1
|
Zimmermann J, Farooqi AR, van Rienen U. Electrical stimulation for cartilage tissue engineering - A critical review from an engineer's perspective. Heliyon 2024; 10:e38112. [PMID: 39416819 PMCID: PMC11481755 DOI: 10.1016/j.heliyon.2024.e38112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Cartilage has a limited intrinsic healing capacity. Hence, cartilage degradation and lesions pose a huge clinical challenge, particularly in an ageing society. Osteoarthritis impacts a significant number of the population and requires the development of repair and tissue engineering methods for hyaline articular cartilage. In this context, electrical stimulation has been investigated for more than 50 years already. Yet, no well-established clinical therapy to treat osteoarthritis by means of electrical stimulation exists. We argue that one reason is the lack of replicability of electrical stimulation devices from a technical perspective together with lacking hypotheses of the biophysical mechanism. Hence, first, the electrical stimulation studies reported in the context of cartilage tissue engineering with a special focus on technical details are summarized. Then, an experimental and numerical approach is discussed to make the electrical stimulation experiments replicable. Finally, biophysical hypotheses have been reviewed on the interaction of electric fields and cells that are relevant for cartilage tissue engineering. With that, the aim is to inspire future research to enable clinical electrical stimulation therapies to fight osteoarthritis.
Collapse
Affiliation(s)
- Julius Zimmermann
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
| | - Abdul Razzaq Farooqi
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Electronic Engineering, Faculty of Engineering, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Ursula van Rienen
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Ageing of Individuals and Society, Interdisciplinary Faculty, University of Rostock, 18051 Rostock, Germany
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany
| |
Collapse
|
2
|
Huang J, Fan X, Jin X, Lyu C, Guo Q, Liu T, Chen J, Davakan A, Lory P, Yan N. Structural basis for human Ca v3.2 inhibition by selective antagonists. Cell Res 2024; 34:440-450. [PMID: 38605177 PMCID: PMC11143251 DOI: 10.1038/s41422-024-00959-8] [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: 12/21/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
The Cav3.2 subtype of T-type calcium channels has been targeted for developing analgesics and anti-epileptics for its role in pain and epilepsy. Here we present the cryo-EM structures of Cav3.2 alone and in complex with four T-type calcium channel selective antagonists with overall resolutions ranging from 2.8 Å to 3.2 Å. The four compounds display two binding poses. ACT-709478 and TTA-A2 both place their cyclopropylphenyl-containing ends in the central cavity to directly obstruct ion flow, meanwhile extending their polar tails into the IV-I fenestration. TTA-P2 and ML218 project their 3,5-dichlorobenzamide groups into the II-III fenestration and place their hydrophobic tails in the cavity to impede ion permeation. The fenestration-penetrating mode immediately affords an explanation for the state-dependent activities of these antagonists. Structure-guided mutational analysis identifies several key residues that determine the T-type preference of these drugs. The structures also suggest the role of an endogenous lipid in stabilizing drug binding in the central cavity.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Xiao Fan
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Laboratory of Neurophysiology and Behavior, The Rockefeller University, New York, NY, USA.
| | - Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Chen Lyu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qinmeng Guo
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tao Liu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiaofeng Chen
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Amaël Davakan
- IGF, Université de Montpellier, CNRS, INSERM, LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Philippe Lory
- IGF, Université de Montpellier, CNRS, INSERM, LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
- Institute of Bio-Architecture and Bio-Interactions, Shenzhen Medical Academy of Research and Translation, Shenzhen, Guangdong, China.
| |
Collapse
|
3
|
Xu F, Cai W, Liu B, Qiu Z, Zhang X. Natural L-type calcium channels antagonists from Chinese medicine. Chin Med 2024; 19:72. [PMID: 38773596 PMCID: PMC11107034 DOI: 10.1186/s13020-024-00944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
L-type calcium channels (LTCCs), the largest subfamily of voltage-gated calcium channels (VGCCs), are the main channels for Ca2+ influx during extracellular excitation. LTCCs are widely present in excitable cells, especially cardiac and cardiovascular smooth muscle cells, and participate in various Ca2+-dependent processes. LTCCs have been considered as worthy drug target for cardiovascular, neurological and psychological diseases for decades. Natural products from Traditional Chinese medicine (TCM) have shown the potential as new drugs for the treatment of LTCCs related diseases. In this review, the basic structure, function of LTCCs, and the related human diseases caused by structural or functional abnormalities of LTCCs, and the natural LTCCs antagonist and their potential usages were summarized.
Collapse
Affiliation(s)
- Fangfang Xu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Wanna Cai
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Bo Liu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Zhenwen Qiu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| | - Xiaoqi Zhang
- Guangdong Provincial Engineering Research Center for Modernization of TCM, NMPA Key Laboratory for Quality Evaluation of TCM, Jinan University, Guangzhou, 510632, People's Republic of China.
| |
Collapse
|
4
|
Ramgoolam KH, Dolphin AC. Capsaicin-Induced Endocytosis of Endogenous Presynaptic Ca V2.2 in DRG-Spinal Cord Co-Cultures Inhibits Presynaptic Function. FUNCTION 2022; 4:zqac058. [PMID: 36540890 PMCID: PMC9761886 DOI: 10.1093/function/zqac058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/26/2022] Open
Abstract
The N-type calcium channel, CaV2.2 is key to neurotransmission from the primary afferent terminals of dorsal root ganglion (DRG) neurons to their postsynaptic targets in the spinal cord. In this study, we have utilized CaV2.2_HA knock-in mice, because the exofacial epitope tag in CaV2.2_HA enables accurate detection and localization of endogenous CaV2.2. CaV2.2_HA knock-in mice were used as a source of DRGs to exclusively study the presynaptic expression of N-type calcium channels in co-cultures between DRG neurons and wild-type spinal cord neurons. CaV2.2_HA is strongly expressed on the cell surface, particularly in TRPV1-positive small and medium DRG neurons. Super-resolution images of the presynaptic terminals revealed an increase in CaV2.2_HA expression and increased association with the postsynaptic marker Homer over time in vitro. Brief application of the TRPV1 agonist, capsaicin, resulted in a significant down-regulation of cell surface CaV2.2_HA expression in DRG neuron somata. At their presynaptic terminals, capsaicin caused a reduction in CaV2.2_HA proximity to and co-localization with the active zone marker RIM 1/2, as well as a lower contribution of N-type channels to single action potential-mediated Ca2+ influx. The mechanism of this down-regulation of CaV2.2_HA involves a Rab11a-dependent trafficking process, since dominant-negative Rab11a (S25N) occludes the effect of capsaicin on presynaptic CaV2.2_HA expression, and also prevents the effect of capsaicin on action potential-induced Ca2+ influx. Taken together, these data suggest that capsaicin causes a decrease in cell surface CaV2.2_HA expression in DRG terminals via a Rab11a-dependent endosomal trafficking pathway.
Collapse
Affiliation(s)
- Krishma H Ramgoolam
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| |
Collapse
|
5
|
Functional Postnatal Maturation of the Medial Olivocochlear Efferent-Outer Hair Cell Synapse. J Neurosci 2020; 40:4842-4857. [PMID: 32430293 DOI: 10.1523/jneurosci.2409-19.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/19/2020] [Accepted: 05/11/2020] [Indexed: 01/07/2023] Open
Abstract
The organ of Corti, the auditory mammalian sensory epithelium, contains two types of mechanotransducer cells, inner hair cells (IHCs) and outer hair cells (OHCs). IHCs are involved in conveying acoustic stimuli to the CNS, while OHCs are implicated in the fine tuning and amplification of sounds. OHCs are innervated by medial olivocochlear (MOC) cholinergic efferent fibers. The functional characteristics of the MOC-OHC synapse during maturation were assessed by electrophysiological and pharmacological methods in mouse organs of Corti at postnatal day 11 (P11)-P13, hearing onset in altricial rodents, and at P20-P22 when the OHCs are morphologically and functionally mature. Synaptic currents were recorded in whole-cell voltage-clamped OHCs while electrically stimulating the MOC fibers. A progressive increase in the number of functional MOC-OHC synapses, as well as in their strength and efficacy, was observed between P11-13 and P20-22. At hearing onset, the MOC-OHC synapse presented facilitation during MOC fibers high-frequency stimulation that disappeared at mature stages. In addition, important changes were found in the VGCC that are coupled to transmitter release. Ca2+ flowing in through L-type VGCCs contribute to trigger ACh release together with P/Q- and R-type VGCCs at P11-P13, but not at P20-P22. Interestingly, N-type VGCCs were found to be involved in this process at P20-P22, but not at hearing onset. Moreover, the degree of compartmentalization of calcium channels with respect to BK channels and presynaptic release components significantly increased from P11-P13 to P20-P22. These results suggest that the MOC-OHC synapse is immature at the onset of hearing.SIGNIFICANCE STATEMENT The functional expression of both VGCCs and BK channels, as well as their localization with respect to the presynaptic components involved in transmitter release, are key elements in determining synaptic efficacy. In this work, we show dynamic changes in the expression of VGCCs and Ca2+-dependent BK K+ channels coupled to ACh release at the MOC-OHC synapse and their shift in compartmentalization during postnatal maturation. These processes most likely set the short-term plasticity pattern and reliability of the MOC-OHC synapse on high-frequency activity.
