1
|
Hilgers RHP, Das KC. Redox Regulation of K + Channel: Role of Thioredoxin. Antioxid Redox Signal 2024. [PMID: 39099341 DOI: 10.1089/ars.2023.0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Significance: Potassium channels regulate the influx and efflux of K+ ions in various cell types that generate and propagate action potential associated with excitation, contraction, and relaxation of various cell types. Although redox active cysteines are critically important for channel activity, the redox regulation of K+ channels by thioredoxin (Trx) has not been systematically reviewed. Recent Advances: Redox regulation of K+ channel is now increasingly recognized as drug targets in the pathological condition of several cardiovascular disease processes. The role of Trx in regulation of these channels and its implication in pathological conditions have not been adequately reviewed. This review specifically focuses on the redox-regulatory role of Trx on K+ channel structure and function in physiological and pathophysiological conditions. Critical Issues: Ion channels, including K+ channel, have been implicated in the functioning of cardiomyocyte excitation-contraction coupling, vascular hyperpolarization, cellular proliferation, and neuronal stimulation in physiological and pathophysiological conditions. Although oxidation-reduction of ion channels is critically important in their function, the role of Trx, redox regulatory protein in regulation of these channels, and its implication in pathological conditions need to be studied to gain further insight into channel function. Future Directions: Future studies need to map all redox regulatory pathways in channel structure and function using novel mouse models and redox proteomic and signal transduction studies, which modulate various currents and altered excitability of relevant cells implicated in a pathological condition. We are yet at infancy of studies related to redox control of various K+ channels and structured and focused studies with novel animal models. Antioxid. Redox Signal. 00, 00-00.
Collapse
Affiliation(s)
- Rob H P Hilgers
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kumuda C Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
2
|
Riddle A, Srivastava T, Wang K, Tellez E, O'Neill H, Gong X, O'Niel A, Bell JA, Raber J, Lattal M, Maylie J, Back SA. Mild neonatal hypoxia disrupts adult hippocampal learning and memory and is associated with CK2-mediated dysregulation of synaptic calcium-activated potassium channel KCNN2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602558. [PMID: 39071376 PMCID: PMC11275740 DOI: 10.1101/2024.07.10.602558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Objective Although nearly half of preterm survivors display persistent neurobehavioral dysfunction including memory impairment without overt gray matter injury, the underlying mechanisms of neuronal or glial dysfunction, and their relationship to commonly observed cerebral white matter injury are unclear. We developed a mouse model to test the hypothesis that mild hypoxia during preterm equivalence is sufficient to persistently disrupt hippocampal neuronal maturation related to adult cellular mechanisms of learning and memory. Methods: Neonatal (P2) mice were exposed to mild hypoxia (8%O 2 ) for 30 min and evaluated for acute injury responses or survived until adulthood for assessment of learning and memory and hippocampal neurodevelopment. Results Neonatal mild hypoxia resulted in clinically relevant oxygen desaturation and tachycardia without bradycardia and was not accompanied by cerebral gray or white matter injury. Neonatal hypoxia exposure was sufficient to cause hippocampal learning and memory deficits and abnormal maturation of CA1 neurons that persisted into adulthood. This was accompanied by reduced hippocampal CA3-CA1 synaptic strength and LTP and reduced synaptic activity of calcium-sensitive SK2 channels, key regulators of spike timing dependent neuroplasticity, including LTP. Structural illumination microscopy revealed reduced synaptic density, but intact SK2 localization at the synapse. Persistent loss of SK2 activity was mediated by altered casein kinase 2 (CK2) signaling. Interpretation Clinically relevant mild hypoxic exposure in the neonatal mouse is sufficient to produce morphometric and functional disturbances in hippocampal neuronal maturation independently of white matter injury. Additionally, we describe a novel persistent mechanism of potassium channel dysregulation after neonatal hypoxia. Collectively our findings suggest an unexplored explanation for the broad spectrum of neurobehavioral, cognitive and learning disabilities that paradoxically persist into adulthood without overt gray matter injury after preterm birth.
Collapse
|
3
|
Abstract
Calcium ions (Ca2+) are the basis of a unique and potent array of cellular responses. Calmodulin (CaM) is a small but vital protein that is able to rapidly transmit information about changes in Ca2+ concentrations to its regulatory targets. CaM plays a critical role in cellular Ca2+ signaling, and interacts with a myriad of target proteins. Ca2+-dependent modulation by CaM is a major component of a diverse array of processes, ranging from gene expression in neurons to the shaping of the cardiac action potential in heart cells. Furthermore, the protein sequence of CaM is highly evolutionarily conserved, and identical CaM proteins are encoded by three independent genes (CALM1-3) in humans. Mutations within any of these three genes may lead to severe cardiac deficits including severe long QT syndrome (LQTS) and/or catecholaminergic polymorphic ventricular tachycardia (CPVT). Research into disease-associated CaM variants has identified several proteins modulated by CaM that are likely to underlie the pathogenesis of these calmodulinopathies, including the cardiac L-type Ca2+ channel (LTCC) CaV1.2, and the sarcoplasmic reticulum Ca2+ release channel, ryanodine receptor 2 (RyR2). Here, we review the research that has been done to identify calmodulinopathic CaM mutations and evaluate the mechanisms underlying their role in disease.
Collapse
Affiliation(s)
- John W. Hussey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Worawan B. Limpitikul
- Department of Medicine, Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ivy E. Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- CONTACT Ivy E. Dick School of Medicine, University of Maryland, Baltimore, MD21210
| |
Collapse
|
4
|
Haji-Ghassemi O, Chen YS, Woll K, Gurrola GB, Valdivia CR, Cai W, Li S, Valdivia HH, Van Petegem F. Cryo-EM analysis of scorpion toxin binding to Ryanodine Receptors reveals subconductance that is abolished by PKA phosphorylation. SCIENCE ADVANCES 2023; 9:eadf4936. [PMID: 37224245 PMCID: PMC10208580 DOI: 10.1126/sciadv.adf4936] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/18/2023] [Indexed: 05/26/2023]
Abstract
Calcins are peptides from scorpion venom with the unique ability to cross cell membranes, gaining access to intracellular targets. Ryanodine Receptors (RyR) are intracellular ion channels that control release of Ca2+ from the endoplasmic and sarcoplasmic reticulum. Calcins target RyRs and induce long-lived subconductance states, whereby single-channel currents are decreased. We used cryo-electron microscopy to reveal the binding and structural effects of imperacalcin, showing that it opens the channel pore and causes large asymmetry throughout the cytosolic assembly of the tetrameric RyR. This also creates multiple extended ion conduction pathways beyond the transmembrane region, resulting in subconductance. Phosphorylation of imperacalcin by protein kinase A prevents its binding to RyR through direct steric hindrance, showing how posttranslational modifications made by the host organism can determine the fate of a natural toxin. The structure provides a direct template for developing calcin analogs that result in full channel block, with potential to treat RyR-related disorders.
Collapse
Affiliation(s)
- Omid Haji-Ghassemi
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Yu Seby Chen
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Kellie Woll
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Georgina B. Gurrola
- Universidad Nacional Autónoma de México, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotechnología, Cuaernavaca, Morelos 62271, Mexico
| | - Carmen R. Valdivia
- Department of Medicine and Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Wenxuan Cai
- Department of Medicine and Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Songhua Li
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hector H. Valdivia
- Department of Medicine and Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
5
|
Nuñez E, Jones F, Muguruza-Montero A, Urrutia J, Aguado A, Malo C, Bernardo-Seisdedos G, Domene C, Millet O, Gamper N, Villarroel A. Redox regulation of K V7 channels through EF3 hand of calmodulin. eLife 2023; 12:e81961. [PMID: 36803414 PMCID: PMC9988260 DOI: 10.7554/elife.81961] [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: 07/18/2022] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
Neuronal KV7 channels, important regulators of cell excitability, are among the most sensitive proteins to reactive oxygen species. The S2S3 linker of the voltage sensor was reported as a site-mediating redox modulation of the channels. Recent structural insights reveal potential interactions between this linker and the Ca2+-binding loop of the third EF-hand of calmodulin (CaM), which embraces an antiparallel fork formed by the C-terminal helices A and B, constituting the calcium responsive domain (CRD). We found that precluding Ca2+ binding to the EF3 hand, but not to EF1, EF2, or EF4 hands, abolishes oxidation-induced enhancement of KV7.4 currents. Monitoring FRET (Fluorescence Resonance Energy Transfer) between helices A and B using purified CRDs tagged with fluorescent proteins, we observed that S2S3 peptides cause a reversal of the signal in the presence of Ca2+ but have no effect in the absence of this cation or if the peptide is oxidized. The capacity of loading EF3 with Ca2+ is essential for this reversal of the FRET signal, whereas the consequences of obliterating Ca2+ binding to EF1, EF2, or EF4 are negligible. Furthermore, we show that EF3 is critical for translating Ca2+ signals to reorient the AB fork. Our data are consistent with the proposal that oxidation of cysteine residues in the S2S3 loop relieves KV7 channels from a constitutive inhibition imposed by interactions between the EF3 hand of CaM which is crucial for this signaling.
Collapse
Affiliation(s)
| | - Frederick Jones
- School of Biomedical Sciences, Faculty of Biological Sciences, University of LeedsLeedsUnited Kingdom
| | | | | | | | | | | | - Carmen Domene
- Department of Chemistry, University of BathBathUnited Kingdom
- Department of Chemistry, University of OxfordOxfordUnited Kingdom
| | - Oscar Millet
- Protein Stability and Inherited Disease Laboratory, CIC bioGUNEDerioSpain
| | - Nikita Gamper
- School of Biomedical Sciences, Faculty of Biological Sciences, University of LeedsLeedsUnited Kingdom
| | | |
Collapse
|
6
|
Halling DB, Philpo AE, Aldrich RW. Calcium dependence of both lobes of calmodulin is involved in binding to a cytoplasmic domain of SK channels. eLife 2022; 11:e81303. [PMID: 36583726 PMCID: PMC9803350 DOI: 10.7554/elife.81303] [Citation(s) in RCA: 2] [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: 06/22/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
KCa2.1-3 Ca2+-activated K+-channels (SK) require calmodulin to gate in response to cellular Ca2+. A model for SK gating proposes that the N-terminal domain (N-lobe) of calmodulin is required for activation, but an immobile C-terminal domain (C-lobe) has constitutive, Ca2+-independent binding. Although structures support a domain-driven hypothesis of SK gate activation by calmodulin, only a partial understanding is possible without measuring both channel activity and protein binding. We measured SK2 (KCa2.2) activity using inside-out patch recordings. Currents from calmodulin-disrupted SK2 channels can be restored with exogenously applied calmodulin. We find that SK2 activity only approaches full activation with full-length calmodulin with both an N- and a C-lobe. We measured calmodulin binding to a C-terminal SK peptide (SKp) using both composition-gradient multi-angle light-scattering and tryptophan emission spectra. Isolated lobes bind to SKp with high affinity, but isolated lobes do not rescue SK2 activity. Consistent with earlier models, N-lobe binding to SKp is stronger in Ca2+, and C-lobe-binding affinity is strong independent of Ca2+. However, a native tryptophan in SKp is sensitive to Ca2+ binding to both the N- and C-lobes of calmodulin at Ca2+ concentrations that activate SK2, demonstrating that the C-lobe interaction with SKp changes with Ca2+. Our peptide-binding data and electrophysiology show that SK gating models need deeper scrutiny. We suggest that the Ca2+-dependent associations of both lobes of calmodulin to SKp are crucial events during gating. Additional investigations are necessary to complete a mechanistic gating model consistent with binding, physiology, and structure.
