151
|
Lieb A, Ortner N, Striessnig J. C-terminal modulatory domain controls coupling of voltage-sensing to pore opening in Cav1.3 L-type Ca(2+) channels. Biophys J 2014; 106:1467-75. [PMID: 24703308 PMCID: PMC3976517 DOI: 10.1016/j.bpj.2014.02.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/11/2014] [Accepted: 02/25/2014] [Indexed: 12/31/2022] Open
Abstract
Activity of voltage-gated Cav1.3 L-type Ca2+ channels is required for proper hearing as well as sinoatrial node and brain function. This critically depends on their negative activation voltage range, which is further fine-tuned by alternative splicing. Shorter variants miss a C-terminal regulatory domain (CTM), which allows them to activate at even more negative potentials than C-terminally long-splice variants. It is at present unclear whether this is due to an increased voltage sensitivity of the Cav1.3 voltage-sensing domain, or an enhanced coupling of voltage-sensor conformational changes to the subsequent opening of the activation gate. We studied the voltage-dependence of voltage-sensor charge movement (QON-V) and of current activation (ICa-V) of the long (Cav1.3L) and a short Cav1.3 splice variant (Cav1.342A) expressed in tsA-201 cells using whole cell patch-clamp. Charge movement (QON) of Cav1.3L displayed a much steeper voltage-dependence and a more negative half-maximal activation voltage than Cav1.2 and Cav3.1. However, a significantly higher fraction of the total charge had to move for activation of Cav1.3 half-maximal conductance (Cav1.3: 68%; Cav1.2: 52%; Cav3.1: 22%). This indicated a weaker coupling of Cav1.3 voltage-sensor charge movement to pore opening. However, the coupling efficiency was strengthened in the absence of the CTM in Cav1.342A, thereby shifting ICa-V by 7.2 mV to potentials that were more negative without changing QON-V. We independently show that the presence of intracellular organic cations (such as n-methyl-D-glucamine) induces a pronounced negative shift of QON-V and a more negative activation of ICa-V of all three channels. These findings illustrate that the voltage sensors of Cav1.3 channels respond more sensitively to depolarization than those of Cav1.2 or Cav3.1. Weak coupling of voltage sensing to pore opening is enhanced in the absence of the CTM, allowing short Cav1.342A splice variants to activate at lower voltages without affecting QON-V.
Collapse
Affiliation(s)
- Andreas Lieb
- Pharmacology and Toxicology, Institute of Pharmacy, and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.
| | - Nadine Ortner
- Pharmacology and Toxicology, Institute of Pharmacy, and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Jörg Striessnig
- Pharmacology and Toxicology, Institute of Pharmacy, and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
152
|
Shoenfeld L, Westenbroek RE, Fisher E, Quinlan KA, Tysseling VM, Powers RK, Heckman CJ, Binder MD. Soma size and Cav1.3 channel expression in vulnerable and resistant motoneuron populations of the SOD1G93A mouse model of ALS. Physiol Rep 2014; 2:2/8/e12113. [PMID: 25107988 PMCID: PMC4246589 DOI: 10.14814/phy2.12113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although the loss of motoneurons is an undisputed feature of amyotrophic lateral sclerosis (ALS) in man and in its animal models (SOD1 mutant mice), how the disease affects the size and excitability of motoneurons prior to their degeneration is not well understood. This study was designed to test the hypothesis that motoneurons in mutant SOD1G93A mice exhibit an enlargement of soma size (i.e., cross‐sectional area) and an increase in Cav1.3 channel expression at postnatal day 30, well before the manifestation of physiological symptoms that typically occur at p90 (Chiu et al. 1995). We made measurements of spinal and hypoglossal motoneurons vulnerable to degeneration, as well as motoneurons in the oculomotor nucleus that are resistant to degeneration. Overall, we found that the somata of motoneurons in male SOD1G93A mutants were larger than those in wild‐type transgenic males. When females were included in the two groups, significance was lost. Expression levels of the Cav1.3 channels were not differentiated by genotype, sex, or any interaction of the two. These results raise the intriguing possibility of an interaction between male sex steroid hormones and the SOD1 mutation in the etiopathogenesis of ALS. This study was designed to test the hypothesis that motoneurons in mutant SOD1G93A mice exhibit an enlargement of soma size (i.e., cross‐sectional area) and an increase in Cav1.3 channel expression at postnatal day 30, well before the manifestation of physiological symptoms that typically occur at p90 (Chiu et al. 1995). We made measurements of spinal and hypoglossal motoneurons vulnerable to degeneration, as well as motoneurons in the oculomotor nucleus that are resistant to degeneration. Overall, we found that the somata of motoneurons in male SOD1G93A mutants were larger than those in wild‐type transgenic males. When females were included in the two groups, significance was lost. These results raise the intriguing possibility of an interaction between male sex steroid hormones and the SOD1 mutation in the etiopathogenesis of ALS.