Collapse
|
6
|
Corpus cavernosum smooth muscle cell dysfunction and phenotype transformation are related to erectile dysfunction in prostatitis rats with chronic prostatitis/chronic pelvic pain syndrome. JOURNAL OF INFLAMMATION-LONDON 2020; 17:2. [PMID: 31911760 PMCID: PMC6945598 DOI: 10.1186/s12950-019-0233-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/29/2019] [Indexed: 01/09/2023]
Abstract
Background The relationship between chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) and erectile dysfunction (ED) has been shown in many studies. However, the specific mechanism remains unclear. This study was to investigate the corpus cavernosum smooth muscle cell function and phenotype transformation in Experimental autoimmune prostatitis (EAP) rats. Methods EAP was induced in rats by using prostate protein supplemented with immuneadjuvant extraction, and the max-ICP and MAP were measured. IHC and Masson staining were done to assess inflammatory infiltration and collagen deposition in the corpus cavernosum, respectively. Subsequently, normal rat and EAP rat CCSMCs were purified by tissue block implantation and differential adherence method. The oxidative stress, smooth muscle phenotype transformation, cell cycle and intracellular calcium ion transport were also evaluated. Results The ratio of max ICP/MAP in EAP rats significantly reduced, and the TNF-α content and collagen deposition in the corpus cavernosum markedly increased as compared to healthy rats. High-purity rat CCSMCs were obtained. Oxidative stress was evident and the cGMP content decreased in the EAP rat CCSMCs. The expression of Cav1.2, IP3R1 and RyR2 increased, but the SERCA2 expression decreased in EAP rat CCSMCs, which was accompanied by increased intracellular calcium. Increased expression of OPN, collagen and KCa3.1, decreased Calponin expression and increased proportion of cells in the S phase were also observed in the EAP rat CCSMCs. Conclusion CP causes oxidative stress and imbalance of intracellular calcium in CCSMCs and promotes CCSMCs transformation from contractile to synthetic state, which may be involved in the pathogenesis of ED.
Collapse
|
7
|
Andrade A, Brennecke A, Mallat S, Brown J, Gomez-Rivadeneira J, Czepiel N, Londrigan L. Genetic Associations between Voltage-Gated Calcium Channels and Psychiatric Disorders. Int J Mol Sci 2019; 20:E3537. [PMID: 31331039 PMCID: PMC6679227 DOI: 10.3390/ijms20143537] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/23/2022] Open
Abstract
Psychiatric disorders are mental, behavioral or emotional disorders. These conditions are prevalent, one in four adults suffer from any type of psychiatric disorders world-wide. It has always been observed that psychiatric disorders have a genetic component, however, new methods to sequence full genomes of large cohorts have identified with high precision genetic risk loci for these conditions. Psychiatric disorders include, but are not limited to, bipolar disorder, schizophrenia, autism spectrum disorder, anxiety disorders, major depressive disorder, and attention-deficit and hyperactivity disorder. Several risk loci for psychiatric disorders fall within genes that encode for voltage-gated calcium channels (CaVs). Calcium entering through CaVs is crucial for multiple neuronal processes. In this review, we will summarize recent findings that link CaVs and their auxiliary subunits to psychiatric disorders. First, we will provide a general overview of CaVs structure, classification, function, expression and pharmacology. Next, we will summarize tools to study risk loci associated with psychiatric disorders. We will examine functional studies of risk variations in CaV genes when available. Finally, we will review pharmacological evidence of the use of CaV modulators to treat psychiatric disorders. Our review will be of interest for those studying pathophysiological aspects of CaVs.
Collapse
Affiliation(s)
- Arturo Andrade
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA.
| | - Ashton Brennecke
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Shayna Mallat
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Julian Brown
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | | | - Natalie Czepiel
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Laura Londrigan
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| |
Collapse
|
8
|
Kearney G, Zorrilla de San Martín J, Vattino LG, Elgoyhen AB, Wedemeyer C, Katz E. Developmental Synaptic Changes at the Transient Olivocochlear-Inner Hair Cell Synapse. J Neurosci 2019; 39:3360-3375. [PMID: 30755493 PMCID: PMC6495135 DOI: 10.1523/jneurosci.2746-18.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/04/2019] [Accepted: 01/30/2019] [Indexed: 12/18/2022] Open
Abstract
In the mature mammalian cochlea, inner hair cells (IHCs) are mainly innervated by afferent fibers that convey sound information to the CNS. During postnatal development, however, medial olivocochlear (MOC) efferent fibers transiently innervate the IHCs. The MOC-IHC synapse, functional from postnatal day 0 (P0) to hearing onset (P12), undergoes dramatic changes in the sensitivity to acetylcholine (ACh) and in the expression of key postsynaptic proteins. To evaluate whether there are associated changes in the properties of ACh release during this period, we used a cochlear preparation from mice of either sex at P4, P6-P7, and P9-P11 and monitored transmitter release from MOC terminals in voltage-clamped IHCs in the whole-cell configuration. The quantum content increased 5.6× from P4 to P9-P11 due to increases in the size and replenishment rate of the readily releasable pool of synaptic vesicles without changes in their probability of release or quantum size. This strengthening in transmission was accompanied by changes in short-term plasticity properties, which switched from facilitation at P4 to depression at P9-P11. We have previously shown that at P9-P11, ACh release is supported by P/Q- and N-type voltage-gated calcium channels (VGCCs) and negatively regulated by BK potassium channels activated by Ca2+ influx through L-type VGCCs. We now show that at P4 and P6-P7, release is mediated by P/Q-, R- and L-type VGCCs. Interestingly, L-type VGCCs have a dual role: they both support release and fuel BK channels, suggesting that at immature stages presynaptic proteins involved in release are less compartmentalized.SIGNIFICANCE STATEMENT During postnatal development before the onset of hearing, cochlear inner hair cells (IHCs) present spontaneous Ca2+ action potentials that release glutamate at the first auditory synapse in the absence of sound stimulation. The IHC Ca2+ action potential frequency pattern, which is crucial for the correct establishment and function of the auditory system, is regulated by the efferent medial olivocochlear (MOC) system that transiently innervates IHCs during this period. We show here that developmental changes in synaptic strength and synaptic plasticity properties at the MOC-IHC synapse upon MOC fiber activation at different frequencies might be crucial for tightly shaping the pattern of afferent activity during this critical period.
Collapse
Affiliation(s)
- Graciela Kearney
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
| | - Javier Zorrilla de San Martín
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
| | - Lucas G Vattino
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, 1121 Ciudad Autónoma de Buenos Aires, Argentina, and
| | - Carolina Wedemeyer
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
| | - Eleonora Katz
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina,
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. Ciudad Universitaria, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
9
|
Teleb M, Rizk OH, Zhang FX, Fronczek FR, Zamponi GW, Fahmy H. Synthesis of some new C2 substituted dihydropyrimidines and their electrophysiological evaluation as L-/T-type calcium channel blockers. Bioorg Chem 2019; 88:102915. [PMID: 31005784 DOI: 10.1016/j.bioorg.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/22/2019] [Accepted: 04/06/2019] [Indexed: 01/05/2023]
Abstract
Drugs targeting different calcium channel subtypes have strong therapeutic potential for future drug development for cardiovascular disorders, neuropsychiatric diseases and cancer. This study aims to design and synthesize a new series of C2 substituted dihydropyrimidines to mimic the structure features of third generation long acting dihydropyridine calcium channel blockers and dihydropyrimidines analogues. The target compounds have been evaluated as blockers for CaV1.2 and CaV3.2 utilizing the whole-cell patch clamp technique. Among the tested compounds, compound 7a showed moderate calcium channel blockade activity against CaV3.2. Moreover, the predicted physicochemical properties and pharmacokinetic profiles of the target compounds recommend that they can be considered as drug-like candidates. The results highlight some significant information for the future design of lead compounds as calcium channel blockers.
Collapse
Affiliation(s)
- Mohamed Teleb
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ola H Rizk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria 21311, Egypt
| | - Fang-Xiong Zhang
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4N1, Canada
| | - Frank R Fronczek
- Department of Chemistry, College of Science, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Gerald W Zamponi
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary T2N 4N1, Canada
| | - Hesham Fahmy
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA.
| |
Collapse
|
10
|
Teleb M, Rizk OH, Zhang FX, Fronczek FR, Zamponi GW, Fahmy H. Design, synthesis and pharmacological evaluation of some substituted dihydropyrimidines with L-/T-type calcium channel blocking activities. Bioorg Chem 2019; 83:354-366. [DOI: 10.1016/j.bioorg.2018.10.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/19/2018] [Accepted: 10/26/2018] [Indexed: 12/24/2022]
|
11
|
Mitochondrial Calcium Handling in Physiology and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:25-47. [PMID: 28551780 DOI: 10.1007/978-3-319-55330-6_2] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Calcium (Ca2+) accumulation inside mitochondria represents a pleiotropic signal controlling a wide range of cellular functions, including key metabolic pathways and life/death decisions. This phenomenon has been first described in the 1960s, but the identity of the molecules controlling this process remained a mystery until just few years ago, when both mitochondrial Ca2+ uptake and release systems were genetically dissected. This finally opened the possibility to develop genetic models to directly test the contribution of mitochondrial Ca2+ homeostasis to cellular functions. Here we summarize our current understanding of the molecular machinery that controls mitochondrial Ca2+ handling and critically evaluate the physiopathological role of mitochondrial Ca2+ signaling, based on recent evidences obtained through in vitro and in vivo models.