Collapse
Affiliation(s)
- David B Halling
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| | - Ashley E Philpo
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| | - Richard W Aldrich
- Department of Neuroscience, The University of Texas at AustinAustinUnited States
| |
Collapse
|
7
|
Tiffner A, Hopl V, Derler I. CRAC and SK Channels: Their Molecular Mechanisms Associated with Cancer Cell Development. Cancers (Basel) 2022; 15:101. [PMID: 36612099 PMCID: PMC9817886 DOI: 10.3390/cancers15010101] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer represents a major health burden worldwide. Several molecular targets have been discovered alongside treatments with positive clinical outcomes. However, the reoccurrence of cancer due to therapy resistance remains the primary cause of mortality. Endeavors in pinpointing new markers as molecular targets in cancer therapy are highly desired. The significance of the co-regulation of Ca2+-permeating and Ca2+-regulated ion channels in cancer cell development, proliferation, and migration make them promising molecular targets in cancer therapy. In particular, the co-regulation of the Orai1 and SK3 channels has been well-studied in breast and colon cancer cells, where it finally leads to an invasion-metastasis cascade. Nevertheless, many questions remain unanswered, such as which key molecular components determine and regulate their interplay. To provide a solid foundation for a better understanding of this ion channel co-regulation in cancer, we first shed light on the physiological role of Ca2+ and how this ion is linked to carcinogenesis. Then, we highlight the structure/function relationship of Orai1 and SK3, both individually and in concert, their role in the development of different types of cancer, and aspects that are not yet known in this context.
Collapse
Affiliation(s)
- Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| |
Collapse
|
8
|
Yang X, Chen S, Zhang S, Shi S, Zong R, Gao Y, Guan B, Gamper N, Gao H. Intracellular zinc protects Kv7 K + channels from Ca 2+/calmodulin-mediated inhibition. J Biol Chem 2022; 299:102819. [PMID: 36549648 PMCID: PMC9852549 DOI: 10.1016/j.jbc.2022.102819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Zinc (Zn) is an essential trace element; it serves as a cofactor for a great number of enzymes, transcription factors, receptors, and other proteins. Zinc is also an important signaling molecule, which can be released from intracellular stores into the cytosol or extracellular space, for example, during synaptic transmission. Amongst cellular effects of zinc is activation of Kv7 (KCNQ, M-type) voltage-gated potassium channels. Here, we investigated relationships between Kv7 channel inhibition by Ca2+/calmodulin (CaM) and zinc-mediated potentiation. We show that Zn2+ ionophore, zinc pyrithione (ZnPy), can prevent or reverse Ca2+/CaM-mediated inhibition of Kv7.2. In the presence of both Ca2+ and Zn2+, the Kv7.2 channels lose most of their voltage dependence and lock in an open state. In addition, we demonstrate that mutations that interfere with CaM binding to Kv7.2 and Kv7.3 reduced channel membrane abundance and activity, but these mutants retained zinc sensitivity. Moreover, the relative efficacy of ZnPy to activate these mutants was generally greater, compared with the WT channels. Finally, we show that zinc sensitivity was retained in Kv7.2 channels assembled with mutant CaM with all four EF hands disabled, suggesting that it is unlikely to be mediated by CaM. Taken together, our findings indicate that zinc is a potent Kv7 stabilizer, which may protect these channels from physiological inhibitory effects of neurotransmitters and neuromodulators, protecting neurons from overactivity.
Collapse
Affiliation(s)
- Xinhe Yang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China,CSPC ZhongQi Pharmaceutical Technology (Shijiazhuang) Co, Ltd, Shijiazhuang, Hebei, China
| | - Shuai Chen
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuo Zhang
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sai Shi
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, China
| | - Rui Zong
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiting Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bingcai Guan
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China; Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK.
| | - Haixia Gao
- Department of Pharmacology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
9
|
Feliziani C, Fernandez M, Quasollo G, Holstein D, Bairo SM, Paton JC, Paton AW, de Batista J, Lechleiter JD, Bollo M. Ca 2+ signalling system initiated by endoplasmic reticulum stress stimulates PERK activation. Cell Calcium 2022; 106:102622. [PMID: 35908318 PMCID: PMC9982837 DOI: 10.1016/j.ceca.2022.102622] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/11/2022] [Accepted: 07/05/2022] [Indexed: 01/25/2023]
Abstract
The accumulation of unfolded proteins within the Endoplasmic Reticulum (ER) activates a signal transduction pathway termed the unfolded protein response (UPR), which attempts to restore ER homoeostasis. If this cannot be done, UPR signalling ultimately induces apoptosis. Ca2+ depletion in the ER is a potent inducer of ER stress. Despite the ubiquity of Ca2+ as an intracellular messenger, the precise mechanism(s) by which Ca2+ release affects the UPR remains unknown. Tethering a genetically encoded Ca2+ indicator (GCamP6) to the ER membrane revealed novel Ca2+ signalling events initiated by Ca2+ microdomains in human astrocytes under ER stress, induced by tunicamycin (Tm), an N-glycosylation inhibitor, as well as in a cell model deficient in all three inositol triphosphate receptor isoforms. Pharmacological and molecular studies indicate that these local events are mediated by translocons and that the Ca2+ microdomains impact (PKR)-like-ER kinase (PERK), an UPR sensor, activation. These findings reveal the existence of a Ca2+ signal mechanism by which stressor-mediated Ca2+ release regulates ER stress.
Collapse
Affiliation(s)
- Constanza Feliziani
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - Macarena Fernandez
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - Gonzalo Quasollo
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - Deborah Holstein
- Department of Cell Systems and Anatomy, UT Health San
Antonio, 8403 Floyd Curl Dr., San Antonio, TX 78229-3904, USA
| | - Sebastián M Bairo
- Instituto de Investigación Médica M y M
Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli,
Córdoba 5016, Argentina
| | - James C Paton
- Research Centre for Infectious Diseases, School of
Molecular and Biomedical Science, University of Adelaide, South Australia 5005,
Australia
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, School of
Molecular and Biomedical Science, University of Adelaide, South Australia 5005,
Australia
| | - Juan de Batista
- Instituto Universitario de Ciencias Biomédicas de
Córdoba (IUCBC), Hospital Privado Universitario de Córdoba, 420
Naciones Unidas, Córdoba 5016, Argentina
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, UT Health San
Antonio, 8403 Floyd Curl Dr., San Antonio, TX 78229-3904, USA
| | - Mariana Bollo
- Instituto de Investigación Médica M y M Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, 2434 Friuli, Córdoba 5016, Argentina.
| |
Collapse
|
10
|
Altinoz MA, Guloksuz S, Ozpinar A. Immunomodifying and neuroprotective effects of noscapine: Implications for multiple sclerosis, neurodegenerative, and psychiatric disorders. Chem Biol Interact 2021; 352:109794. [PMID: 34963564 DOI: 10.1016/j.cbi.2021.109794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/20/2021] [Indexed: 12/20/2022]
Abstract
Noscapine is a phthalide isoquinoline alkaloid with antitussive activity. Noscapine protects oligodendroglia from ischemic and chemical injury, binds to bitter taste receptors, antagonizes the bradykinin and histaminergic systems, which may be of benefit in treatment of multiple sclerosis. Noscapine normalizes axonal transport and exerts significant therapeutic efficacy in animal models of Parkinson's Disease and Amyotrophic Lateral Sclerosis. Noscapine exerts neuroprotective effects on oxygen- and glucose-deprived fetal cortical neuronal cells and reduces ischemic brain damage in neonatal rat pups. Pilot clinical studies indicated some beneficial effects of noscapine in stroke. Noscapine harbours anxiolytic activity and methyl-noscapine blocks small conductance SK channels, which is beneficial in alleviating anxiety and depression. Noscapine exerts anticholinesterase activity and acts inhibitory on the inflammatory transcription factor NF-κB, which may be harnessed in treatment of Alzheimer's Disease. With its blood-brain barrier traversing features and versatile actions, noscapine may be a promising agent in the armamentarium against neurodegenerative and psychiatric diseases.
Collapse
Affiliation(s)
- Meric A Altinoz
- Department of Medical Biochemistry, Acibadem University, Istanbul, Turkey; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, the Netherlands.
| | - Sinan Guloksuz
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, the Netherlands; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Aysel Ozpinar
- Department of Medical Biochemistry, Acibadem University, Istanbul, Turkey
| |
Collapse
|
11
|
Mahling R, Hovey L, Isbell HM, Marx DC, Miller MS, Kilpatrick AM, Weaver LD, Yoder JB, Kim EH, Andresen CNJ, Li S, Shea MA. Na V1.2 EFL domain allosterically enhances Ca 2+ binding to sites I and II of WT and pathogenic calmodulin mutants bound to the channel CTD. Structure 2021; 29:1339-1356.e7. [PMID: 33770503 PMCID: PMC8458505 DOI: 10.1016/j.str.2021.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 12/23/2020] [Accepted: 03/03/2021] [Indexed: 11/23/2022]
Abstract
Neuronal voltage-gated sodium channel NaV1.2 C-terminal domain (CTD) binds calmodulin (CaM) constitutively at its IQ motif. A solution structure (6BUT) and other NMR evidence showed that the CaM N domain (CaMN) is structurally independent of the C-domain (CaMC) whether CaM is bound to the NaV1.2IQp (1,901-1,927) or NaV1.2CTD (1,777-1,937) with or without calcium. However, in the CaM + NaV1.2CTD complex, the Ca2+ affinity of CaMN was more favorable than in free CaM, while Ca2+ affinity for CaMC was weaker than in the CaM + NaV1.2IQp complex. The CTD EF-like (EFL) domain allosterically widened the energetic gap between CaM domains. Cardiomyopathy-associated CaM mutants (N53I(N54I), D95V(D96V), A102V(A103V), E104A(E105A), D129G(D130G), and F141L(F142L)) all bound the NaV1.2 IQ motif favorably under resting (apo) conditions and bound calcium normally at CaMN sites. However, only N53I and A102V bound calcium at CaMC sites at [Ca2+] < 100 μM. Thus, they are expected to respond like wild-type CaM to Ca2+ spikes in excitable cells.
Collapse
Affiliation(s)
- Ryan Mahling
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Liam Hovey
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Holly M Isbell
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Dagan C Marx
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Mark S Miller
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Adina M Kilpatrick
- Department of Physics and Astronomy, Drake University, Des Moines, IA 50311-4516, USA
| | - Lisa D Weaver
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Jesse B Yoder
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Elaine H Kim
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Corinne N J Andresen
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Shuxiang Li
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Madeline A Shea
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA.
| |
Collapse
|
12
|
Daniel NH, Aravind A, Thakur P. Are ion channels potential therapeutic targets for Parkinson's disease? Neurotoxicology 2021; 87:243-257. [PMID: 34699791 DOI: 10.1016/j.neuro.2021.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is primarily associated with the progressive neurodegeneration of the dopaminergic neurons in the substantia nigra region of the brain. The resulting motor symptoms are managed with the help of dopamine replacement therapies. However, these therapeutics do not prevent the neurodegeneration underlying the disease and therefore lose their effectiveness in managing disease symptoms over time. Thus, there is an urgent need to develop newer therapeutics for the benefit of patients. The release of dopamine and the firing activity of substantia nigra neurons is regulated by several ion channels that act in concert. Dysregulations of these channels cause the aberrant movement of various ions in the intracellular milieu. This eventually leads to disruption of intracellular signalling cascades, alterations in cellular homeostasis, and bioenergetic deficits. Therefore, ion channels play a central role in driving the high vulnerability of dopaminergic neurons to degenerate during PD. Targeting ion channels offers an attractive mechanistic strategy to combat the process of neurodegeneration. In this review, we highlight the evidence pointing to the role of various ion channels in driving the PD processes. In addition, we also discuss the various drugs or compounds that target the ion channels and have shown neuroprotective potential in the in-vitro and in-vivo models of PD. We also discuss the current clinical status of various drugs targeting the ion channels in the context of PD.