Collapse
Affiliation(s)
- Liza Shoenfeld
- Graduate Program in Neurobiology & Behavior, University of Washington, Seattle, Washington, USA
| | - Ruth E Westenbroek
- Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Erika Fisher
- Department of Physiology & Biophysics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Katharina A Quinlan
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Vicki M Tysseling
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA Department of Physical Therapy and Human Movement Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Randall K Powers
- Department of Physiology & Biophysics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Charles J Heckman
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA Department of Physical Therapy and Human Movement Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Marc D Binder
- Graduate Program in Neurobiology & Behavior, University of Washington, Seattle, Washington, USA Department of Physiology & Biophysics, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
153
|
Pan B, Guo Y, Kwok WM, Hogan Q, Wu HE. Sigma-1 receptor antagonism restores injury-induced decrease of voltage-gated Ca2+ current in sensory neurons. J Pharmacol Exp Ther 2014; 350:290-300. [PMID: 24891452 PMCID: PMC4109486 DOI: 10.1124/jpet.114.214320] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/29/2014] [Indexed: 01/01/2023] Open
Abstract
Sigma-1 receptor (σ1R), an endoplasmic reticulum-chaperone protein, can modulate painful response after peripheral nerve injury. We have demonstrated that voltage-gated calcium current is inhibited in axotomized sensory neurons. We examined whether σ1R contributes to the sensory dysfunction of voltage-gated calcium channel (VGCC) after peripheral nerve injury through electrophysiological approach in dissociated rat dorsal root ganglion (DRG) neurons. Animals received either skin incision (Control) or spinal nerve ligation (SNL). Both σ1R agonists, (+)pentazocine (PTZ) and DTG [1,3-di-(2-tolyl)guanidine], dose dependently inhibited calcium current (ICa) with Ba(2+) as charge carrier in control sensory neurons. The inhibitory effect of σ1R agonists on ICa was blocked by σ1R antagonist, BD1063 (1-[2-(3,4-dichlorophenyl)ethyl]-4-methylpiperazine dihydrochloride) or BD1047 (N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide). PTZ and DTG showed similar effect on ICa in axotomized fifth DRG neurons (SNL L5). Both PTZ and DTG shifted the voltage-dependent activation and steady-state inactivation of VGCC to the left and accelerated VGCC inactivation rate in both Control and axotomized L5 SNL DRG neurons. The σ1R antagonist, BD1063 (10 μM), increases ICa in SNL L5 neurons but had no effect on Control and noninjured fourth lumbar neurons in SNL rats. Together, the findings suggest that activation of σR1 decreases ICa in sensory neurons and may play a pivotal role in pain generation.
Collapse
Affiliation(s)
- Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin (B.P., Y.G., W.-M.K., Q.H., H.-e.W.); and Department of Anesthesiology, Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin (Q.H.)
| | - Yuan Guo
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin (B.P., Y.G., W.-M.K., Q.H., H.-e.W.); and Department of Anesthesiology, Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin (Q.H.)