Collapse
|
12
|
Zamponi GW. Targeting voltage-gated calcium channels in neurological and psychiatric diseases. Nat Rev Drug Discov 2015; 15:19-34. [DOI: 10.1038/nrd.2015.5] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
13
|
Jha A, Singh AK, Weissgerber P, Freichel M, Flockerzi V, Flavell RA, Jha MK. Essential roles for Cavβ2 and Cav1 channels in thymocyte development and T cell homeostasis. Sci Signal 2015; 8:ra103. [DOI: 10.1126/scisignal.aac7538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
14
|
Marques R, Peres CG, Vaz CV, Gomes IM, Figueira MI, Cairrão E, Verde I, Maia CJ, Socorro S. 5α-Dihydrotestosterone regulates the expression of L-type calcium channels and calcium-binding protein regucalcin in human breast cancer cells with suppression of cell growth. Med Oncol 2015; 32:228. [PMID: 26255063 DOI: 10.1007/s12032-015-0676-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/20/2022]
Abstract
Androgens have been associated with the development of normal breast, and their role in mammary gland carcinogenesis has also been described. Several studies reported that androgens inhibit breast cancer cell growth, whereas others linked their action with the modulation of calcium (Ca(2+)) pumps, Ca(2+) channels and Ca(2+)-binding proteins. Also, it is known that deregulated Ca(2+) homeostasis has been implicated in the pathophysiology of breast. The L-type Ca(2+) channels (LTCCs) were found to be up-regulated in colon, colorectal and prostate cancer, but their presence in breast tissues remains uncharacterized. On the other hand, regucalcin (RGN) is a Ca(2+)-binding protein involved in the control of mammary gland cell proliferation, which has been identified as an androgen target gene in distinct tissues except breast. This study aimed to confirm the expression and activity of LTCCs in human breast cancer cells and investigate the effect of androgens in regulating the expression of α1C subunit (Cav1.2) of LTCCs and Ca(2+)-binding protein RGN. PCR, Western blot, immunofluorescence and electrophysiological experiments demonstrated the expression and activity of Cav1.2 subunit in MCF-7 cells. The MCF-7 cells were treated with 1, 10 or 100 nM of 5α-dihydrotestosterone (DHT) for 24-72 h. The obtained results showed that 1 nM DHT up-regulated the expression of Cav1.2 subunit while diminishing RGN protein levels, which was underpinned by reduced cell viability. These findings first confirmed the presence of LTCCs in breast cancer cells and opened new perspectives for the development of therapeutic approaches targeting Ca(2+) signaling.
Collapse
Affiliation(s)
- Ricardo Marques
- Faculdade de Ciências da Saúde, CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Albrizio M, Moramarco AM, Nicassio M, Micera E, Zarrilli A, Lacalandra GM. Localization and functional modification of L-type voltage-gated calcium channels in equine spermatozoa from fresh and frozen semen. Theriogenology 2014; 83:421-9. [PMID: 25459425 DOI: 10.1016/j.theriogenology.2014.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 10/01/2014] [Accepted: 10/05/2014] [Indexed: 01/13/2023]
Abstract
It is well known that insemination of cryopreserved semen always results in lower fertility when compared with fresh semen, but there is an increased interest and demand for frozen equine semen by the major breeder associations because of the utility arising from semen already "on hand" at breeding time. In this article, we report that equine sperm cells express L-type voltage-gated calcium channels; their localization is restricted to sperm neck and to the principal piece of the tail in both fresh and frozen-thawed spermatozoa. We also studied the causes of cryoinjury at the membrane level focusing on the function of L-type calcium channels. We report that in cryopreserved spermatozoa the mean basal value of [Ca(2+)]i is higher than that of spermatozoa from fresh semen (447.130 vs. 288.3 nM; P < 0.001) and L-type channels function differently in response to their agonist and antagonist in relation to semen condition (fresh or frozen-thawed). We found that on addition of agonist to the culture medium, the increase in intracellular calcium concentrations ([Ca(2+)]i) was greater in frozen semen than in fresh semen (Δ[Ca(2+)]i = 124.59 vs. 16.04 nM; P < 0.001), whereas after the addition of antagonist the decrease in [Ca(2+)]i was lower in frozen semen than in fresh semen (Δ[Ca(2+)]i = 32.5 vs. 82.5 nM; P < 0.001). In this article, we also discuss the impact of cryopreservation on sperm physiology.
Collapse
Affiliation(s)
- M Albrizio
- Department of Emergency and Organs Transplantation (DETO), Section of Veterinary Clinics and Animal Productions, University of Bari "Aldo Moro," Valenzano, Bari, Italy.
| | - A M Moramarco
- Department of Emergency and Organs Transplantation (DETO), Section of Veterinary Clinics and Animal Productions, University of Bari "Aldo Moro," Valenzano, Bari, Italy
| | - M Nicassio
- Department of Emergency and Organs Transplantation (DETO), Section of Veterinary Clinics and Animal Productions, University of Bari "Aldo Moro," Valenzano, Bari, Italy
| | - E Micera
- Department of Emergency and Organs Transplantation (DETO), Section of Veterinary Clinics and Animal Productions, University of Bari "Aldo Moro," Valenzano, Bari, Italy
| | - A Zarrilli
- Department of Emergency and Organs Transplantation (DETO), Section of Veterinary Clinics and Animal Productions, University of Bari "Aldo Moro," Valenzano, Bari, Italy
| | - G M Lacalandra
- Department of Emergency and Organs Transplantation (DETO), Section of Veterinary Clinics and Animal Productions, University of Bari "Aldo Moro," Valenzano, Bari, Italy
| |
Collapse
|
16
|
Maintz J, Cavdar M, Tamborski J, Kwaaitaal M, Huisman R, Meesters C, Kombrink E, Panstruga R. Comparative Analysis of MAMP-induced Calcium Influx in Arabidopsis Seedlings and Protoplasts. ACTA ACUST UNITED AC 2014; 55:1813-25. [DOI: 10.1093/pcp/pcu112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
Abstract
SNX-482, a peptide toxin isolated from tarantula venom, has become widely used as an inhibitor of Cav2.3 voltage-gated calcium channels. Unexpectedly, we found that SNX-482 dramatically reduced the A-type potassium current in acutely dissociated dopamine neurons from mouse substantia nigra pars compacta. The inhibition persisted when calcium was replaced by cobalt, showing that it was not secondary to a reduction of calcium influx. Currents from cloned Kv4.3 channels expressed in HEK-293 cells were inhibited by SNX-482 with an IC50 of <3 nM, revealing substantially greater potency than for SNX-482 inhibition of Cav2.3 channels (IC50 20-60 nM). At sub-saturating concentrations, SNX-482 produced a depolarizing shift in the voltage dependence of activation of Kv4.3 channels and slowed activation kinetics. Similar effects were seen on gating of cloned Kv4.2 channels, but the inhibition was less pronounced and required higher toxin concentrations. These results reveal SNX-482 as the most potent inhibitor of Kv4.3 channels yet identified. Because of the effects on both Kv4.3 and Kv4.2 channels, caution is needed when interpreting the effects of SNX-482 on cells and circuits where these channels are present.
Collapse
|
18
|
Predicting changes in cardiac myocyte contractility during early drug discovery with in vitro assays. Toxicol Appl Pharmacol 2014; 279:87-94. [DOI: 10.1016/j.taap.2014.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/12/2014] [Accepted: 06/07/2014] [Indexed: 11/22/2022]
|
19
|
Arranz-Tagarro JA, de los Ríos C, García AG, Padín JF. Recent patents on calcium channel blockers: emphasis on CNS diseases. Expert Opin Ther Pat 2014; 24:959-77. [DOI: 10.1517/13543776.2014.940892] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Senatore A, Guan W, Boone AN, Spafford JD. T-type channels become highly permeable to sodium ions using an alternative extracellular turret region (S5-P) outside the selectivity filter. J Biol Chem 2014; 289:11952-11969. [PMID: 24596098 DOI: 10.1074/jbc.m114.551473] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
T-type (Cav3) channels are categorized as calcium channels, but invertebrate ones can be highly sodium-selective channels. We illustrate that the snail LCav3 T-type channel becomes highly sodium-permeable through exon splicing of an extracellular turret and descending helix in domain II of the four-domain Cav3 channel. Highly sodium-permeable T-type channels are generated without altering the invariant ring of charged residues in the selectivity filter that governs calcium selectivity in calcium channels. The highly sodium-permeant T-type channel expresses in the brain and is the only splice isoform expressed in the snail heart. This unique splicing of turret residues offers T-type channels a capacity to serve as a pacemaking sodium current in the primitive heart and brain in lieu of Nav1-type sodium channels and to substitute for voltage-gated sodium channels lacking in many invertebrates. T-type channels would also contribute substantially to sodium leak conductances at rest in invertebrates because of their large window currents.
Collapse
Affiliation(s)
- Adriano Senatore
- Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Wendy Guan
- Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Adrienne N Boone
- Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - J David Spafford
- Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada.
| |
Collapse
|
21
|
Kuryshev YA, Brown AM, Duzic E, Kirsch GE. Evaluating state dependence and subtype selectivity of calcium channel modulators in automated electrophysiology assays. Assay Drug Dev Technol 2014; 12:110-9. [PMID: 24579774 PMCID: PMC4657039 DOI: 10.1089/adt.2013.552] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Voltage-gated Ca2+ channels play essential roles in control of neurosecretion and muscle contraction. The pharmacological significance of Cav channels stem from their identification as the molecular targets of calcium blockers used in the treatment of cardiovascular diseases, such as hypertension, angina, and arrhythmia, and neurologic diseases, such as pain and seizure. It has been proposed that state-dependent Cav inhibitors, that is, those that preferentially bind to channels in open or inactivated states, may improve the therapeutic window over relatively state-independent Cav inhibitors. High-throughput fluorescent-based functional assays have been useful in screening chemical libraries to identify Cav inhibitors. However, hit confirmation, mechanism of action, and subtype selectivity are better suited to automated patch clamp assays that have sufficient capacity to handle the volume of compounds identified during screening, even of modest sized libraries (≤500,000 compounds), and the flexible voltage control that allows evaluation of state-dependent drug blocks. IonWorks Barracuda (IWB), the newest generation of IonWorks instruments, provides the opportunity to accelerate the Cav drug discovery studies in an automated patch clamp platform in 384-well format capable of medium throughput screening and profiling studies. We have validated hCav1.2, hCav2.1, hCav2.2, and hCav3.2 channels assays on the IWB platform (population patch clamp mode) and demonstrated that the biophysical characteristics of the channels (activation, inactivation, and steady-state inactivation) obtained with the IWB system are consistent with known subtype-specific characteristics. Using standard reference compounds (nifedipine, BAY K8644, verapamil, mibefradil, and pimozide), we demonstrated subtype-selective and state- and use-dependent characteristics of drug-channel interactions. Here we describe the design and validation of novel robust high-throughput Cav channel assays on the IWB platform. The assays can be used to screen focused compound libraries for state-dependent Cav channel antagonists, to prioritize compounds for potency or to counterscreen for Cav subtype selectivity.