Collapse
Affiliation(s)
- Neha Hanna Daniel
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Ananya Aravind
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Poonam Thakur
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India.
| |
Collapse
|
13
|
Ledford HA, Park S, Muir D, Woltz RL, Ren L, Nguyen PT, Sirish P, Wang W, Sihn CR, George AL, Knollmann BC, Yamoah EN, Yarov-Yarovoy V, Zhang XD, Chiamvimonvat N. Different arrhythmia-associated calmodulin mutations have distinct effects on cardiac SK channel regulation. J Gen Physiol 2021; 152:211546. [PMID: 33211795 PMCID: PMC7681919 DOI: 10.1085/jgp.202012667] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/25/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Calmodulin (CaM) plays a critical role in intracellular signaling and regulation of Ca2+-dependent proteins and ion channels. Mutations in CaM cause life-threatening cardiac arrhythmias. Among the known CaM targets, small-conductance Ca2+-activated K+ (SK) channels are unique, since they are gated solely by beat-to-beat changes in intracellular Ca2+. However, the molecular mechanisms of how CaM mutations may affect the function of SK channels remain incompletely understood. To address the structural and functional effects of these mutations, we introduced prototypical human CaM mutations in human induced pluripotent stem cell–derived cardiomyocyte-like cells (hiPSC-CMs). Using structural modeling and molecular dynamics simulation, we demonstrate that human calmodulinopathy-associated CaM mutations disrupt cardiac SK channel function via distinct mechanisms. CaMD96V and CaMD130G mutants reduce SK currents through a dominant-negative fashion. By contrast, specific mutations replacing phenylalanine with leucine result in conformational changes that affect helix packing in the C-lobe, which disengage the interactions between apo-CaM and the CaM-binding domain of SK channels. Distinct mutant CaMs may result in a significant reduction in the activation of the SK channels, leading to a decrease in the key Ca2+-dependent repolarization currents these channels mediate. The findings in this study may be generalizable to other interactions of mutant CaMs with Ca2+-dependent proteins within cardiac myocytes.
Collapse
Affiliation(s)
- Hannah A Ledford
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Seojin Park
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Duncan Muir
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Ryan L Woltz
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Lu Ren
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Phuong T Nguyen
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA
| | - Wenying Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Choong-Ryoul Sihn
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Björn C Knollmann
- Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, School of Medicine, Vanderbilt University, Nashville, TN
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, Reno, NV
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA.,Department of Veterans Affairs, Northern California Health Care System, Mather, CA
| |
Collapse
|
14
|
Sun J, Liu Y, Baudry M, Bi X. SK2 channel regulation of neuronal excitability, synaptic transmission, and brain rhythmic activity in health and diseases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118834. [PMID: 32860835 PMCID: PMC7541745 DOI: 10.1016/j.bbamcr.2020.118834] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 11/20/2022]
Abstract
Small conductance calcium-activated potassium channels (SKs) are solely activated by intracellular Ca2+ and their activation leads to potassium efflux, thereby repolarizing/hyperpolarizing membrane potential. Thus, these channels play a critical role in synaptic transmission, and consequently in information transmission along the neuronal circuits expressing them. SKs are widely but not homogeneously distributed in the central nervous system (CNS). Activation of SKs requires submicromolar cytoplasmic Ca2+ concentrations, which are reached following either Ca2+ release from intracellular Ca2+ stores or influx through Ca2+ permeable membrane channels. Both Ca2+ sensitivity and synaptic levels of SKs are regulated by protein kinases and phosphatases, and degradation pathways. SKs in turn control the activity of multiple Ca2+ channels. They are therefore critically involved in coordinating diverse Ca2+ signaling pathways and controlling Ca2+ signal amplitude and duration. This review highlights recent advances in our understanding of the regulation of SK2 channels and of their roles in normal brain functions, including synaptic plasticity, learning and memory, and rhythmic activities. It will also discuss how alterations in their expression and regulation might contribute to various brain disorders such as Angelman Syndrome, Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Yan Liu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Michel Baudry
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America.
| |
Collapse
|
15
|
Weisbrod D. Small and Intermediate Calcium Activated Potassium Channels in the Heart: Role and Strategies in the Treatment of Cardiovascular Diseases. Front Physiol 2020; 11:590534. [PMID: 33329039 PMCID: PMC7719780 DOI: 10.3389/fphys.2020.590534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium-activated potassium channels are a heterogeneous family of channels that, despite their different biophysical characteristics, structures, and pharmacological signatures, play a role of transducer between the ubiquitous intracellular calcium signaling and the electric variations of the membrane. Although this family of channels was extensively described in various excitable and non-excitable tissues, an increasing amount of evidences shows their functional role in the heart. This review aims to focus on the physiological role and the contribution of the small and intermediate calcium-activated potassium channels in cardiac pathologies.
Collapse
|
16
|
Diniz AFA, Ferreira RC, de Souza ILL, da Silva BA. Ionic Channels as Potential Therapeutic Targets for Erectile Dysfunction: A Review. Front Pharmacol 2020; 11:1120. [PMID: 32848741 PMCID: PMC7396897 DOI: 10.3389/fphar.2020.01120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
Erectile dysfunction (ED) is a prevalent condition, especially in men over 40 years old, characterized by the inability to obtain and/or maintain penile erection sufficient for satisfactory sexual intercourse. Several psychological and/or organic factors are involved in the etiopathogenesis of ED. In this context, we gathered evidence of the involvement of Large-conductance, Ca2+-activated K+ channels (BKCa), Small-conductance, Ca2+-activated K+ channels (SKCa), KCNQ-encoded voltage-dependent K+ channels (KV7), Transient Receptor Potential channels (TRP), and Calcium-activated Chloride channels (CaCC) dysfunctions on ED. In addition, the use of modulating agents of these channels are involved in relaxation of the cavernous smooth muscle cell and, consequent penile erection, suggesting that these channels are promising therapeutic targets for the treatment of erectile dysfunction.
Collapse
Affiliation(s)
- Anderson Fellyp Avelino Diniz
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Rafael Carlos Ferreira
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Iara Leão Luna de Souza
- Departamento de Ciências Biológicas e da Saúde, Universidade Estadual de Roraima, Boa Vista, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| |
Collapse
|
17
|
SAC3B is a target of CML19, the centrin 2 of Arabidopsis thaliana. Biochem J 2020; 477:173-189. [PMID: 31860002 DOI: 10.1042/bcj20190674] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 01/15/2023]
Abstract
Arabidopsis centrin 2, also known as calmodulin-like protein 19 (CML19), is a member of the EF-hand superfamily of calcium (Ca2+)-binding proteins. In addition to the notion that CML19 interacts with the nucleotide excision repair protein RAD4, CML19 was suggested to be a component of the transcription export complex 2 (TREX-2) by interacting with SAC3B. However, the molecular determinants of this interaction have remained largely unknown. Herein, we identified a CML19-binding site within the C-terminus of SAC3B and characterized the binding properties of the corresponding 26-residue peptide (SAC3Bp), which exhibits the hydrophobic triad centrin-binding motif in a reversed orientation (I8W4W1). Using a combination of spectroscopic and calorimetric experiments, we shed light on the SAC3Bp-CML19 complex structure in solution. We demonstrated that the peptide interacts not only with Ca2+-saturated CML19, but also with apo-CML19 to form a protein-peptide complex with a 1 : 1 stoichiometry. Both interactions involve hydrophobic and electrostatic contributions and include the burial of Trp residues of SAC3Bp. However, the peptide likely assumes different conformations upon binding to apo-CML19 or Ca2+-CML19. Importantly, the peptide dramatically increases the affinity for Ca2+ of CML19, especially of the C-lobe, suggesting that in vivo the protein would be Ca2+-saturated and bound to SAC3B even at resting Ca2+-levels. Our results, providing direct evidence that Arabidopsis SAC3B is a CML19 target and proposing that CML19 can bind to SAC3B through its C-lobe independent of a Ca2+ stimulus, support a functional role for these proteins in TREX-2 complex and mRNA export.
Collapse
|
18
|
Park EYJ, Baik JY, Kwak M, So I. The role of calmodulin in regulating calcium-permeable PKD2L1 channel activity. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:219-227. [PMID: 31080352 PMCID: PMC6488707 DOI: 10.4196/kjpp.2019.23.3.219] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 11/15/2022]
Abstract
Polycystic kidney disease 2-like-1 (PKD2L1), polycystin-L or transient receptor potential polycystin 3 (TRPP3) is a TRP superfamily member. It is a calcium-permeable non-selective cation channel that regulates intracellular calcium concentration and thereby calcium signaling. Although the calmodulin (CaM) inhibitor, calmidazolium, is an activator of the PKD2L1 channel, the activating mechanism remains unclear. The purpose of this study is to clarify whether CaM takes part in the regulation of the PKD2L1 channel, and if so, how. With patch clamp techniques, we observed the current amplitudes of PKD2L1 significantly reduced when coexpressed with CaM and CaMΔN. This result suggests that the N-lobe of CaM carries a more crucial role in regulating PKD2L1 and guides us into our next question on the different functions of two lobes of CaM. We also identified the predicted CaM binding site, and generated deletion and truncation mutants. The mutants showed significant reduction in currents losing PKD2L1 current-voltage curve, suggesting that the C-terminal region from 590 to 600 is crucial for maintaining the functionality of the PKD2L1 channel. With PKD2L1608Stop mutant showing increased current amplitudes, we further examined the functional importance of EF-hand domain. Along with co-expression of CaM, ΔEF-hand mutant also showed significant changes in current amplitudes and potentiation time. Our findings suggest that there is a constitutive inhibition of EF-hand and binding of CaM C-lobe on the channel in low calcium concentration. At higher calcium concentration, calcium ions occupy the N-lobe as well as the EF-hand domain, allowing the two to compete to bind to the channel.
Collapse
Affiliation(s)
- Eunice Yon June Park
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Julia Young Baik
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Misun Kwak
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
19
|
Archer CR, Enslow BT, Taylor AB, De la Rosa V, Bhattacharya A, Shapiro MS. A mutually induced conformational fit underlies Ca 2+-directed interactions between calmodulin and the proximal C terminus of KCNQ4 K + channels. J Biol Chem 2019; 294:6094-6112. [PMID: 30808708 PMCID: PMC6463706 DOI: 10.1074/jbc.ra118.006857] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/24/2019] [Indexed: 11/06/2022] Open
Abstract
Calmodulin (CaM) conveys intracellular Ca2+ signals to KCNQ (Kv7, "M-type") K+ channels and many other ion channels. Whether this "calmodulation" involves a dramatic structural rearrangement or only slight perturbations of the CaM/KCNQ complex is as yet unclear. A consensus structural model of conformational shifts occurring between low nanomolar and physiologically high intracellular [Ca2+] is still under debate. Here, we used various techniques of biophysical chemical analyses to investigate the interactions between CaM and synthetic peptides corresponding to the A and B domains of the KCNQ4 subtype. We found that in the absence of CaM, the peptides are disordered, whereas Ca2+/CaM imposed helical structure on both KCNQ A and B domains. Isothermal titration calorimetry revealed that Ca2+/CaM has higher affinity for the B domain than for the A domain of KCNQ2-4 and much higher affinity for the B domain when prebound with the A domain. X-ray crystallography confirmed that these discrete peptides spontaneously form a complex with Ca2+/CaM, similar to previous reports of CaM binding KCNQ-AB domains that are linked together. Microscale thermophoresis and heteronuclear single-quantum coherence NMR spectroscopy indicated the C-lobe of Ca2+-free CaM to interact with the KCNQ4 B domain (Kd ∼10-20 μm), with increasing Ca2+ molar ratios shifting the CaM-B domain interactions via only the CaM C-lobe to also include the N-lobe. Our findings suggest that in response to increased Ca2+, CaM undergoes lobe switching that imposes a dramatic mutually induced conformational fit to both the proximal C terminus of KCNQ4 channels and CaM, likely underlying Ca2+-dependent regulation of KCNQ gating.