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin (B.P., Y.G., W.-M.K., Q.H., H.-e.W.); and Department of Anesthesiology, Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin (Q.H.)
| | - Quinn Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin (B.P., Y.G., W.-M.K., Q.H., H.-e.W.); and Department of Anesthesiology, Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin (Q.H.)
| | - Hsiang-en Wu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin (B.P., Y.G., W.-M.K., Q.H., H.-e.W.); and Department of Anesthesiology, Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin (Q.H.)
| |
Collapse
|
154
|
Huang H, Ng CY, Yu D, Zhai J, Lam Y, Soong TW. Modest CaV1.342-selective inhibition by compound 8 is β-subunit dependent. Nat Commun 2014; 5:4481. [PMID: 25057870 PMCID: PMC4124865 DOI: 10.1038/ncomms5481] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/20/2014] [Indexed: 01/20/2023] Open
Abstract
Two voltage-gated calcium channel subtypes—CaV1.2 and CaV1.3—underlie the major L-type Ca2+ currents in the mammalian central nervous system. Owing to their high sequence homology, the two channel subtypes share similar pharmacological properties, and at high doses classic calcium channel blockers, such as dihydropyridines, phenylalkylamines and benzothiazepines, do not discriminate between the two channel subtypes. Recent progress in treating Parkinson’s disease (PD) was marked by the discovery of synthetic compound 8, which was reported to be a highly selective inhibitor of the CaV1.3 L-type calcium channels (LTCC). However, despite a previously reported IC50 of ~24 μM, in our hands inhibition of the full-length CaV1.342 by compound 8 at 50 μM reaches a maximum of 45%. Moreover, we find that the selectivity of compound 8 towards CaV1.3 relative to CaV1.2B15 channels is greatly influenced by the β-subunit type and its splice isoform variants. Compound 8-1-(3-chlorophenethyl)-3-cyclopentylpyrimidine-2,4,6(1H,3H,5H)-trione was previously reported to be a selective inhibitor of the CaV1.3 calcium channel. Now, Huang et al. demonstrate that selectivity towards CaV1.3 relative to CaV1.2 is dependent on the type of β-subunit and CaV1.3 splice variant assayed.
Collapse
Affiliation(s)
- Hua Huang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Cheng Yang Ng
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Dejie Yu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jing Zhai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yulin Lam
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Tuck Wah Soong
- 1] Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore [2] NUS Graduate School for Integrative Sciences and Engineering, Singapore 117456, Singapore [3] Neurobiology/Ageing Programme, National University of Singapore, Singapore 117456, Singapore [4] National Neuroscience Institute, Jalan Tan Tock Seng, Singapore 308433, Singapore
| |
Collapse
|
155
|
The role of L-type voltage-gated calcium channels Cav1.2 and Cav1.3 in normal and pathological brain function. Cell Tissue Res 2014; 357:463-76. [DOI: 10.1007/s00441-014-1936-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
|
156
|
Pyrimidine-2,4,6-triones are a new class of voltage-gated L-type Ca2+ channel activators. Nat Commun 2014; 5:3897. [PMID: 24941892 PMCID: PMC4083433 DOI: 10.1038/ncomms4897] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/15/2014] [Indexed: 12/15/2022] Open
Abstract
Cav1.2 and Cav1.3 are the main L-type Ca2+ channel subtypes in the brain. Cav1.3 channels have recently been implicated in the pathogenesis of Parkinson’s disease. Therefore, Cav1.3-selective blockers are developed as promising neuroprotective drugs. We studied the pharmacological properties of a pyrimidine-2,4,6-trione derivative (1-(3-chlorophenethyl)-3-cyclopentylpyrimidine-2,4,6-(1H,3H,5H)-trione, Cp8) recently reported as the first highly selective Cav1.3 blocker. Here we show, in contrast to this previous study, that Cp8 reproducibly increases inward Ca2+ currents of Cav1.3 and Cav1.2 channels expressed in tsA-201 cells by slowing activation, inactivation and enhancement of tail currents. Similar effects are also observed for native Cav1.3 and Cav1.2 channels in mouse chromaffin cells, while non-L-type currents are unaffected. Evidence for a weak and non-selective inhibition of Cav1.3 and Cav1.2 currents is only observed in a minority of cells using Ba2+ as charge carrier. Therefore, our data identify pyrimidine-2,4,6-triones as Ca2+ channel activators. Selective inhibitors of the L-type Ca2+ channel Cav1.3 are being developed as neuroprotective drugs. Here, Ortner et al. assess the pharmacological properties of a recently reported, selective Cav1.3 blocker and show that this agent is a Ca2+ channel activator.