Collapse
|
22
|
Ambrosino P, Soldovieri MV, Russo C, Taglialatela M. Activation and desensitization of TRPV1 channels in sensory neurons by the PPARα agonist palmitoylethanolamide. Br J Pharmacol 2013; 168:1430-44. [PMID: 23083124 DOI: 10.1111/bph.12029] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 10/03/2012] [Accepted: 10/10/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Palmitoylethanolamide (PEA) is an endogenous fatty acid amide displaying anti-inflammatory and analgesic actions. To investigate the molecular mechanism responsible for these effects, the ability of PEA and of pain-inducing stimuli such as capsaicin (CAP) or bradykinin (BK) to influence intracellular calcium concentrations ([Ca²⁺](i)) in peripheral sensory neurons, has been assessed in the present study. The potential involvement of the transcription factor PPARα and of TRPV1 channels in PEA-induced effects was also studied. EXPERIMENTAL APPROACH [Ca²⁺](i) was evaluated by single-cell microfluorimetry in differentiated F11 cells. Activation of TRPV1 channels was assessed by imaging and patch-clamp techniques in CHO cells transiently-transfected with rat TRPV1 cDNA. KEY RESULTS In F11 cells, PEA (1-30 μM) dose-dependently increased [Ca²⁺](i). The TRPV1 antagonists capsazepine (1 μM) and SB-366791 (1 μM), as well as the PPARα antagonist GW-6471 (10 μM), inhibited PEA-induced [Ca²⁺](i) increase; blockers of cannabinoid receptors were ineffective. PEA activated TRPV1 channels heterologously expressed in CHO cells; this effect appeared to be mediated at least in part by PPARα. When compared with CAP, PEA showed similar potency and lower efficacy, and caused stronger TRPV1 currents desensitization. Sub-effective PEA concentrations, closer to those found in vivo, counteracted CAP- and BK-induced [Ca²⁺](i) transients, as well as CAP-induced TRPV1 activation. CONCLUSIONS AND IMPLICATIONS Activation of PPARα and TRPV1 channels, rather than of cannabinoid receptors, largely mediate PEA-induced [Ca²⁺](i) transients in sensory neurons. Differential TRPV1 activation and desensitization by CAP and PEA might contribute to their distinct pharmacological profile, possibly translating into potentially relevant clinical differences.
Collapse
Affiliation(s)
- Paolo Ambrosino
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | | | | | | |
Collapse
|
23
|
Shin WS, Oh S, An SW, Park GM, Kwon D, Ham J, Lee S, Park BG. 5E- and 5Z-farnesylacetones from Sargassum siliquastrum as novel selective L-type calcium channel blockers. Vascul Pharmacol 2013; 58:299-306. [PMID: 23416245 DOI: 10.1016/j.vph.2013.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 01/23/2013] [Accepted: 02/04/2013] [Indexed: 11/21/2022]
Abstract
A specific blocker of L-type Ca(2+) channels may be useful in decreasing arterial tone by reducing the open-state probability of L-type Ca(2+) channels. The aim of the present study was to evaluate the farnesylacetones, which are major active constituents of Sargassum siliquastrum, regarding their vasodilatation efficacies, selectivities toward L-type Ca(2+) channels, and in vivo antihypertensive activities. The application of 5E-(farnesylacetone 311) or 5Z-farnesylacetone (farnesylacetone 312) induced concentration-dependent vasodilatation effects on the basilar artery that was pre-contracted with depolarization and showed an ignorable potential role of endothelial-derived nitric oxide. We also tested farnesylacetone 311 or 312 to determine their pharmacological profiles for the blockade of native L-type Ca(2+) channels in basilar arterial smooth muscle cells (BASMCs) and ventricular myocytes (VMCs), cloned L- (α1C/β2a/α2δ), N- (α1B/β1b/α2δ), and T-type Ca(2+) channels (α1G, α1H, and α1I). Farnesylacetone 311 or 312 showed greater selectivity toward the L-type Ca(2+) channels among the tested voltage-gated Ca(2+) channels. The ranked order of the potency for farnesylacetone 311 was cloned α1C≒L-type (BASMC)≒L-type (VMCs)>α1B>α1H>α1I>α1G and that for farnesylacetone 312 was cloned α1C≒L-type (BASMCs)≒L-type (VMCs)>α1H>α1G>α1B>α1I. The oral administration of the farnesylacetone 311 (80mg/kg) conferred potent, long-lasting antihypertensive activity in spontaneous hypertensive rats, but it did not alter the heart rate.
Collapse
Affiliation(s)
- Woon-Seob Shin
- Department of Microbiology, College of Medicine, Kwandong University, Gangneung, 210-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Farkas V, Szentandrássy N, Bárándi L, Hegyi B, Ruzsnavszky F, Ruzsnavszky O, Horváth B, Bányász T, Magyar J, Márton I, Nánási PP. Interaction between Ca(2+) channel blockers and isoproterenol on L-type Ca(2+) current in canine ventricular cardiomyocytes. Acta Physiol (Oxf) 2012; 206:42-50. [PMID: 22520840 DOI: 10.1111/j.1748-1716.2012.02448.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 01/31/2012] [Accepted: 04/17/2012] [Indexed: 11/28/2022]
Abstract
AIM The aim of this work was to study antagonistic interactions between the effects of various types of Ca(2+) channel blockers and isoproterenol on the amplitude of L-type Ca(2+) current in canine ventricular cells. METHODS Whole-cell version of the patch clamp technique was used to study the effect of isoproterenol on Ca(2+) current in the absence and presence of Ca(2+) channel-blocking agents, including nifedipine, nisoldipine, diltiazem, verapamil, CoCl(2) and MnCl(2) . RESULTS Five micromolar Nifedipine, 1 μM nisoldipine, 10 μM diltiazem, 5 μM verapamil, 3 mM CoCl(2) and 5 mM MnCl(2) evoked uniformly a 90-95% blockade of Ca(2+) current in the absence of isoproterenol. Isoproterenol (100 nM) alone increased the amplitude of Ca(2+) current from 6.8 ± 1.3 to 23.7 ± 2.2 pA/pF in a reversible manner. Isoproterenol caused a marked enhancement of Ca(2+) current even in the presence of nifedipine, nisoldipine, diltiazem and verapamil, but not in the presence of CoCl(2) or MnCl(2) . CONCLUSION The results indicate that the action of isoproterenol is different in the presence of organic and inorganic Ca(2+) channel blockers. CoCl(2) and MnCl(2) were able to fully prevent the effect of isoproterenol on Ca(2+) current, while the organic Ca(2+) channel blockers failed to do so. This has to be born in mind when the effects of organic Ca(2+) channel blockers are evaluated either experimentally or clinically under conditions of increased sympathetic tone.
Collapse
Affiliation(s)
- V. Farkas
- Department of Dentistry; University of Debrecen; Debrecen; Hungary
| | - N. Szentandrássy
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - L. Bárándi
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - B. Hegyi
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - F. Ruzsnavszky
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - O. Ruzsnavszky
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - B. Horváth
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - T. Bányász
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - J. Magyar
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| | - I. Márton
- Department of Dentistry; University of Debrecen; Debrecen; Hungary
| | - P. P. Nánási
- Department of Physiology; University of Debrecen; Debrecen; Hungary
| |
Collapse
|
25
|
Expression of a recombinant Phoneutria toxin active in calcium channels. Toxicon 2012; 60:907-18. [PMID: 22659539 DOI: 10.1016/j.toxicon.2012.05.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 04/24/2012] [Accepted: 05/24/2012] [Indexed: 11/20/2022]
Abstract
PnTx3-4 is a toxin isolated from the venom of the spider Phoneutria nigriventer that blocks N-, P/Q-, and R-type voltage-gated calcium channels and has great potential for clinical applications. In this report we used the SUMO system to express large amounts of recombinant PnTx3-4 peptide, which was found in both soluble and insoluble fractions of bacterial extracts. We purified the recombinant toxin from both fractions and showed that the recombinant peptide showed biological activity similar to the native PnTx3-4. In silico analysis of the primary sequence of PnTx3-4 indicated that the peptide conforms to all the criteria of a knottin scaffold. Additionally, circular dichroism spectrum analysis of the recombinant PnTx3-4 predicted that the toxin structure is composed of approximately 53% turns/unordered, 31% α-helix and 16% β-strand, which is consistent with predicted model of the PnTx3-4 knottin scaffold available at the knottin database (http://knottin.cbs.cnrs.fr). These studies provide the basis for future large scale production and structure-function investigation of PnTx3-4.
Collapse
|
26
|
Rosa JM, Nanclares C, Orozco A, Colmena I, de Pascual R, García AG, Gandía L. Regulation by L-Type Calcium Channels of Endocytosis: An Overview. J Mol Neurosci 2012; 48:360-7. [DOI: 10.1007/s12031-012-9786-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 04/22/2012] [Indexed: 11/29/2022]
|
27
|
O'Malley D, Liston M, Hyland NP, Dinan TG, Cryan JF. Colonic soluble mediators from the maternal separation model of irritable bowel syndrome activate submucosal neurons via an interleukin-6-dependent mechanism. Am J Physiol Gastrointest Liver Physiol 2011; 300:G241-52. [PMID: 21109592 DOI: 10.1152/ajpgi.00385.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Irritable bowel syndrome (IBS) is characterized by episodic bouts of abdominal pain, bloating, and altered bowel habit. Accumulating evidence has linked immune activation with IBS, including reports of increases in circulating levels of the proinflammatory cytokine interleukin (IL)-6. However, it is unknown whether IL-6 contributes directly to disease manifestation. As enteric nervous activity mediates motility and secretory function, the aims of this study were to determine the effects of IL-6 on submucosal neurons and related gastrointestinal (GI) function. In these studies, we examined the colons of maternally separated (MS) rats, which exhibit elevated circulating levels of IL-6 in addition to GI dysfunction. To our knowledge, these studies are the first to provide evidence of the sensitivity of submucosal neurons to colonic secretions from MS rats (n = 50, P < 0.05), thus recapitulating clinical biopsy data. Moreover, we demonstrated that the excitatory action is IL-6 dependent. Thereafter, the impact of IL-6 on neuronal and glial activation and absorpto/secretory function was pharmacologically characterized. Other proinflammatory cytokines including IL-8 (n = 30, P > 0.05), IL-1β (n = 56, P > 0.05), and TNF-α (n = 56, P > 0.05) excited fewer neurons. Both muscarinic and nicotinic cholinergic receptors participate in the effect and cause downstream activation of ERK, JAK-STAT, and NF-κB signaling cascades. Functionally, IL-6 increases transepithelial resistance and enhances neurally and cholinergically mediated ion transport. These data provide a role for IL-6 in colonic secretory functions and relate these effects to GI dysfunction in an animal model of IBS, thereby elucidating a potential relationship between circulating levels of IL-6 and aberrant GI function.