Collapse
Affiliation(s)
- Crystal R Archer
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229; Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Benjamin T Enslow
- the Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Alexander B Taylor
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Victor De la Rosa
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Akash Bhattacharya
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Mark S Shapiro
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229.
| |
Collapse
|
20
|
Limpitikul WB, Greenstein JL, Yue DT, Dick IE, Winslow RL. A bilobal model of Ca 2+-dependent inactivation to probe the physiology of L-type Ca 2+ channels. J Gen Physiol 2018; 150:1688-1701. [PMID: 30470716 PMCID: PMC6279366 DOI: 10.1085/jgp.201812115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/01/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
L-type calcium channels undergo Ca2+-dependent inactivation (CDI) in order to precisely control the entry of Ca2+ into cells such as cardiomyocytes. Limpitikul et al. develop a bilobal model of CDI and use it to understand the pathogenesis of arrhythmias associated with mutations in CaM. L-type calcium channels (LTCCs) are critical elements of normal cardiac function, playing a major role in orchestrating cardiac electrical activity and initiating downstream signaling processes. LTCCs thus use feedback mechanisms to precisely control calcium (Ca2+) entry into cells. Of these, Ca2+-dependent inactivation (CDI) is significant because it shapes cardiac action potential duration and is essential for normal cardiac rhythm. This important form of regulation is mediated by a resident Ca2+ sensor, calmodulin (CaM), which is comprised of two lobes that are each capable of responding to spatially distinct Ca2+ sources. Disruption of CaM-mediated CDI leads to severe forms of long-QT syndrome (LQTS) and life-threatening arrhythmias. Thus, a model capable of capturing the nuances of CaM-mediated CDI would facilitate increased understanding of cardiac (patho)physiology. However, one critical barrier to achieving a detailed kinetic model of CDI has been the lack of quantitative data characterizing CDI as a function of Ca2+. This data deficit stems from the experimental challenge of uncoupling the effect of channel gating on Ca2+ entry. To overcome this obstacle, we use photo-uncaging of Ca2+ to deliver a measurable Ca2+ input to CaM/LTCCs, while simultaneously recording CDI. Moreover, we use engineered CaMs with Ca2+ binding restricted to a single lobe, to isolate the kinetic response of each lobe. These high-resolution measurements enable us to build mathematical models for each lobe of CaM, which we use as building blocks for a full-scale bilobal model of CDI. Finally, we use this model to probe the pathogenesis of LQTS associated with mutations in CaM (calmodulinopathies). Each of these models accurately recapitulates the kinetics and steady-state properties of CDI in both physiological and pathological states, thus offering powerful new insights into the mechanistic alterations underlying cardiac arrhythmias.
Collapse
Affiliation(s)
- Worawan B Limpitikul
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph L Greenstein
- Institute for Computational Medicine, Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - David T Yue
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ivy E Dick
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD .,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Raimond L Winslow
- Institute for Computational Medicine, Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| |
Collapse
|
21
|
Bajorat R, Porath K, Kuhn J, Goßla E, Goerss D, Sellmann T, Köhling R, Kirschstein T. Oral administration of the casein kinase 2 inhibitor TBB leads to persistent K Ca2.2 channel up-regulation in the epileptic CA1 area and cortex, but lacks anti-seizure efficacy in the pilocarpine epilepsy model. Epilepsy Res 2018; 147:42-50. [PMID: 30219695 DOI: 10.1016/j.eplepsyres.2018.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/31/2018] [Accepted: 08/29/2018] [Indexed: 02/04/2023]
Abstract
Temporal lobe epilepsy (TLE) is the most common epileptic syndrome in adults and often presents with seizures that prove intractable with currently available anticonvulsants. Thus, there is still a need for new anti-seizure drugs in this condition. Recently, we found that the casein kinase 2 inhibitor 4,5,6,7-tetrabromotriazole (TBB) prevented the emergence of spontaneous epileptic discharges in an acute in vitro epilepsy model. This prompted us to study the anti-seizure effects of TBB in the pilocarpine model of chronic epilepsy in vivo. To this end, we performed long-term video-EEG monitoring lasting 78-167 days of nine chronically epileptic rats and obtained a baseline seizure rate of 3.3 ± 1.3 per day (baseline of 27-80 days). We found a significant age effect with more pronounced seizure rates in older animals as compared to younger ones. However, the seizure rate increased to 6.3 ± 2.2 per day during the oral TBB administration (treatment period of 21-50 days), and following discontinuation of TBB, this rate remained stable with 5.2 ± 1.4 seizures per day (follow-up of 30-55 days). After completing the video-EEG during the follow-up the hippocampal tissue was prepared and studied for the expression of the Ca2+-activated K+ channel KCa2.2. We found a significant up-regulation of KCa2.2 in the epileptic CA1 region and in the neocortex, but in no other hippocampal subfield. Hence, our findings indicate that oral administration of TBB leads to persistent up-regulation of KCa2.2 in the epileptic CA1 subfield and in the neocortex, but lacks anti-seizure efficacy in the pilocarpine epilepsy model.
Collapse
Affiliation(s)
- Rika Bajorat
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany; Department of Anesthesiology and Intensive Care Medicine, Rostock University Medical Centre, Rostock, Germany.
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Johannes Kuhn
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Elke Goßla
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Doreen Goerss
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany; Department of Psychosomatic and Psychotherapeutic Medicine, Rostock University Medical Centre, Rostock, Germany.
| | - Tina Sellmann
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| |
Collapse
|
22
|
Lee CH, MacKinnon R. Activation mechanism of a human SK-calmodulin channel complex elucidated by cryo-EM structures. Science 2018; 360:508-513. [PMID: 29724949 DOI: 10.1126/science.aas9466] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/16/2018] [Indexed: 12/17/2022]
Abstract
Small-conductance Ca2+-activated K+ (SK) channels mediate neuron excitability and are associated with synaptic transmission and plasticity. They also regulate immune responses and the size of blood cells. Activation of SK channels requires calmodulin (CaM), but how CaM binds and opens SK channels has been unclear. Here we report cryo-electron microscopy (cryo-EM) structures of a human SK4-CaM channel complex in closed and activated states at 3.4- and 3.5-angstrom resolution, respectively. Four CaM molecules bind to one channel tetramer. Each lobe of CaM serves a distinct function: The C-lobe binds to the channel constitutively, whereas the N-lobe interacts with the S4-S5 linker in a Ca2+-dependent manner. The S4-S5 linker, which contains two distinct helices, undergoes conformational changes upon CaM binding to open the channel pore. These structures reveal the gating mechanism of SK channels and provide a basis for understanding SK channel pharmacology.
Collapse
Affiliation(s)
- Chia-Hsueh Lee
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, 1230 York Avenue, New York, NY 10065, USA
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
23
|
Chang A, Abderemane-Ali F, Hura GL, Rossen ND, Gate RE, Minor DL. A Calmodulin C-Lobe Ca 2+-Dependent Switch Governs Kv7 Channel Function. Neuron 2018; 97:836-852.e6. [PMID: 29429937 DOI: 10.1016/j.neuron.2018.01.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/07/2017] [Accepted: 01/12/2018] [Indexed: 12/22/2022]
Abstract
Kv7 (KCNQ) voltage-gated potassium channels control excitability in the brain, heart, and ear. Calmodulin (CaM) is crucial for Kv7 function, but how this calcium sensor affects activity has remained unclear. Here, we present X-ray crystallographic analysis of CaM:Kv7.4 and CaM:Kv7.5 AB domain complexes that reveal an Apo/CaM clamp conformation and calcium binding preferences. These structures, combined with small-angle X-ray scattering, biochemical, and functional studies, establish a regulatory mechanism for Kv7 CaM modulation based on a common architecture in which a CaM C-lobe calcium-dependent switch releases a shared Apo/CaM clamp conformation. This C-lobe switch inhibits voltage-dependent activation of Kv7.4 and Kv7.5 but facilitates Kv7.1, demonstrating that mechanism is shared by Kv7 isoforms despite the different directions of CaM modulation. Our findings provide a unified framework for understanding how CaM controls different Kv7 isoforms and highlight the role of membrane proximal domains for controlling voltage-gated channel function. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Aram Chang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Greg L Hura
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Nathan D Rossen
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Rachel E Gate
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA; California Institute for Quantitative Biomedical Research, University of California San Francisco, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
24
|
Sultana R, Ghandi T, M. Davila A, Lee CC, Ogundele OM. Upregulated SK2 Expression and Impaired CaMKII Phosphorylation Are Shared Synaptic Defects Between 16p11.2del and 129S: Δdisc1 Mutant Mice. ASN Neuro 2018; 10:1759091418817641. [PMID: 33592687 PMCID: PMC6295693 DOI: 10.1177/1759091418817641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/29/2022] Open
Abstract
Ion channel gating and kinase regulation of N-methyl-D-aspartate receptor 1 activity are fundamental mechanisms that govern synaptic plasticity. In this study, we showed that two mutant models (16p11.2del and Δdisc1 ) that recapitulate aspects of human cognitive disorders shared a similar defect in N-methyl-D-aspartate receptor 1-dependent synaptic function. Our results demonstrate that the expression of small-conductance potassium channels (SK2 or KCa2.2) was significantly upregulated in the hippocampus and prefrontal cortex of 16p11.2del and 129S:Δdisc1 mutant mice. Likewise, both mutant strains exhibited an impairment of T286 phosphorylation of calcium-calmodulin-dependent kinase II (CaMKII) in the hippocampus and prefrontal cortex. In vivo neural recordings revealed that increased SK2 expression and impaired T286 phosphorylation of CaMKII coincide with a prolonged interspike interval in the hippocampal cornu ammonis-1 (CA1) field for both 16p11.2del and 129S:Δdisc1 mutant mice. These findings suggest that alteration of small conductance channels and T286 phosphorylation of CaMKII are likely shared factors underlying behavioral changes in these two genetic mouse models.
Collapse
Affiliation(s)
- Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Tanya Ghandi
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Alexandra M. Davila
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| |
Collapse
|
25
|
Tobelaim WS, Dvir M, Lebel G, Cui M, Buki T, Peretz A, Marom M, Haitin Y, Logothetis DE, Hirsch JA, Attali B. Ca 2+-Calmodulin and PIP2 interactions at the proximal C-terminus of Kv7 channels. Channels (Austin) 2017; 11:686-695. [PMID: 28976808 PMCID: PMC5786183 DOI: 10.1080/19336950.2017.1388478] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/18/2017] [Accepted: 09/29/2017] [Indexed: 10/18/2022] Open
Abstract
In the heart, co-assembly of Kv7.1 with KCNE1 produces the slow IKS potassium current, which repolarizes the cardiac action potential and mutations in human Kv7.1 and KCNE1 genes cause cardiac arrhythmias. The proximal Kv7.1 C-terminus binds calmodulin (CaM) and phosphatidylinositol-4,5-bisphosphate (PIP2) and recently we revealed the competition of PIP2 with the calcified CaM N-lobe to a previously unidentified site in Kv7.1 helix B, also known to harbor a LQT mutation. Data indicated that PIP2 and Ca2+-CaM perform the same function on IKS channel gating to stabilize the channel open state. Here we show that similar features were observed for Kv7.1 currents expressed alone. We also find that conservation of homologous residues in helix B of other Kv7 subtypes confer similar competition of Ca2+-CaM with PIP2 binding to their proximal C-termini and suggest that PIP2-CaM interactions converge to Kv7 helix B to modulates channel activity in a Kv7 subtype-dependent manner.
Collapse
Affiliation(s)
- William S. Tobelaim
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Meidan Dvir
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Guy Lebel
- Department of Biochemistry & Molecular Biology, George S. Wise Faculty of Life Sciences, Institute of Structural Biology, Tel Aviv University, Tel Aviv, Israel
| | - Meng Cui
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Tal Buki
- Laboratory of Structural Physiology, Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Asher Peretz
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Milit Marom
- Laboratory of Structural Physiology, Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yoni Haitin
- Laboratory of Structural Physiology, Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Joel A. Hirsch
- Department of Biochemistry & Molecular Biology, George S. Wise Faculty of Life Sciences, Institute of Structural Biology, Tel Aviv University, Tel Aviv, Israel
| | - Bernard Attali
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
Guarina L, Vandael DHF, Carabelli V, Carbone E. Low pH o boosts burst firing and catecholamine release by blocking TASK-1 and BK channels while preserving Cav1 channels in mouse chromaffin cells. J Physiol 2017; 595:2587-2609. [PMID: 28026020 DOI: 10.1113/jp273735] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 12/07/2016] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Mouse chromaffin cells (MCCs) generate spontaneous burst-firing that causes large increases of Ca2+ -dependent catecholamine release, and is thus a key mechanism for regulating the functions of MCCs. With the aim to uncover a physiological role for burst-firing we investigated the effects of acidosis on MCC activity. Lowering the extracellular pH (pHo ) from 7.4 to 6.6 induces cell depolarizations of 10-15 mV that generate bursts of ∼330 ms at 1-2 Hz and a 7.4-fold increase of cumulative catecholamine-release. Burst-firing originates from the inhibition of the pH-sensitive TASK-1-channels and a 60% reduction of BK-channel conductance at pHo 6.6. Blockers of the two channels (A1899 and paxilline) mimic the effects of pHo 6.6, and this is reverted by the Cav1 channel blocker nifedipine. MCCs act as pH-sensors. At low pHo , they depolarize, undergo burst-firing and increase catecholamine-secretion, generating an effective physiological response that may compensate for the acute acidosis and hyperkalaemia generated during heavy exercise and muscle fatigue. ABSTRACT Mouse chromaffin cells (MCCs) generate action potential (AP) firing that regulates the Ca2+ -dependent release of catecholamines (CAs). Recent findings indicate that MCCs possess a variety of spontaneous firing modes that span from the common 'tonic-irregular' to the less frequent 'burst' firing. This latter is evident in a small fraction of MCCs but occurs regularly when Nav1.3/1.7 channels are made less available or when the Slo1β2-subunit responsible for BK channel inactivation is deleted. Burst firing causes large increases of Ca2+ -entry and potentiates CA release by ∼3.5-fold and thus may be a key mechanism for regulating MCC function. With the aim to uncover a physiological role for burst-firing we investigated the effects of acidosis on MCC activity. Lowering the extracellular pH (pHo ) from 7.4 to 7.0 and 6.6 induces cell depolarizations of 10-15 mV that generate repeated bursts. Bursts at pHo 6.6 lasted ∼330 ms, occurred at 1-2 Hz and caused an ∼7-fold increase of CA cumulative release. Burst firing originates from the inhibition of the pH-sensitive TASK-1/TASK-3 channels and from a 40% BK channel conductance reduction at pHo 7.0. The same pHo had little or no effect on Nav, Cav, Kv and SK channels that support AP firing in MCCs. Burst firing of pHo 6.6 could be mimicked by mixtures of the TASK-1 blocker A1899 (300 nm) and BK blocker paxilline (300 nm) and could be prevented by blocking L-type channels by adding 3 μm nifedipine. Mixtures of the two blockers raised cumulative CA-secretion even more than low pHo (∼12-fold), showing that the action of protons on vesicle release is mainly a result of the ionic conductance changes that increase Ca2+ -entry during bursts. Our data provide direct evidence suggesting that MCCs respond to low pHo with sustained depolarization, burst firing and enhanced CA-secretion, thus mimicking the physiological response of CCs to acute acidosis and hyperkalaemia generated during heavy exercise and muscle fatigue.
Collapse
Affiliation(s)
- Laura Guarina
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, NIS Centre, CNISM Unit, Torino, Italy
| | - David H F Vandael
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, NIS Centre, CNISM Unit, Torino, Italy.,Present address: Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, Austria
| | - Valentina Carabelli
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, NIS Centre, CNISM Unit, Torino, Italy
| | - Emilio Carbone
- Department of Drug Science, Laboratory of Cellular and Molecular Neuroscience, NIS Centre, CNISM Unit, Torino, Italy
| |
Collapse
|
27
|
Competition of calcified calmodulin N lobe and PIP2 to an LQT mutation site in Kv7.1 channel. Proc Natl Acad Sci U S A 2017; 114:E869-E878. [PMID: 28096388 DOI: 10.1073/pnas.1612622114] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Voltage-gated potassium 7.1 (Kv7.1) channel and KCNE1 protein coassembly forms the slow potassium current IKS that repolarizes the cardiac action potential. The physiological importance of the IKS channel is underscored by the existence of mutations in human Kv7.1 and KCNE1 genes, which cause cardiac arrhythmias, such as the long-QT syndrome (LQT) and atrial fibrillation. The proximal Kv7.1 C terminus (CT) binds calmodulin (CaM) and phosphatidylinositol-4,5-bisphosphate (PIP2), but the role of CaM in channel function is still unclear, and its possible interaction with PIP2 is unknown. Our recent crystallographic study showed that CaM embraces helices A and B with the apo C lobe and calcified N lobe, respectively. Here, we reveal the competition of PIP2 and the calcified CaM N lobe to a previously unidentified site in Kv7.1 helix B, also known to harbor an LQT mutation. Protein pulldown, molecular docking, molecular dynamics simulations, and patch-clamp recordings indicate that residues K526 and K527 in Kv7.1 helix B form a critical site where CaM competes with PIP2 to stabilize the channel open state. Data indicate that both PIP2 and Ca2+-CaM perform the same function on IKS channel gating by producing a left shift in the voltage dependence of activation. The LQT mutant K526E revealed a severely impaired channel function with a right shift in the voltage dependence of activation, a reduced current density, and insensitivity to gating modulation by Ca2+-CaM. The results suggest that, after receptor-mediated PIP2 depletion and increased cytosolic Ca2+, calcified CaM N lobe interacts with helix B in place of PIP2 to limit excessive IKS current inhibition.
Collapse
|
28
|
Mullins FM, Yen M, Lewis RS. Orai1 pore residues control CRAC channel inactivation independently of calmodulin. ACTA ACUST UNITED AC 2016; 147:137-52. [PMID: 26809793 PMCID: PMC4727942 DOI: 10.1085/jgp.201511437] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Researchers have reevaluated the role of calmodulin and previously identified calmodulin binding sites in the mechanism by which Ca2+-release activated Ca2+ channels can be inactivated as Ca2+ ions enter cells. Ca2+ entry through CRAC channels causes fast Ca2+-dependent inactivation (CDI). Previous mutagenesis studies have implicated Orai1 residues W76 and Y80 in CDI through their role in binding calmodulin (CaM), in agreement with the crystal structure of Ca2+–CaM bound to an Orai1 N-terminal peptide. However, a subsequent Drosophila melanogaster Orai crystal structure raises concerns about this model, as the side chains of W76 and Y80 are predicted to face the pore lumen and create a steric clash between bound CaM and other Orai1 pore helices. We further tested the functional role of CaM using several dominant-negative CaM mutants, none of which affected CDI. Given this evidence against a role for pretethered CaM, we altered side-chain volume and charge at the Y80 and W76 positions to better understand their roles in CDI. Small side chain volume had different effects at the two positions: it accelerated CDI at position Y80 but reduced the extent of CDI at position W76. Positive charges at Y80 and W76 permitted partial CDI with accelerated kinetics, whereas introducing negative charge at any of five consecutive pore-lining residues (W76, Y80, R83, K87, or R91) completely eliminated CDI. Noise analysis of Orai1 Y80E and Y80K currents indicated that reductions in CDI for these mutations could not be accounted for by changes in unitary current or open probability. The sensitivity of CDI to negative charge introduced into the pore suggested a possible role for anion binding in the pore. However, although Cl− modulated the kinetics and extent of CDI, we found no evidence that CDI requires any single diffusible cytosolic anion. Together, our results argue against a CDI mechanism involving CaM binding to W76 and Y80, and instead support a model in which Orai1 residues Y80 and W76 enable conformational changes within the pore, leading to CRAC channel inactivation.
Collapse
Affiliation(s)
- Franklin M Mullins
- Department of Pathology and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Michelle Yen
- Department of Pathology and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Richard S Lewis
- Department of Pathology and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
29
|
Yu CC, Ko JS, Ai T, Tsai WC, Chen Z, Rubart M, Vatta M, Everett TH, George AL, Chen PS. Arrhythmogenic calmodulin mutations impede activation of small-conductance calcium-activated potassium current. Heart Rhythm 2016; 13:1716-23. [PMID: 27165696 DOI: 10.1016/j.hrthm.2016.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Indexed: 01/27/2023]
Abstract
BACKGROUND Apamin-sensitive small-conductance calcium-activated potassium (SK) channels are gated by intracellular Ca(2+) through a constitutive interaction with calmodulin. OBJECTIVE We hypothesize that arrhythmogenic human calmodulin mutations impede activation of SK channels. METHODS We studied 5 previously published calmodulin mutations (N54I, N98S, D96V, D130G, and F90L). Plasmids encoding either wild-type or mutant calmodulin were transiently transfected into human embryonic kidney 293 cells that stably express subtype 2 of SK protein channels (SK2 cells). Whole-cell voltage-clamp recording was used to determine apamin-sensitive current densities. We also performed optical mapping studies in normal murine hearts to determine the effects of apamin in hearts with (n=7) or without (n=3) pretreatment with sea anemone toxin. RESULTS SK2 cells transfected with wild-type calmodulin exhibited an apamin-sensitive current density of 33.6 pA/pF (31.4-36.5 pA/pF) (median and confidence interval 25th-75th percentile), which was significantly higher than that observed for cells transfected with N54I (17.0 pA/pF [14.0-27.7 pA/pF]; P = .016), F90L (22.6 pA/pF [20.3-24.3 pA/pF]; P = .011), D96V (13.0 pA/pF [10.9-15.8 pA/pF]; P = .003), N98S (13.7 pA/pF [8.8-20.4 pA/pF]; P = .005), and D130G (17.6 pA/pF [13.8-24.6 pA/pF]; P = .003). The decrease in SK2 current densities was not associated with a decrease in membrane protein expression or intracellular distribution of the channel protein. Apamin increased the ventricular action potential duration at 80% repolarization (from 79.6 ms [63.4-93.3 ms] to 121.8 ms [97.9-127.2 ms]; P = .010) in hearts pretreated with anemone toxin but not in control hearts. CONCLUSION Human arrhythmogenic calmodulin mutations impede the activation of SK2 channels in human embryonic kidney 293 cells.