Collapse
Affiliation(s)
- Dervla O'Malley
- School of Pharmacy, Cavanagh Pharmacy Bldg., University College Cork, Cork, Ireland
| | | | | | | | | |
Collapse
|
28
|
Peptide neurotoxins that affect voltage-gated calcium channels: a close-up on ω-agatoxins. Toxins (Basel) 2011; 3:17-42. [PMID: 22069688 PMCID: PMC3210452 DOI: 10.3390/toxins3010017] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 12/23/2010] [Accepted: 12/30/2010] [Indexed: 12/02/2022] Open
Abstract
Peptide neurotoxins found in animal venoms have gained great interest in the field of neurotransmission. As they are high affinity ligands for calcium, potassium and sodium channels, they have become useful tools for studying channel structure and activity. Peptide neurotoxins represent the clinical potential of ion-channel modulators across several therapeutic fields, especially in developing new strategies for treatment of ion channel-related diseases. The aim of this review is to overview the latest updates in the domain of peptide neurotoxins that affect voltage-gated calcium channels, with a special focus on ω-agatoxins.
Collapse
|
29
|
Participation of L-type voltage-gated calcium channels in facilitation of long-term potentiation during the formation of morphine dependence in rats. Bull Exp Biol Med 2010; 150:203-5. [DOI: 10.1007/s10517-010-1105-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Singh A, Hildebrand ME, Garcia E, Snutch TP. The transient receptor potential channel antagonist SKF96365 is a potent blocker of low-voltage-activated T-type calcium channels. Br J Pharmacol 2010; 160:1464-75. [PMID: 20590636 DOI: 10.1111/j.1476-5381.2010.00786.x] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE SKF96365 (SKF), originally identified as a blocker of receptor-mediated calcium entry, is widely used diagnostically, as a blocker of transient receptor potential canonical type (TRPC) channels. While SKF has been used as a tool to define the functional roles of TRPC channels in various cell and tissue types, there are notable overlapping physiological and pathophysiological associations between TRPC channels and low-voltage-activated (LVA) T-type calcium channels. The activity of SKF against T-type Ca channels has not been previously explored, and here we systematically investigated the effects of SKF on recombinant and native voltage-gated Ca channel-mediated currents. EXPERIMENTAL APPROACH Effects of SKF on recombinant Ca channels were studied under whole-cell patch clamp conditions after expression in HEK293 cells. The effect of SKF on cerebellar Purkinje cells (PCs) expressing native T-type Ca channels was also assessed. KEY RESULTS SKF blocked recombinant Ca channels, representative of each of the three main molecular genetic classes (Ca(V)1, Ca(V)2 and Ca(V)3) at concentrations typically utilized to assay TRPC function (10 microM). Particularly, human Ca(V)3.1 T-type Ca channels were more potently inhibited by SKF (IC(50) approximately 560 nM) in our experiments than previously reported for similarly expressed TRPC channels. SKF also inhibited native Ca(V)3.1 T-type currents in a rat cerebellar PC slice preparation. CONCLUSIONS AND IMPLICATIONS SKF was a potent blocker of LVA T-type Ca channels. We suggest caution in the interpretation of results using SKF alone as a diagnostic agent for TRPC activity in native tissues.
Collapse
Affiliation(s)
- A Singh
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
31
|
Ca(2+) and Ca(2+)-activated K(+) channels that support and modulate transmitter release at the olivocochlear efferent-inner hair cell synapse. J Neurosci 2010; 30:12157-67. [PMID: 20826678 DOI: 10.1523/jneurosci.2541-10.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In the mammalian auditory system, the synapse between efferent olivocochlear (OC) neurons and sensory cochlear hair cells is cholinergic, fast, and inhibitory. This efferent synapse is mediated by the nicotinic alpha9alpha10 receptor coupled to the activation of SK2 Ca(2+)-activated K(+) channels that hyperpolarize the cell. So far, the ion channels that support and/or modulate neurotransmitter release from the OC terminals remain unknown. To identify these channels, we used an isolated mouse cochlear preparation and monitored transmitter release from the efferent synaptic terminals in inner hair cells (IHCs) voltage clamped in the whole-cell recording configuration. Acetylcholine (ACh) release was evoked by electrically stimulating the efferent fibers that make axosomatic contacts with IHCs before the onset of hearing. Using the specific antagonists for P/Q- and N-type voltage-gated calcium channels (VGCCs), omega-agatoxin IVA and omega-conotoxin GVIA, respectively, we show that Ca(2+) entering through both types of VGCCs support the release process at this synapse. Interestingly, we found that Ca(2+) entering through the dihydropiridine-sensitive L-type VGCCs exerts a negative control on transmitter release. Moreover, using immunostaining techniques combined with electrophysiology and pharmacology, we show that BK Ca(2+)-activated K(+) channels are transiently expressed at the OC efferent terminals contacting IHCs and that their activity modulates the release process at this synapse. The effects of dihydropiridines combined with iberiotoxin, a specific BK channel antagonist, strongly suggest that L-type VGCCs negatively regulate the release of ACh by fueling BK channels that are known to curtail the duration of the terminal action potential in several types of neurons.
Collapse
|
32
|
Abbadie C, McManus OB, Sun SY, Bugianesi RM, Dai G, Haedo RJ, Herrington JB, Kaczorowski GJ, Smith MM, Swensen AM, Warren VA, Williams B, Arneric SP, Eduljee C, Snutch TP, Tringham EW, Jochnowitz N, Liang A, Euan MacIntyre D, McGowan E, Mistry S, White VV, Hoyt SB, London C, Lyons KA, Bunting PB, Volksdorf S, Duffy JL. Analgesic effects of a substituted N-triazole oxindole (TROX-1), a state-dependent, voltage-gated calcium channel 2 blocker. J Pharmacol Exp Ther 2010; 334:545-55. [PMID: 20439438 DOI: 10.1124/jpet.110.166363] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Voltage-gated calcium channel (Ca(v))2.2 (N-type calcium channels) are key components in nociceptive transmission pathways. Ziconotide, a state-independent peptide inhibitor of Ca(v)2.2 channels, is efficacious in treating refractory pain but exhibits a narrow therapeutic window and must be administered intrathecally. We have discovered an N-triazole oxindole, (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1), as a small-molecule, state-dependent blocker of Ca(v)2 channels, and we investigated the therapeutic advantages of this compound for analgesia. TROX-1 preferentially inhibited potassium-triggered calcium influx through recombinant Ca(v)2.2 channels under depolarized conditions (IC(50) = 0.27 microM) compared with hyperpolarized conditions (IC(50) > 20 microM). In rat dorsal root ganglion (DRG) neurons, TROX-1 inhibited omega-conotoxin GVIA-sensitive calcium currents (Ca(v)2.2 channel currents), with greater potency under depolarized conditions (IC(50) = 0.4 microM) than under hyperpolarized conditions (IC(50) = 2.6 microM), indicating state-dependent Ca(v)2.2 channel block of native as well as recombinant channels. TROX-1 fully blocked calcium influx mediated by a mixture of Ca(v)2 channels in calcium imaging experiments in rat DRG neurons, indicating additional block of all Ca(v)2 family channels. TROX-1 reversed inflammatory-induced hyperalgesia with maximal effects equivalent to nonsteroidal anti-inflammatory drugs, and it reversed nerve injury-induced allodynia to the same extent as pregabalin and duloxetine. In contrast, no significant reversal of hyperalgesia was observed in Ca(v)2.2 gene-deleted mice. Mild impairment of motor function in the Rotarod test and cardiovascular functions were observed at 20- to 40-fold higher plasma concentrations than required for analgesic activities. TROX-1 demonstrates that an orally available state-dependent Ca(v)2 channel blocker may achieve a therapeutic window suitable for the treatment of chronic pain.
Collapse
Affiliation(s)
- Catherine Abbadie
- Department of Pharmacology,Merck Research Laboratories, Rahway, New Jersey 07065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yarotskyy V, Elmslie KS. Open-state occupancy prevents gating charge relaxation of N-type (CaV2.2) calcium channels. Biophys J 2010; 97:2446-55. [PMID: 19883587 DOI: 10.1016/j.bpj.2009.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 07/17/2009] [Accepted: 08/06/2009] [Indexed: 10/20/2022] Open
Abstract
N-type and L-type channels have significant gating differences, and we wondered whether some of these differences are linked to the relationship between charge movement and channel opening. The time constants for N-channel closing (tau(Deact)) and Off-gating charge movement (tauQ(Off)) were compared over a range of voltages. tauQ(Off) was significantly larger than tau(Deact) at voltages < -10 mV, and the voltage dependence of the tauQ(Off) was less steep than that for tau(Deact), which suggests that gating charge relaxation does not limit channel closing. Roscovitine, a drug that slows N-channel closing by holding the channel in a high open-probability state, was found to slow both tauQ(Off) and tau(Deact), and thus the time courses of channel closing and gating charge relaxation were similar. Our gating current results were reproduced with the addition of a voltage-independent, closed-closed transition to our previously published two-open-state N-channel model. This work suggests that, like L-type channels, there is a voltage-independent transition along the N-channel activation/deactivation pathway, but this transition occurs between closed states instead of the closed-open states of the L-channel. Also unlike L-type channels, the gating charge appears to be locked into the activated position by the N-channel open state.
Collapse
Affiliation(s)
- Viktor Yarotskyy
- Department of Anesthesiology, Penn State College of Medicine, Penn State University, Hershey, Pennsylvania, USA
| | | |
Collapse
|
34
|
Baliño P, Pastor R, Aragon CMG. Participation of L-type calcium channels in ethanol-induced behavioral stimulation and motor incoordination: effects of diltiazem and verapamil. Behav Brain Res 2010; 209:196-204. [PMID: 20122967 DOI: 10.1016/j.bbr.2010.01.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 01/19/2010] [Accepted: 01/24/2010] [Indexed: 10/19/2022]
Abstract
Calcium flux through voltage gate calcium channels (VGCC) is involved in many neuronal processes such as membrane depolarization, gene expression, hormone secretion, and neurotransmitter release. Several studies have shown that either acute or chronic exposure to ethanol modifies calcium influx through high voltage activated channels. Of special relevance is the L-type VGCC. Pharmacological manipulation of L-type calcium channels affects ethanol intake, ethanol discrimination and manifestations of withdrawal syndrome. The present study investigates the role of L-type channels on the psychomotor effects (stimulation and sedation/ataxia) of ethanol by testing the effects of different L-type calcium channel blockers (CCB) on such behaviors. Mice were pretreated intraperitoneally with the CCB, diltiazem (0-40 mg/kg) or verapamil (0-30 mg/kg) 30 min before ethanol (0-3.5 g/kg). Locomotion was measured in an open field chamber for 20 min immediately after ethanol. The two CCB tested prevented locomotor stimulation, but not locomotor suppression produced by ethanol. Doses of the two CCB which reduced ethanol stimulation, did not alter spontaneous locomotion. The ataxic effects of ethanol (1.25 g/kg), measured with an accelerating rotarod task, were not affected by diltiazem (20mg/kg) or verapamil (15 mg/kg). In addition, our results indicated that ethanol is more sensitive to the antagonism of L-type calcium channels than other drugs with stimulant properties; doses of the two CCB that reduced ethanol stimulation did not reduce the psychomotor effects of amphetamine, caffeine or cocaine. In conclusion, these data provide further evidence of the important involvement of L-type calcium channels in the behavioral effects produced by ethanol.
Collapse
Affiliation(s)
- Pablo Baliño
- Area de Psicobiología, Universtitat Jaume I, Avda. Sos Baynat s/n, 12071 Castellón, Spain
| | | | | |
Collapse
|
35
|
Yarishkin OV, Hwang EM, Kim D, Yoo JC, Kang SS, Kim DR, Shin JHJ, Chung HJ, Jeong HS, Kang D, Han J, Park JY, Hong SG. Diclofenac, a Non-steroidal Anti-inflammatory Drug, Inhibits L-type Ca Channels in Neonatal Rat Ventricular Cardiomyocytes. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2009; 13:437-42. [PMID: 20054489 DOI: 10.4196/kjpp.2009.13.6.437] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 11/17/2009] [Accepted: 11/19/2009] [Indexed: 11/15/2022]
Abstract
A non-steroidal anti-inflammatory drug (NSAID) has many adverse effects including cardiovascular (CV) risk. Diclofenac among the nonselective NSAIDs has the highest CV risk such as congestive heart failure, which resulted commonly from the impaired cardiac pumping due to a disrupted excitation-contraction (E-C) coupling. We investigated the effects of diclofenac on the L-type calcium channels which are essential to the E-C coupling at the level of single ventricular myocytes isolated from neonatal rat heart, using the whole-cell voltage-clamp technique. Only diclofenac of three NSAIDs, including naproxen and ibuprofen, significantly reduced inward whole cell currents. At concentrations higher than 3 microM, diclofenac inhibited reversibly the Na(+) current and did irreversibly the L-type Ca(2+) channels-mediated inward current (IC(50)=12.89+/-0.43 microM) in a dose-dependent manner. However, nifedipine, a well-known L-type channel blocker, effectively inhibited the L-type Ca(2+) currents but not the Na(+) current. Our finding may explain that diclofenac causes the CV risk by the inhibition of L-type Ca(2+) channel, leading to the impairment of E-C coupling in cardiac myocytes.
Collapse
Affiliation(s)
- Oleg V Yarishkin
- Department of Physiology, Institute of Health Sciences, and Medical Research Center for Neural Dysfunction, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhang P, Shi J, Shen B, Li X, Gao Y, Zhu Z, Zhu Z, Ji Y, Teng M, Niu L. Stejnihagin, a novel snake metalloproteinase from Trimeresurus stejnegeri venom, inhibited L-type Ca2+ channels. Toxicon 2008; 53:309-15. [PMID: 19114053 DOI: 10.1016/j.toxicon.2008.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2008] [Revised: 10/18/2008] [Accepted: 12/01/2008] [Indexed: 11/18/2022]
Abstract
Snake venom metalloproteinases (SVMPs) mainly distribute in Crotalid and Viperid snake venom and are classified into the Reprolysin subfamily of the M12 family of metalloproteinases. Previous function investigations have suggested that SVMPs are the key toxins involved in a variety of snake venom-induced pathogenesis including systemic injury, local damage, hemorrhage, edema, hypotension, hypovolemia, inflammation and necrosis. However, up to now, there is no report on ion channels blocking activity about SVMPs. Here, from Trimeresurus stejnegeri venom we purified a component Stejnihagin containing a mixture of Stejnihagin-A and -B, with 86% sequences identity, both as members of SVMPs. In the study, whole-cell patch clamp and vessel tension measurement were employed to identify the effect of Stejnihagin on L-type Ca2+ channels and vessel contraction. The results show that Stejnihagin inhibited L-type Ca2+ channels in A7r5 cells with an IC50 about 37 nM and simultaneously blocked 60 mM K+-induced vessel contraction. Besides, the inhibitory effect of Stejnihagin on L-type Ca2+ channels was also independent of the enzymatic activity. This finding offers new insight into the snake venom metalloproteinase functions and provides a novel pathogenesis of T. stejnegeri venom. Furthermore, it may also provide a clue to study the structure-function relationship of animal toxins and voltage-gated Ca2+ channel.
Collapse
Affiliation(s)
- Ping Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Higley MJ, Sabatini BL. Calcium Signaling in Dendrites and Spines: Practical and Functional Considerations. Neuron 2008; 59:902-13. [DOI: 10.1016/j.neuron.2008.08.020] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 08/18/2008] [Accepted: 08/19/2008] [Indexed: 11/25/2022]
|
38
|
Kubista H, Mafra RA, Chong Y, Nicholson GM, Beirão PSL, Cruz JS, Boehm S, Nentwig W, Kuhn-Nentwig L. CSTX-1, a toxin from the venom of the hunting spider Cupiennius salei, is a selective blocker of L-type calcium channels in mammalian neurons. Neuropharmacology 2007; 52:1650-62. [PMID: 17517422 DOI: 10.1016/j.neuropharm.2007.03.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 02/28/2007] [Accepted: 03/21/2007] [Indexed: 12/01/2022]
Abstract
The inhibitor cystine-knot motif identified in the structure of CSTX-1 from Cupiennius salei venom suggests that this toxin may act as a blocker of ion channels. Whole-cell patch-clamp experiments performed on cockroach neurons revealed that CSTX-1 produced a slow voltage-independent block of both mid/low- (M-LVA) and high-voltage-activated (HVA) insect Ca(v) channels. Since C. salei venom affects both insect as well as rodent species, we investigated whether Ca(v) channel currents of rat neurons are also inhibited by CSTX-1. CSTX-1 blocked rat neuronal L-type, but no other types of HVA Ca(v) channels, and failed to modulate LVA Ca(v) channel currents. Using neuroendocrine GH3 and GH4 cells, CSTX-1 produced a rapid voltage-independent block of L-type Ca(v) channel currents. The concentration-response curve was biphasic in GH4 neurons and the subnanomolar IC(50) values were at least 1000-fold lower than in GH3 cells. L-type Ca(v) channel currents of skeletal muscle myoballs and other voltage-gated ion currents of rat neurons, such as I(Na(v)) or I(K(v)) were not affected by CSTX-1. The high potency and selectivity of CSTX-1 for a subset of L-type channels in mammalian neurons may enable the toxin to be used as a molecular tool for the investigation of this family of Ca(v) channels.
Collapse
Affiliation(s)
- Helmut Kubista
- Center for Biomolecular Medicine and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringerstrasse 13a, A-1090 Vienna, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Doering CJ, McRory JE. Effects of extracellular pH on neuronal calcium channel activation. Neuroscience 2007; 146:1032-43. [PMID: 17434266 DOI: 10.1016/j.neuroscience.2007.02.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 02/23/2007] [Accepted: 02/28/2007] [Indexed: 11/16/2022]
Abstract
Previous studies have shown that extracellular pH (pHo) alters gating and permeation properties of cardiac L- and T-type channels. However, a comprehensive study investigating the effects of pHo on all other voltage-gated calcium channels is lacking. Here, we report the effects of pHo on activation parameters slope factor (S), half-activation potential (Va), reversal potential (Erev), and maximum slope conductance (Gmax) of the nine known neuronal voltage-gated calcium channels transiently expressed in tsA-201 cells. In all cases, acidification of the extracellular bathing solution results in a depolarizing shift in the activation curve and reduction in peak current amplitudes. Relative to a physiological pHo of 7.25, statistically significant depolarizing shifts in Va were observed for all channels at pHo 7.00 except Cav1.3 and 3.2, which showed significant shifts at pHo 6.75 and below. All channels displayed significant reductions in Gmax relative to pHo 7.25 at pHo 7.00 except Cav1.2, 2.1, and 3.1 which required acidification to pHo 6.75. Upon acidification Cav3 channels displayed the largest changes in Vas and exhibited the largest reduction in Gmax compared with other channel subtypes. Taken together, these results suggest that significant modulation of calcium channel currents can occur with changes in pHo. Acidification of the external solution did not produce significant shifts in observed Erevs or blockade of outward currents for any of the nine channel subtypes. Finally, we tested a simple Woodhull-type model of current block by assuming blockade of the pore by a single proton. In all cases, the amount of blockade observed could not be explained in these simple terms, suggesting that proton modulation is more complicated, involving more than one site or gating modification as has been previously described for cardiac L- and T-type channels.