Collapse
Affiliation(s)
- Chih-Chieh Yu
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana; Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jum-Suk Ko
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana; Division of Cardiology, Department of Internal Medicine, Wonkwang University School of Medicine and Hospital, Iksan, Korea
| | - Tomohiko Ai
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana; Department of Molecular Pathogenesis, Medical Research Institute; Department of Emergency Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Wen-Chin Tsai
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana; Hualien Tzu-Chi General Hospital, Hualien City, Taiwan
| | - Zhenhui Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Michael Rubart
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana; Department of Pediatrics, Riley Heart Research Center
| | - Matteo Vatta
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Thomas H Everett
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Peng-Sheng Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
30
|
Fast-Response Calmodulin-Based Fluorescent Indicators Reveal Rapid Intracellular Calcium Dynamics. Sci Rep 2015; 5:15978. [PMID: 26527405 PMCID: PMC4630588 DOI: 10.1038/srep15978] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/01/2015] [Indexed: 01/16/2023] Open
Abstract
Faithful reporting of temporal patterns of intracellular Ca2+ dynamics requires the working range of indicators to match the signals. Current genetically encoded calmodulin-based fluorescent indicators are likely to distort fast Ca2+ signals by apparent saturation and integration due to their limiting fluorescence rise and decay kinetics. A series of probes was engineered with a range of Ca2+ affinities and accelerated kinetics by weakening the Ca2+-calmodulin-peptide interactions. At 37 °C, the GCaMP3-derived probe termed GCaMP3fast is 40-fold faster than GCaMP3 with Ca2+ decay and rise times, t1/2, of 3.3 ms and 0.9 ms, respectively, making it the fastest to-date. GCaMP3fast revealed discreet transients with significantly faster Ca2+ dynamics in neonatal cardiac myocytes than GCaMP6f. With 5-fold increased two-photon fluorescence cross-section for Ca2+ at 940 nm, GCaMP3fast is suitable for deep tissue studies. The green fluorescent protein serves as a reporter providing important novel insights into the kinetic mechanism of target recognition by calmodulin. Our strategy to match the probe to the signal by tuning the affinity and hence the Ca2+ kinetics of the indicator is applicable to the emerging new generations of calmodulin-based probes.
Collapse
|
31
|
Manganese-Enhanced MRI Reflects Both Activity-Independent and Activity-Dependent Uptake within the Rat Habenulomesencephalic Pathway. PLoS One 2015; 10:e0127773. [PMID: 26009889 PMCID: PMC4443977 DOI: 10.1371/journal.pone.0127773] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/20/2015] [Indexed: 11/19/2022] Open
Abstract
Manganese-enhanced magnetic resonance imaging (MEMRI) is a powerful technique for assessing the functional connectivity of neurons within the central nervous system. Despite the widely held proposition that MEMRI signal is dependent on neuronal activity, few studies have directly tested this implicit hypothesis. In the present series of experiments, MnCl2 was injected into the habenula of urethane-anesthetized rats alone or in combination with drugs known to alter neuronal activity by modulating specific voltage- and/or ligand-gated ion channels. Continuous quantitative T1 mapping was used to measure Mn2+ accumulation in the interpeduncular nucleus, a midline structure in which efferents from the medial habenula terminate. Microinjection of MnCl2 into the habenular complex using a protocol that maintained spontaneous neuronal activity resulted in a time-dependent increase in MEMRI signal intensity in the interpeduncular nucleus consistent with fast axonal transport of Mn2+ between these structures. Co-injection of the excitatory amino-acid agonist AMPA, increased the Mn2+-enhanced signal intensity within the interpeduncular nucleus. AMPA-induced increases in MEMRI signal were attenuated by co-injection of either the sodium channel blocker, TTX, or broad-spectrum Ca2+ channel blocker, Ni2+, and were occluded in the presence of both channel blockers. However, neither Ni2+ nor TTX, alone or in combination, attenuated the increase in signal intensity following injection of Mn2+ into the habenula. These results support the premise that changes in neuronal excitability are reflected by corresponding changes in MEMRI signal intensity. However, they also suggest that basal rates of Mn2+ uptake by neurons in the medial habenula may also occur via activity-independent mechanisms.
Collapse
|
32
|
Abstract
Calcium ions are Nature's most widely used signaling mechanism, mediating communication between pathways at virtually every physiological level. Ion channels are no exception, as the activities of a wide range of ion channels are intricately shaped by fluctuations in intracellular Ca(2+) levels. Mirroring the importance and the breadth of Ca(2+) signaling, free Ca(2+) levels are tightly controlled, and a myriad of Ca(2+) binding proteins transduce Ca(2+) signals, each with its own nuance, comprising a constantly changing symphony of metabolic activity. The founding member of Ca(2+) binding proteins is calmodulin (CaM), a small, acidic, modular protein endowed with gymnastic-like flexibility and E-F hand motifs that chelate Ca(2+) ions. In this review, I will trace the history that led to the realization that CaM serves as the Ca(2+)-gating cue for SK channels, the experiments that revealed that CaM is an intrinsic subunit of SK channels, and itself a target of regulation.
Collapse
Affiliation(s)
- John P Adelman
- a Vollum Institute ; Oregon Health & Science University ; Portland , OR USA
| |
Collapse
|
33
|
Magnolol and honokiol regulate the calcium-activated potassium channels signaling pathway in Enterotoxigenic Escherichia coli-induced diarrhea mice. Eur J Pharmacol 2015; 755:66-73. [DOI: 10.1016/j.ejphar.2015.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 12/19/2022]
|
34
|
Lu L, Sirish P, Zhang Z, Woltz RL, Li N, Timofeyev V, Knowlton AA, Zhang XD, Yamoah EN, Chiamvimonvat N. Regulation of gene transcription by voltage-gated L-type calcium channel, Cav1.3. J Biol Chem 2014; 290:4663-4676. [PMID: 25538241 DOI: 10.1074/jbc.m114.586883] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Cav1.3 L-type Ca(2+) channel is known to be highly expressed in neurons and neuroendocrine cells. However, we have previously demonstrated that the Cav1.3 channel is also expressed in atria and pacemaking cells in the heart. The significance of the tissue-specific expression of the channel is underpinned by our previous demonstration of atrial fibrillation in a Cav1.3 null mutant mouse model. Indeed, a recent study has confirmed the critical roles of Cav1.3 in the human heart (Baig, S. M., Koschak, A., Lieb, A., Gebhart, M., Dafinger, C., Nürnberg, G., Ali, A., Ahmad, I., Sinnegger-Brauns, M. J., Brandt, N., Engel, J., Mangoni, M. E., Farooq, M., Khan, H. U., Nürnberg, P., Striessnig, J., and Bolz, H. J. (2011) Nat. Neurosci. 14, 77-84). These studies suggest that detailed knowledge of Cav1.3 may have broad therapeutic ramifications in the treatment of cardiac arrhythmias. Here, we tested the hypothesis that there is a functional cross-talk between the Cav1.3 channel and a small conductance Ca(2+)-activated K(+) channel (SK2), which we have documented to be highly expressed in human and mouse atrial myocytes. Specifically, we tested the hypothesis that the C terminus of Cav1.3 may translocate to the nucleus where it functions as a transcriptional factor. Here, we reported for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcriptional regulator to modulate the function of Ca(2+)-activated K(+) channels in atrial myocytes. Nuclear translocation of the C-terminal domain of Cav1.3 is directly regulated by intracellular Ca(2+). Utilizing a Cav1.3 null mutant mouse model, we demonstrate that ablation of Cav1.3 results in a decrease in the protein expression of myosin light chain 2, which interacts and increases the membrane localization of SK2 channels.
Collapse
Affiliation(s)
- Ling Lu
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616,; the College of Life Sciences, Nanjing Normal University, Nanjing 210046, China.
| | - Padmini Sirish
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616
| | - Zheng Zhang
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616
| | - Ryan L Woltz
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616
| | - Ning Li
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616
| | - Valeriy Timofeyev
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616
| | - Anne A Knowlton
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616,; the Department of Veterans Affairs, Northern California Health Care System, Mather, California 95655
| | - Xiao-Dong Zhang
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616
| | - Ebenezer N Yamoah
- the Department of Physiology, School of Medicine, University of Nevada, Reno, Nevada 89557, and.
| | - Nipavan Chiamvimonvat
- From the Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, California 95616,; the Department of Veterans Affairs, Northern California Health Care System, Mather, California 95655,.
| |
Collapse
|
35
|
Abstract
There is an urgent need to identify novel interventions for mitigating the progression of diabetic nephropathy. Diabetic nephropathy is characterized by progressive renal fibrosis, in which tubulointerstitial fibrosis has been shown to be the final common pathway of all forms of chronic progressive renal disease, including diabetic nephropathy. Therefore targeting the possible mechanisms that drive this process may provide novel therapeutics which allow the prevention and potentially retardation of the functional decline in diabetic nephropathy. Recently, the Ca2+-activated K+ channel KCa3.1 (KCa3.1) has been suggested as a potential therapeutic target for nephropathy, based on its ability to regulate Ca2+ entry into cells and modulate Ca2+-signalling processes. In the present review, we focus on the physiological role of KCa3.1 in those cells involved in the tubulointerstitial fibrosis, including proximal tubular cells, fibroblasts, inflammatory cells (T-cells and macrophages) and endothelial cells. Collectively these studies support further investigation into KCa3.1 as a therapeutic target in diabetic nephropathy.
Collapse
|
36
|
Pachuau J, Li DP, Chen SR, Lee HA, Pan HL. Protein kinase CK2 contributes to diminished small conductance Ca2+-activated K+ channel activity of hypothalamic pre-sympathetic neurons in hypertension. J Neurochem 2014; 130:657-67. [PMID: 24806793 DOI: 10.1111/jnc.12758] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/05/2014] [Accepted: 05/06/2014] [Indexed: 11/29/2022]
Abstract
Small conductance calcium-activated K(+) (SK) channels regulate neuronal excitability. However, little is known about changes in SK channel activity of pre-sympathetic neurons in the hypothalamic paraventricular nucleus (PVN) in essential hypertension. SK channels, calmodulin, and casein kinase II (CK2) form a molecular complex. Because CK2 is up-regulated in the PVN in spontaneously hypertensive rats (SHRs), we hypothesized that CK2 increases calmodulin phosphorylation and contributes to diminished SK channel activity in PVN pre-sympathetic neurons in SHRs. Perforated whole-cell recordings were performed on retrogradely labeled spinally projecting PVN neurons in Wistar-Kyoto (WKY) rats and SHRs. Blocking SK channels with apamin significantly increased the firing rate of PVN neurons in WKY rats but not in SHRs. CK2 inhibition restored the stimulatory effect of apamin on the firing activity of PVN neurons in SHRs. Furthermore, apamin-sensitive SK currents and depolarization-induced medium after-hyperpolarization potentials of PVN neurons were significantly larger in WKY rats than in SHRs. CK2 inhibition significantly increased the SK channel current and medium after-depolarization potential of PVN neurons in SHRs. In addition, CK2-mediated calmodulin phosphorylation level in the PVN was significantly higher in SHRs than in WKY rats. Although SK3 was detected in the PVN, its expression level did not differ significantly between SHRs and WKY rats. Our findings suggest that CK2-mediated calmodulin phosphorylation is increased and contributes to diminished SK channel function of PVN pre-sympathetic neurons in SHRs. This information advances our understanding of the mechanisms underlying hyperactivity of PVN pre-sympathetic neurons and increased sympathetic vasomotor tone in hypertension. Small conductance calcium-activated K(+) (SK) channels, calmodulin, and protein kinase CK2 form a molecular complex and regulate neuronal excitability. Our study suggests that augmented CK2 activity in hypertension can increase calmodulin (CaM) phosphorylation, which leads to diminished SK channel function in pre-sympathetic neurons. Diminished SK channel activity plays a role in hyperactivity of pre-sympathetic neurons in the hypothalamus in hypertension.