Collapse
Affiliation(s)
- C J Doering
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | | |
Collapse
|
40
|
Felix R. Calcium channelopathies. Neuromolecular Med 2007; 8:307-18. [PMID: 16775382 DOI: 10.1385/nmm:8:3:307] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2005] [Revised: 11/30/1999] [Accepted: 01/20/2006] [Indexed: 11/11/2022]
Abstract
Intracellular calcium ([Ca2+]i) is highly regulated in eukaryotic cells. The free [Ca2+]i is approximately four orders of magnitude less than that in the extracellular environment. It is, therefore, an electrochemical gradient favoring Ca2+ entry, and transient cellular activation increasing Ca2+ permeability will lead to a transient increase in [Ca2+]i. These transient rises of [Ca2+]i trigger or regulate diverse intracellular events, including metabolic processes, muscle contraction, secretion of hormones and neurotransmitters, cell differentiation, and gene expression. Hence, changes in [Ca2+]i act as a second messenger system coordinating modifications in the external environment with intracellular processes. Notably, information on the molecular genetics of the membrane channels responsible for the influx of Ca2+ ions has led to the discovery that mutations in these proteins are linked to human disease. Ca2+ channel dysfunction is now known to be the basis for several neurological and muscle disorders such as migraine, ataxia, and periodic paralysis. In contrast to other types of genetic diseases, Ca2+ channelopathies can be studied with precision by electrophysiological methods, and in some cases, the results have been highly rewarding with a biophysical phenotype that correlates with the ultimate clinical phenotype. This review outlines recent advances in genetic, molecular, and pathophysiological aspects of human Ca2+ channelopathies.
Collapse
Affiliation(s)
- Ricardo Felix
- Department of Cell Biology, Center for Research and Advanced Studies, National Polytechnic Institute (Cinvestav-IPN), Mexico City, Mexico.
| |
Collapse
|
41
|
King GF. Modulation of insect Cav channels by peptidic spider toxins. Toxicon 2007; 49:513-30. [PMID: 17197008 DOI: 10.1016/j.toxicon.2006.11.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Accepted: 11/17/2006] [Indexed: 10/23/2022]
Abstract
Insects have a much smaller repertoire of voltage-gated calcium (Ca(V)) channels than vertebrates. Drosophila melanogaster harbors only a single ortholog of each of the vertebrate Ca(V)1, Ca(V)2, and Ca(V)3 subtypes, although its basal inventory is expanded by alternative splicing and editing of Ca(V) channel transcripts. Nevertheless, there appears to be little functional plasticity within this limited panel of insect Ca(V) channels, since severe loss-of-function mutations in genes encoding the pore-forming alpha1 subunits in Drosophila are embryonic lethal. Since the primary role of spider venom is to paralyze or kill insect prey, it is not surprising that most, if not all, spider venoms contain peptides that potently modify the activity of these functionally critical insect Ca(V) channels. Unfortunately, it has proven difficult to determine the precise ion channel subtypes recognized by these peptide toxins since insect Ca(V) channels have significantly different pharmacology to their vertebrate counterparts, and cloned insect Ca(V) channels are not available for electrophysiological studies. However, biochemical and genetic studies indicate that some of these spider toxins might ultimately become the defining pharmacology for certain subtypes of insect Ca(V) channels. This review focuses on peptidic spider toxins that specifically target insect Ca(V) channels. In addition to providing novel molecular tools for ion channel characterization, some of these toxins are being used as leads to develop new methods for controlling insect pests.
Collapse
Affiliation(s)
- Glenn F King
- Division of Chemical and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane Qld. 4072, Australia.
| |
Collapse
|
42
|
Galli GLJ, Taylor EW, Shiels HA. Calcium flux in turtle ventricular myocytes. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1781-9. [PMID: 16887918 DOI: 10.1152/ajpregu.00421.2006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The relative contribution of the sarcoplasmic reticulum (SR), the L-type Ca2+channel and the Na+/Ca2+exchanger (NCX) were assessed in turtle ventricular myocytes using epifluorescent microscopy and electrophysiology. Confocal microscopy images of turtle myocytes revealed spindle-shaped cells, which lacked T-tubules and had a large surface area-to-volume ratio. Myocytes loaded with the fluorescent Ca2+-sensitive dye Fura-2 elicited Ca2+transients, which were insensitive to ryanodine and thapsigargin, indicating the SR plays a small role in the regulation of contraction and relaxation in the turtle ventricle. Sarcolemmal Ca2+currents were measured using the perforated-patch voltage-clamp technique. Depolarizing voltage steps to 0 mV elicited an inward current that could be blocked by nifedipine, indicating the presence of Ca2+currents originating from L-type Ca2+channels (ICa). The density of ICawas 3.2 ± 0.5 pA/pF, which led to an overall total Ca2+influx of 64.1 ± 9.3 μM/l. NCX activity was measured as the Ni+-sensitive current at two concentrations of intracellular Na+(7 and 14 mM). Total Ca2+influx through the NCX during depolarizing voltage steps to 0 mV was 58.5 ± 7.7 μmol/l and 26.7 ± 3.2 μmol/l at 14 and 7 mM intracellular Na+, respectively. In the absence of the SR and L-type Ca2+channels, the NCX is able to support myocyte contraction independently. Our results indicate turtle ventricular myocytes are primed for sarcolemmal Ca2+transport, and most of the Ca2+used for contraction originates from the L-type Ca2+channel.
Collapse
Affiliation(s)
- Gina L J Galli
- Faculty of Life Sciemces, The University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT UK.
| | | | | |
Collapse
|
43
|
Yoshihara S, Morimoto H, Ohori M, Yamada Y, Abe T, Arisaka O. Cannabinoid receptor agonists inhibit Ca(2+) influx to synaptosomes from rat brain. Pharmacology 2006; 76:157-62. [PMID: 16446560 DOI: 10.1159/000091228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Accepted: 11/14/2005] [Indexed: 11/19/2022]
Abstract
We examined the effects of cannabinoid receptor agonists on (45)Ca(2+) uptake in rat brain synaptosomes. A cannabinoid receptor agonist, (R)-(+)-[2,3-dihydro-5-methyl-3-[(4-merpholino)methyl]pyrrolo-[1,2,3-de]-1,4-benzoxazin-6-yl](1-naphthyl)methanone (WIN 55212-2) dose-dependently inhibited (45)Ca(2+) uptake in rat synaptosomes. Only an endogenous cannabinoid receptor agonist, anandamide, dose-dependently inhibited (45)Ca(2+) uptake in rat synaptosomes, but not an endogenous cannabinoid receptor agonist, palmitoylethanolamide. Only a cannabinoid CB1 antagonist, [N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamidehydrochloride] (SR 141716A), reversed the inhibitory effect of these WIN 55212-2 and anandamide on (45)Ca(2+) uptake in rat synaptosomes, but not a cannabinoid CB2 receptor antagonist, [N-[(1S)-endo-1,3,3-trimethylbicyclo[2.2.1]heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)pyrazole-3-carboxamide] (SR 144528). The inhibitory effects of WIN 55212-2 and anandamide on (45)Ca(2+) uptake in rat synaptosomes were reversed by the pretreatment of a voltage-sensitive A-type K(+) channel blocker, dendrotoxin, but no other type of K(+) channel blockers, i.e. iberiotoxin, charybdotoxin or glibenclamide. These findings suggest that cannabinoid receptors inhibit Ca(2+) influx into rat brain nerves via the activation of CB1 receptors and the opening of voltage-sensitive A-type K(+) channels.
Collapse
Affiliation(s)
- Shigemi Yoshihara
- Department of Pediatrics, Dokkyo University School of Medicine, Mibu-machi, Japan.
| | | | | | | | | | | |
Collapse
|
44
|
Etheredge JA, Murchison D, Abbott LC, Griffith WH. Functional compensation by other voltage-gated Ca2+ channels in mouse basal forebrain neurons with Ca(V)2.1 mutations. Brain Res 2005; 1140:105-19. [PMID: 16364258 DOI: 10.1016/j.brainres.2005.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2005] [Revised: 11/10/2005] [Accepted: 11/14/2005] [Indexed: 01/19/2023]
Abstract
Tottering (tg/tg) and leaner (tg(la)/tg(la)) mutant mice exhibit distinct mutations in the gene encoding the voltage-activated Ca(2+) channel alpha(1A) subunit (CACNA1A), the pore-forming subunit of the Ca(V)2.1 (P/Q type) Ca(2+) channels. These mice exhibit absence seizures and deficiencies in motor control and other functions. Previous work in cerebellar Purkinje neurons has shown that these mutations cause dramatic reductions in calcium channel function. Because Purkinje cell somata primarily express the Ca(V)2.1 channels, the general decrease in Ca(V)2.1 channel function is observed as a profound decrease in whole-cell current. In contrast to Purkinje cells, basal forebrain (BF) neurons express all of the Ca(2+) channel alpha(1) subunits, with Ca(V)2.1 contributing approximately 30% to the whole-cell current in wild-type (+/+) mice. Here, we show that whole-cell Ba(2+) current densities in BF neurons are not reduced in the mutant genotypes despite a reduction in the Ca(V)2.1 contribution. By blocking the different Ca(2+) channel subtypes with specific pharmacological agents, we found a significant increase in the proportion of Ca(V)1 Ca(2+) current in mutant phenotypes. There was no change in tissue mRNA expression of calcium channel subtypes Ca(V)2.1, Ca(V)2.2, Ca(V)1.2, Ca(V)1.3, and Ca(V)2.3 in the tottering and leaner mutant mice. These results suggest that Ca(V)1 channels may functionally upregulate to compensate for reduced Ca(V)2.1 function in the mutants without an increase in Ca(v)1 message. Single-cell reverse transcription polymerase chain reaction (RT-PCR) experiments in a subset of sampled neurons revealed that approximately 90% of the cells could be considered cholinergic based on choline acetyltransferase (ChAT) mRNA expression.
Collapse
Affiliation(s)
- Jason A Etheredge
- Department of Medical Pharmacology and Toxicology, College of Medicine, Reynolds Medical Science Building, Texas A&M University System Health Science Center, 1114-TAMU, College Station, TX 77843-1114, USA
| | | | | | | |
Collapse
|
45
|
Abstract
Voltage-gated Ca2+ (Ca(v)) channels are found in all excitable cells and many nonexcitable cells, in which they govern Ca2+ influx, thereby contributing to determine a host of important physiological processes including gene transcription, muscle contraction, hormone secretion, and neurotransmitter release. The past years have seen some significant advances in our understanding of the functional, pharmacological, and molecular properties of Ca(v) channels. Molecular studies have revealed that several of these channels are oligomeric complexes consisting of an ion-conducting alpha1 subunit and auxiliary alpha2delta, beta, and gamma subunits. In addition, cloning of multiple Ca(v) channel alpha1 subunits has offered the opportunity to investigate the regulation of these proteins at the molecular level. The regulation of Ca(v) channels by intracellular second messengers constitutes a key mechanism for controlling Ca2+ influx. This review summarizes recent advances that have provided important clues to the underlying molecular mechanisms involved in the regulation of Ca(v) channels by protein phosphorylation, G-protein activation, and interactions with Ca(2+)-binding and SNARE proteins.
Collapse
Affiliation(s)
- Ricardo Felix
- Department of Physiology Biophysics, and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico.
| |
Collapse
|
46
|
French RJ, Zamponi GW. Voltage-gated sodium and calcium channels in nerve, muscle, and heart. IEEE Trans Nanobioscience 2005; 4:58-69. [PMID: 15816172 DOI: 10.1109/tnb.2004.842500] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Voltage-gated ion channels are membrane proteins which underlie rapid electrical signals among neurons and the spread of excitation in skeletal muscle and heart. We outline some recent advances in the study of voltage-sensitive sodium and calcium channels. Investigations are providing insight into the changes in molecular conformation associated with open-closed gating of the channels, the mechanisms by which they allow only specific ion species to pass through and carry an electric current, and the pathological consequences of small perturbations in channel structure which result from genetic mutations. Determination of three-dimensional structures, coupled with molecular manipulations by site-directed mutagenesis, and parallel electrophysiological analyses of currents through the ion channels, are providing an understanding of the roles and function of these channels at an unprecedented level of molecular detail. Crucial to these advances are studies of bacterial homologues of ion channels from man and other eukaryotes, and the use of naturally occurring peptide toxins which target different ion channel types with exquisite specificity.
Collapse
Affiliation(s)
- Robert J French
- Department of Physiology and Biophysics, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | | |
Collapse
|
47
|
Wood CD, Nishigaki T, Furuta T, Baba SA, Darszon A. Real-time analysis of the role of Ca(2+) in flagellar movement and motility in single sea urchin sperm. J Cell Biol 2005; 169:725-31. [PMID: 15928204 PMCID: PMC2171626 DOI: 10.1083/jcb.200411001] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Accepted: 04/29/2005] [Indexed: 11/22/2022] Open
Abstract
Eggs of many marine and mammalian species attract sperm by releasing chemoattractants that modify the bending properties of flagella to redirect sperm paths toward the egg. This process, called chemotaxis, is dependent on extracellular Ca(2+). We used stroboscopic fluorescence imaging to measure intracellular Ca(2+) concentration ([Ca(2+)]i) in the flagella of swimming sea urchin sperm. Uncaging of cyclic GMP induced Ca(2+) entry via at least two distinct pathways, and we identified a nimodipine-sensitive pathway, compartmentalized in the flagella, as a key regulator of flagellar bending and directed motility changes. We found that, contrary to current models, the degree of flagellar bending does not vary in proportion to the overall [Ca(2+)]i. Instead we propose a new model whereby flagella bending is increased by Ca(2+) flux through the nimodipine-sensitive pathway, and is unaffected by [Ca(2+)]i increases through alternative pathways.
Collapse
Affiliation(s)
- Christopher D Wood
- Department of Developmental Genetics and Molecular Physiology, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos 62210, Mexico.
| | | | | | | | | |
Collapse
|
48
|
Vieira LB, Kushmerick C, Hildebrand ME, Garcia E, Stea A, Cordeiro MN, Richardson M, Gomez MV, Snutch TP. Inhibition of High Voltage-Activated Calcium Channels by Spider Toxin PnTx3-6. J Pharmacol Exp Ther 2005; 314:1370-7. [PMID: 15933156 DOI: 10.1124/jpet.105.087023] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Animal peptide toxins have become powerful tools to study structure-function relationships and physiological roles of voltage-activated Ca(2+) channels. In the present study, we investigated the effects of PnTx3-6, a neurotoxin purified from the venom of the spider Phoneutria nigriventer on cloned mammalian Ca(2+) channels expressed in human embryonic kidney 293 cells and endogenous Ca(2+) channels in N18 neuroblastoma cells. Whole-cell patch-clamp measurements indicate that PnTx3-6 reversibly inhibited L-(alpha(1C)/Ca(v)1.2), N-(alpha(1B)/Ca(v)2.2), P/Q-(alpha(1A)/Ca(v)2.1), and R-(alpha(1E)/Ca(v)2.3) type channels with varying potency (alpha(1B) > alpha(1E) > alpha(1A) > alpha(1C)) and IC(50) values of 122, 136, 263, and 607 nM, respectively. Inhibition occurred without alteration of the kinetics or the voltage dependence of the exogenously expressed Ca(2+) channels. In N18 cells, PnTx3-6 exhibited highest potency against N-type (conotoxin-GVIA-sensitive) current. In contrast to its effects on high voltage-activated Ca(2+) channels subtypes, application of 1 microM PnTx3-6 did not affect alpha(1G)/Ca(v)3.1 T-type Ca(2+) channels. Based on our study, we suggest that PnTx3-6 acts as a omega-toxin that targets high voltage-activated Ca(2+) channels, with a preference for the Ca(v)2 subfamily (N-, P/Q-, and R-types).
Collapse
Affiliation(s)
- Luciene B Vieira
- Biotechnology Laboratory, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schönherr R. Clinical Relevance of Ion Channels for Diagnosis and Therapy of Cancer. J Membr Biol 2005; 205:175-84. [PMID: 16362505 DOI: 10.1007/s00232-005-0782-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Indexed: 12/29/2022]
Abstract
Ion channels have a critical role in cell proliferation and it is well documented that channel blockers can inhibit the growth of cancer cells. The concept of ion channels as therapeutic targets or prognostic biomarkers attracts increasing interest, but the lack of potent and selective channel modulators has hampered a critical verification for many years. Today, the knowledge of human ion channel genes is almost complete and molecular correlates for many native currents have already been identified. This information triggered a wave of experimental results, identifying individual ion channels with relevance for specific cancer types. The current pattern of cancer-related ion channels is not arbitrary, but can be reduced to few members from each ion channel family. This review aims to provide an overview of the molecularly identified ion channels that might be relevant for the most common human cancer types. Possible applications of these candidates for a targeted cancer therapy or for clinical diagnosis are discussed.
Collapse
Affiliation(s)
- R Schönherr
- Research Unit Molecular and Cellular Biophysics, Medical Faculty of the Friedrich Schiller University Jena, Drackendorfer St. 1, Jena, D-07747, Germany.
| |
Collapse
|
50
|
Ouardouz M, Zamponi GW, Barr W, Kiedrowski L, Stys PK. Protection of ischemic rat spinal cord white matter: Dual action of KB-R7943 on Na+/Ca2+ exchange and L-type Ca2+ channels. Neuropharmacology 2005; 48:566-75. [PMID: 15755484 DOI: 10.1016/j.neuropharm.2004.12.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2004] [Revised: 12/10/2004] [Accepted: 12/15/2004] [Indexed: 10/25/2022]
Abstract
The effect of the Na+/Ca(2+)-exchange inhibitor KB-R7943 was investigated in spinal cord dorsal column ischemia in vitro. Oxygen/glucose deprivation at 37 degrees C for 1 h causes severe injury even in the absence of external Ca2+. KB-R7943 was very protective in the presence and absence of external Ca2+ implicating mechanisms in addition to extracellular Ca2+ influx through Na+/Ca(2+)-exchange, such as activation of ryanodine receptors by L-type Ca2+ channels. Indeed, blockade of L-type Ca2+ by nimodipine confers a certain degree of protection of dorsal column against ischemia; combined application of nimodipine and KB-R7943 was not additive suggesting that KB-R7943 may also act on Ca2+ channels. KB-R7943 reduced inward Ba2+ current with IC50 = 7 microM in tsA-201 cells expressing Ca(v)1.2. Moreover, nifedipine and KB-R7943 both reduced depolarization-induced [Ca2+]i increases in forebrain neurons and effects were not additive. Nimodipine or KB-R7943 also reduced ischemic axoplasmic Ca2+ increase, which persisted in 0Ca2+/EGTA perfusate in dorsal column during ischemia. While KB-R7943 cannot be considered to be a specific Na+/Ca2+ exchange inhibitor, its profile makes it a very useful neuroprotectant in dorsal columns by: reducing Ca2+ import through reverse Na+/Ca2+ exchange; reducing influx through L-type Ca2+ channels, and indirectly inhibiting Ca2+ release from the ER through activation of ryanodine receptors.
Collapse
Affiliation(s)
- M Ouardouz
- Ottawa Health Research Institute, Division of Neuroscience, University of Ottawa, Ottawa, ON, Canada K1Y 4K9
| | | | | | | | | |
Collapse
|