Collapse
Affiliation(s)
- Judith Pachuau
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
37
|
Mu YH, Zhao WC, Duan P, Chen Y, Zhao WD, Wang Q, Tu HY, Zhang Q. RyR2 modulates a Ca2+-activated K+ current in mouse cardiac myocytes. PLoS One 2014; 9:e94905. [PMID: 24747296 PMCID: PMC3991633 DOI: 10.1371/journal.pone.0094905] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 03/21/2014] [Indexed: 11/19/2022] Open
Abstract
In cardiomyocytes, Ca2+ entry through voltage-dependent Ca2+ channels (VDCCs) binds to and activates RyR2 channels, resulting in subsequent Ca2+ release from the sarcoplasmic reticulum (SR) and cardiac contraction. Previous research has documented the molecular coupling of small-conductance Ca2+-activated K+ channels (SK channels) to VDCCs in mouse cardiac muscle. Little is known regarding the role of RyRs-sensitive Ca2+ release in the SK channels in cardiac muscle. In this study, using whole-cell patch clamp techniques, we observed that a Ca2+-activated K+ current (IK,Ca) recorded from isolated adult C57B/L mouse atrial myocytes was significantly decreased by ryanodine, an inhibitor of ryanodine receptor type 2 (RyR2), or by the co-application of ryanodine and thapsigargin, an inhibitor of the sarcoplasmic reticulum calcium ATPase (SERCA) (p<0.05, p<0.01, respectively). The activation of RyR2 by caffeine increased the IK,Ca in the cardiac cells (p<0.05, p<0.01, respectively). We further analyzed the effect of RyR2 knockdown on IK,Ca and Ca2+ in isolated adult mouse cardiomyocytes using a whole-cell patch clamp technique and confocal imaging. RyR2 knockdown in mouse atrial cells transduced with lentivirus-mediated small hairpin interference RNA (shRNA) exhibited a significant decrease in IK,Ca (p<0.05) and [Ca2+]i fluorescence intensity (p<0.01). An immunoprecipitated complex of SK2 and RyR2 was identified in native cardiac tissue by co-immunoprecipitation assays. Our findings indicate that RyR2-mediated Ca2+ release is responsible for the activation and modulation of SK channels in cardiac myocytes.
Collapse
Affiliation(s)
- Yong-hui Mu
- Department of Physiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Science, Xinxiang Medical College, Xinxiang, Henan, China
| | - Wen-chao Zhao
- Department of Physiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Ping Duan
- Department of Physiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yun Chen
- Department of Physiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Wei-da Zhao
- Department of Biological Engineering, University of Henan, Kaifeng, Henan, China
| | - Qian Wang
- Department of Physiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Hui-yin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Qian Zhang
- Department of Physiology, School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
- * E-mail:
| |
Collapse
|
38
|
KCa and Ca(2+) channels: the complex thought. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2322-33. [PMID: 24613282 DOI: 10.1016/j.bbamcr.2014.02.019] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/13/2014] [Accepted: 02/26/2014] [Indexed: 01/30/2023]
Abstract
Potassium channels belong to the largest and the most diverse super-families of ion channels. Among them, Ca(2+)-activated K(+) channels (KCa) comprise many members. Based on their single channel conductance they are divided into three subfamilies: big conductance (BKCa), intermediate conductance (IKCa) and small conductance (SKCa; SK1, SK2 and SK3). Ca(2+) channels are divided into two main families, voltage gated/voltage dependent Ca(2+) channels and non-voltage gated/voltage independent Ca(2+) channels. Based on their electrophysiological and pharmacological properties and on the tissue where there are expressed, voltage gated Ca(2+) channels (Cav) are divided into 5 families: T-type, L-type, N-type, P/Q-type and R-type Ca(2+). Non-voltage gated Ca(2+) channels comprise the TRP (TRPC, TRPV, TRPM, TRPA, TRPP, TRPML and TRPN) and Orai (Orai1 to Orai3) families and their partners STIM (STIM1 to STIM2). A depolarization is needed to activate voltage-gated Ca(2+) channels while non-voltage gated Ca(2+) channels are activated by Ca(2+) depletion of the endoplasmic reticulum stores (SOCs) or by receptors (ROCs). These two Ca(2+) channel families also control constitutive Ca(2+) entries. For reducing the energy consumption and for the fine regulation of Ca(2+), KCa and Ca(2+) channels appear associated as complexes in excitable and non-excitable cells. Interestingly, there is now evidence that KCa-Ca(2+) channel complexes are also found in cancer cells and contribute to cancer-associated functions such as cell proliferation, cell migration and the capacity to develop metastases. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.
Collapse
|
39
|
Alaimo A, Alberdi A, Gomis-Perez C, Fernández-Orth J, Bernardo-Seisdedos G, Malo C, Millet O, Areso P, Villarroel A. Pivoting between calmodulin lobes triggered by calcium in the Kv7.2/calmodulin complex. PLoS One 2014; 9:e86711. [PMID: 24489773 PMCID: PMC3904923 DOI: 10.1371/journal.pone.0086711] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/12/2013] [Indexed: 11/25/2022] Open
Abstract
Kv7.2 (KCNQ2) is the principal molecular component of the slow voltage gated M-channel, which strongly influences neuronal excitability. Calmodulin (CaM) binds to two intracellular C-terminal segments of Kv7.2 channels, helices A and B, and it is required for exit from the endoplasmic reticulum. However, the molecular mechanisms by which CaM controls channel trafficking are currently unknown. Here we used two complementary approaches to explore the molecular events underlying the association between CaM and Kv7.2 and their regulation by Ca2+. First, we performed a fluorometric assay using dansylated calmodulin (D-CaM) to characterize the interaction of its individual lobes to the Kv7.2 CaM binding site (Q2AB). Second, we explored the association of Q2AB with CaM by NMR spectroscopy, using 15N-labeled CaM as a reporter. The combined data highlight the interdependency of the N- and C-lobes of CaM in the interaction with Q2AB, suggesting that when CaM binds Ca2+ the binding interface pivots between the N-lobe whose interactions are dominated by helix B and the C-lobe where the predominant interaction is with helix A. In addition, Ca2+ makes CaM binding to Q2AB more difficult and, reciprocally, the channel weakens the association of CaM with Ca2+.
Collapse
Affiliation(s)
- Alessandro Alaimo
- Unidad de Biofísica, CSIC, UPV/EHU, Universidad del País Vasco, Leioa, Spain
| | - Araitz Alberdi
- Unidad de Biofísica, CSIC, UPV/EHU, Universidad del País Vasco, Leioa, Spain
| | | | | | | | - Covadonga Malo
- Unidad de Biofísica, CSIC, UPV/EHU, Universidad del País Vasco, Leioa, Spain
| | - Oscar Millet
- Structural Biology Unit, CICbioGUNE, Bizkaia Technology Park, Derio, Spain
| | - Pilar Areso
- Departamento de Farmacología, UPV/EHU, Universidad del País Vasco, Leioa, Spain
| | - Alvaro Villarroel
- Unidad de Biofísica, CSIC, UPV/EHU, Universidad del País Vasco, Leioa, Spain
- * E-mail:
| |
Collapse
|
40
|
Halling DB, Kenrick SA, Riggs AF, Aldrich RW. Calcium-dependent stoichiometries of the KCa2.2 (SK) intracellular domain/calmodulin complex in solution. ACTA ACUST UNITED AC 2014; 143:231-52. [PMID: 24420768 PMCID: PMC4001768 DOI: 10.1085/jgp.201311007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biophysical analyses indicate that the Ca2+-activated K+ channel SK2 binds calmodulin with multiple stoichiometries, distinct from the two SK2-two calmodulin stoichiometry identified by crystallography. Ca2+ activates SK Ca2+-activated K+ channels through the protein Ca2+ sensor, calmodulin (CaM). To understand how SK channels operate, it is necessary to determine how Ca2+ regulates CaM binding to its target on SK. Tagless, recombinant SK peptide (SKp), was purified for binding studies with CaM at low and high Ca2+ concentrations. Composition gradient multi-angle light scattering accurately measures the molar mass, stoichiometry, and affinity of protein complexes. In 2 mM Ca2+, SKp and CaM bind with three different stoichiometries that depend on the molar ratio of SKp:CaM in solution. These complexes include 28 kD 1SKp/1CaM, 39 kD 2SKp/1CaM, and 44 kD 1SKp/2CaM. A 2SKp/2CaM complex, observed in prior crystallographic studies, is absent. At <5 nM Ca2+, 1SKp/1CaM and 2SKp/1CaM were observed; however, 1SKp/2CaM was absent. Analytical ultracentrifugation was used to characterize the physical properties of the three SKp/CaM stoichiometries. In high Ca2+, the sedimentation coefficient is smaller for a 1SKp:1CaM solution than it is for either 2SKp:1CaM or 1SKp:2CaM. At low Ca2+ and at >100 µM protein concentrations, a molar excess of SKp over CaM causes aggregation. Aggregation is not observed in Ca2+ or with CaM in molar excess. In low Ca2+ both 1SKp:1CaM and 1SKp:2CaM solutions have similar sedimentation coefficients, which is consistent with the absence of a 1SKp/2CaM complex in low Ca2+. These results suggest that complexes with stoichiometries other than 2SKp/2CaM are important in gating.
Collapse
Affiliation(s)
- D Brent Halling
- Department of Neuroscience, 2 Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
| | | | | | | |
Collapse
|
41
|
Scholl ES, Pirone A, Cox DH, Duncan RK, Jacob MH. Alternative splice isoforms of small conductance calcium-activated SK2 channels differ in molecular interactions and surface levels. Channels (Austin) 2014; 8:62-75. [PMID: 24394769 PMCID: PMC4048344 DOI: 10.4161/chan.27470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Small conductance Ca2+-sensitive potassium (SK2) channels are voltage-independent, Ca2+-activated ion channels that conduct potassium cations and thereby modulate the intrinsic excitability and synaptic transmission of neurons and sensory hair cells. In the cochlea, SK2 channels are functionally coupled to the highly Ca2+ permeant α9/10-nicotinic acetylcholine receptors (nAChRs) at olivocochlear postsynaptic sites. SK2 activation leads to outer hair cell hyperpolarization and frequency-selective suppression of afferent sound transmission. These inhibitory responses are essential for normal regulation of sound sensitivity, frequency selectivity, and suppression of background noise. However, little is known about the molecular interactions of these key functional channels. Here we show that SK2 channels co-precipitate with α9/10-nAChRs and with the actin-binding protein α-actinin-1. SK2 alternative splicing, resulting in a 3 amino acid insertion in the intracellular 3′ terminus, modulates these interactions. Further, relative abundance of the SK2 splice variants changes during developmental stages of synapse maturation in both the avian cochlea and the mammalian forebrain. Using heterologous cell expression to separately study the 2 distinct isoforms, we show that the variants differ in protein interactions and surface expression levels, and that Ca2+ and Ca2+-bound calmodulin differentially regulate their protein interactions. Our findings suggest that the SK2 isoforms may be distinctly modulated by activity-induced Ca2+ influx. Alternative splicing of SK2 may serve as a novel mechanism to differentially regulate the maturation and function of olivocochlear and neuronal synapses.
Collapse
Affiliation(s)
- Elizabeth Storer Scholl
- Department of Neuroscience; Tufts University Sackler School of Graduate Biomedical Sciences; Boston, MA USA
| | - Antonella Pirone
- Department of Neuroscience; Tufts University Sackler School of Graduate Biomedical Sciences; Boston, MA USA
| | - Daniel H Cox
- Department of Neuroscience; Tufts University Sackler School of Graduate Biomedical Sciences; Boston, MA USA
| | - R Keith Duncan
- Department of Otolaryngology; University of Michigan; Ann Arbor, MI USA
| | - Michele H Jacob
- Department of Neuroscience; Tufts University Sackler School of Graduate Biomedical Sciences; Boston, MA USA
| |
Collapse
|
42
|
Tian C, Zhu R, Zhu L, Qiu T, Cao Z, Kang T. Potassium Channels: Structures, Diseases, and Modulators. Chem Biol Drug Des 2013; 83:1-26. [DOI: 10.1111/cbdd.12237] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Chuan Tian
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| | - Ruixin Zhu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Lixin Zhu
- Department of Pediatrics; Digestive Diseases and Nutrition Center; The State University of New York at Buffalo; Buffalo NY 14226 USA
| | - Tianyi Qiu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Zhiwei Cao
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Tingguo Kang
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| |
Collapse
|
43
|
González C, Baez-Nieto D, Valencia I, Oyarzún I, Rojas P, Naranjo D, Latorre R. K(+) channels: function-structural overview. Compr Physiol 2013; 2:2087-149. [PMID: 23723034 DOI: 10.1002/cphy.c110047] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Potassium channels are particularly important in determining the shape and duration of the action potential, controlling the membrane potential, modulating hormone secretion, epithelial function and, in the case of those K(+) channels activated by Ca(2+), damping excitatory signals. The multiplicity of roles played by K(+) channels is only possible to their mammoth diversity that includes at present 70 K(+) channels encoding genes in mammals. Today, thanks to the use of cloning, mutagenesis, and the more recent structural studies using x-ray crystallography, we are in a unique position to understand the origins of the enormous diversity of this superfamily of ion channels, the roles they play in different cell types, and the relations that exist between structure and function. With the exception of two-pore K(+) channels that are dimers, voltage-dependent K(+) channels are tetrameric assemblies and share an extremely well conserved pore region, in which the ion-selectivity filter resides. In the present overview, we discuss in the function, localization, and the relations between function and structure of the five different subfamilies of K(+) channels: (a) inward rectifiers, Kir; (b) four transmembrane segments-2 pores, K2P; (c) voltage-gated, Kv; (d) the Slo family; and (e) Ca(2+)-activated SK family, SKCa.
Collapse
Affiliation(s)
- Carlos González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | | | | | | | | | | |
Collapse
|
44
|
Brehme H, Kirschstein T, Schulz R, Köhling R. In vivo treatment with the casein kinase 2 inhibitor 4,5,6,7-tetrabromotriazole augments the slow afterhyperpolarizing potential and prevents acute epileptiform activity. Epilepsia 2013; 55:175-83. [DOI: 10.1111/epi.12474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hannes Brehme
- Oscar Langendorff Institute of Physiology; University of Rostock; Rostock Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology; University of Rostock; Rostock Germany
| | - Robert Schulz
- Oscar Langendorff Institute of Physiology; University of Rostock; Rostock Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology; University of Rostock; Rostock Germany
| |
Collapse
|
45
|
Ferreira R, Schlichter LC. Selective activation of KCa3.1 and CRAC channels by P2Y2 receptors promotes Ca(2+) signaling, store refilling and migration of rat microglial cells. PLoS One 2013; 8:e62345. [PMID: 23620825 PMCID: PMC3631179 DOI: 10.1371/journal.pone.0062345] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/20/2013] [Indexed: 12/26/2022] Open
Abstract
Microglial activation involves Ca(2+) signaling, and numerous receptors can evoke elevation of intracellular Ca(2+). ATP released from damaged brain cells can activate ionotropic and metabotropic purinergic receptors, and act as a chemoattractant for microglia. Metabotropic P2Y receptors evoke a Ca(2+) rise through release from intracellular Ca(2+) stores and store-operated Ca(2+) entry, and some have been implicated in microglial migration. This Ca(2+) rise is expected to activate small-conductance Ca(2+)-dependent K(+) (SK) channels, if present. We previously found that SK3 (KCa2.3) and KCa3.1 (SK4/IK1) are expressed in rat microglia and contribute to LPS-mediated activation and neurotoxicity. However, neither current has been studied by elevating Ca(2+) during whole-cell recordings. We hypothesized that, rather than responding only to Ca(2+), each channel type might be coupled to different receptor-mediated pathways. Here, our objective was to determine whether the channels are differentially activated by P2Y receptors, and, if so, whether they play differing roles. We used primary rat microglia and a rat microglial cell line (MLS-9) in which riluzole robustly activates both SK3 and KCa3.1 currents. Using electrophysiological, Ca(2+) imaging and pharmacological approaches, we show selective functional coupling of KCa3.1 to UTP-mediated P2Y2 receptor activation. KCa3.1 current is activated by Ca(2+) entry through Ca(2+)-release-activated Ca(2+) (CRAC/Orai1) channels, and both CRAC/Orai1 and KCa3.1 channels facilitate refilling of Ca(2+) stores. The Ca(2+) dependence of KCa3.1 channel activation was skewed to abnormally high concentrations, and we present evidence for a close physical association of the two channel types. Finally, migration of primary rat microglia was stimulated by UTP and inhibited by blocking either KCa3.1 or CRAC/Orai1 channels. This is the first report of selective coupling of one type of SK channel to purinergic stimulation of microglia, transactivation of KCa3.1 channels by CRAC/Orai1, and coordinated roles for both channels in store refilling, Ca(2+) signaling and microglial migration.
Collapse
Affiliation(s)
- Roger Ferreira
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Lyanne C. Schlichter
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Masada N, Schaks S, Jackson SE, Sinz A, Cooper DMF. Distinct mechanisms of calmodulin binding and regulation of adenylyl cyclases 1 and 8. Biochemistry 2012; 51:7917-29. [PMID: 22971080 PMCID: PMC3466776 DOI: 10.1021/bi300646y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Calmodulin (CaM), by mediating the stimulation of the activity of two adenylyl cyclases (ACs), plays a key role in integrating the cAMP and Ca(2+) signaling systems. These ACs, AC1 and AC8, by decoding discrete Ca(2+) signals can contribute to fine-tuning intracellular cAMP dynamics, particularly in neurons where they predominate. CaM comprises an α-helical linker separating two globular regions at the N-terminus and the C-terminus that each bind two Ca(2+) ions. These two lobes have differing affinities for Ca(2+), and they can interact with target proteins independently. This study explores previous indications that the two lobes of CaM can regulate AC1 and AC8 differently and thereby yield different responses to cellular transitions in [Ca(2+)](i). We first compared by glutathione S-transferase pull-down assays and offline nanoelectrospray ionization mass spectrometry the interaction of CaM and Ca(2+)-binding deficient mutants of CaM with the internal CaM binding domain (CaMBD) of AC1 and the two terminal CaMBDs of AC8. We then examined the influence of these three CaMBDs on Ca(2+) binding by native and mutated CaM in stopped-flow experiments to quantify their interactions. The three CaMBDs show quite distinct interactions with the two lobes of CaM. These findings establish the critical kinetic differences between the mechanisms of Ca(2+)-CaM activation of AC1 and AC8, which may underpin their different physiological roles.
Collapse
Affiliation(s)
- Nanako Masada
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | | | | | | | | |
Collapse
|
47
|
Balut CM, Hamilton KL, Devor DC. Trafficking of intermediate (KCa3.1) and small (KCa2.x) conductance, Ca(2+)-activated K(+) channels: a novel target for medicinal chemistry efforts? ChemMedChem 2012; 7:1741-55. [PMID: 22887933 DOI: 10.1002/cmdc.201200226] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 07/09/2012] [Indexed: 12/22/2022]
Abstract
Ca(2+)-activated K(+) (KCa) channels play a pivotal role in the physiology of a wide variety of tissues and disease states, including vascular endothelia, secretory epithelia, certain cancers, red blood cells (RBC), neurons, and immune cells. Such widespread involvement has generated an intense interest in elucidating the function and regulation of these channels, with the goal of developing pharmacological strategies aimed at selective modulation of KCa channels in various disease states. Herein we give an overview of the molecular and functional properties of these channels and their therapeutic importance. We discuss the achievements made in designing pharmacological tools that control the function of KCa channels by modulating their gating properties. Moreover, this review discusses the recent advances in our understanding of KCa channel assembly and anterograde trafficking toward the plasma membrane, the micro-domains in which these channels are expressed within the cell, and finally the retrograde trafficking routes these channels take following endocytosis. As the regulation of intracellular trafficking by agonists as well as the protein-protein interactions that modify these events continue to be explored, we anticipate this will open new therapeutic avenues for the targeting of these channels based on the pharmacological modulation of KCa channel density at the plasma membrane.
Collapse
Affiliation(s)
- Corina M Balut
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
48
|
Expression and Role of the Intermediate-Conductance Calcium-Activated Potassium Channel KCa3.1 in Glioblastoma. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:421564. [PMID: 22675627 PMCID: PMC3362965 DOI: 10.1155/2012/421564] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 03/15/2012] [Indexed: 12/29/2022]
Abstract
Glioblastomas are characterized by altered expression of several ion channels that have important consequences in cell functions associated with their aggressiveness, such as cell survival, proliferation, and migration. Data on the altered expression and function of the intermediate-conductance calcium-activated K (KCa3.1) channels in glioblastoma cells have only recently become available. This paper aims to (i) illustrate the main structural, biophysical, pharmacological, and modulatory properties of the KCa3.1 channel, (ii) provide a detailed account of data on the expression of this channel in glioblastoma cells, as compared to normal brain tissue, and (iii) critically discuss its major functional roles. Available data suggest that KCa3.1 channels (i) are highly expressed in glioblastoma cells but only scantly in the normal brain parenchima, (ii) play an important role in the control of glioblastoma cell migration. Altogether, these data suggest KCa3.1 channels as potential candidates for a targeted therapy against this tumor.
Collapse
|
49
|
Deignan J, Luján R, Bond C, Riegel A, Watanabe M, Williams JT, Maylie J, Adelman JP. SK2 and SK3 expression differentially affect firing frequency and precision in dopamine neurons. Neuroscience 2012; 217:67-76. [PMID: 22554781 DOI: 10.1016/j.neuroscience.2012.04.053] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 03/29/2012] [Accepted: 04/21/2012] [Indexed: 11/30/2022]
Abstract
The firing properties of dopamine (DA) neurons in the substantia nigra (SN) pars compacta are strongly influenced by the activity of apamin-sensitive small conductance Ca(2+)-activated K(+) (SK) channels. Of the three SK channel genes expressed in central neurons, only SK3 expression has been identified in DA neurons. The present findings show that SK2 was also expressed in DA neurons. Immuno-electron microscopy (iEM) showed that SK2 was primarily expressed in the distal dendrites, while SK3 was heavily expressed in the soma and, to a lesser extent, throughout the dendritic arbor. Electrophysiological recordings of the effects of the SK channel blocker apamin on DA neurons from wild type and SK(-/-) mice show that SK2-containing channels contributed to the precision of action potential (AP) timing, while SK3-containing channels influenced AP frequency. The expression of SK2 in DA neurons may endow distinct signaling and subcellular localization to SK2-containing channels.
Collapse
Affiliation(s)
- J Deignan
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Due to its high external and low internal concentration the Ca(2+) ion is used ubiquitously as an intracellular signaling molecule, and a great many Ca(2+)-sensing proteins have evolved to receive and propagate Ca(2+) signals. Among them are ion channel proteins, whose Ca(2+) sensitivity allows internal Ca(2+) to influence the electrical activity of cell membranes and to feedback-inhibit further Ca(2+) entry into the cytoplasm. In this review I will describe what is understood about the Ca(2+) sensing mechanisms of the three best studied classes of Ca(2+)-sensitive ion channels: Large-conductance Ca(2+)-activated K(+) channels, small-conductance Ca(2+)-activated K(+) channels, and voltage- gated Ca(2+) channels. Great strides in mechanistic understanding have be made for each of these channel types in just the past few years.
Collapse
Affiliation(s)
- Daniel H Cox
- Department of Neuroscience, Tufts University School of Medicine, Boston MA, 02420, USA.
| |
Collapse
|