Collapse
|
157
|
Simms BA, Zamponi GW. Neuronal voltage-gated calcium channels: structure, function, and dysfunction. Neuron 2014; 82:24-45. [PMID: 24698266 DOI: 10.1016/j.neuron.2014.03.016] [Citation(s) in RCA: 420] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Voltage-gated calcium channels are the primary mediators of depolarization-induced calcium entry into neurons. There is great diversity of calcium channel subtypes due to multiple genes that encode calcium channel α1 subunits, coassembly with a variety of ancillary calcium channel subunits, and alternative splicing. This allows these channels to fulfill highly specialized roles in specific neuronal subtypes and at particular subcellular loci. While calcium channels are of critical importance to brain function, their inappropriate expression or dysfunction gives rise to a variety of neurological disorders, including, pain, epilepsy, migraine, and ataxia. This Review discusses salient aspects of voltage-gated calcium channel function, physiology, and pathophysiology.
Collapse
Affiliation(s)
- Brett A Simms
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
158
|
Liu Y, Harding M, Pittman A, Dore J, Striessnig J, Rajadhyaksha A, Chen X. Cav1.2 and Cav1.3 L-type calcium channels regulate dopaminergic firing activity in the mouse ventral tegmental area. J Neurophysiol 2014; 112:1119-30. [PMID: 24848473 DOI: 10.1152/jn.00757.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dopaminergic projections from the ventral tegmental area (VTA) constitute the mesolimbocortical system that underlies addiction and psychosis primarily as a result of increased dopaminergic transmission. Dopamine release is spike dependent. L-type calcium channels (LTCCs) play an important role in regulating firing activities, but the contribution of specific subtypes remains unclear. This article describes different functions of Cav1.2 and Cav1.3 subtypes in regulating firing properties with two transgenic mouse strains. For basal firing, Cav1.3-deficient (Cav1.3(-/-)) mice had a lower basal firing frequency. The dihydropyridine (DHP) channel blocker nifedipine reduced single-spike firing in mice expressing DHP-insensitive Cav1.2 channels (Cav1.2DHP(-/-) mice), confirming the significant contribution from the Cav1.3 subtype in basal firing. Moreover, the DHP channel activator (S)-(-)-Bay K8644 and the non-DHP channel activator FPL 64176 converted firing patterns from single spiking to bursting in Cav1.2DHP(-/-) mice. Nifedipine inhibited burst firing induced by both activators, suggesting that Cav1.3 also serves an essential role in burst firing. However, FPL 64176 also induced bursting in Cav1.3(-/-) mice. These results indicate that the Cav1.3 subtype is crucial to regulation of basal single-spike firing, while activation of both Cav1.2 and Cav1.3 can support burst firing of VTA neurons.
Collapse
Affiliation(s)
- Yudan Liu
- Department of Physiology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning, China;
| | - Meghan Harding
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Andrea Pittman
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Jules Dore
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria; and
| | - Anjali Rajadhyaksha
- Division of Pediatric Neurology, Department of Pediatrics and Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York
| | - Xihua Chen
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
159
|
Calcium signaling in Parkinson's disease. Cell Tissue Res 2014; 357:439-54. [PMID: 24781149 DOI: 10.1007/s00441-014-1866-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 03/06/2014] [Indexed: 12/16/2022]
Abstract
Calcium (Ca(2+)) is an almost universal second messenger that regulates important activities of all eukaryotic cells. It is of critical importance to neurons, which have developed extensive and intricate pathways to couple the Ca(2+) signal to their biochemical machinery. In particular, Ca(2+) participates in the transmission of the depolarizing signal and contributes to synaptic activity. During aging and in neurodegenerative disease processes, the ability of neurons to maintain an adequate energy level can be compromised, thus impacting on Ca(2+) homeostasis. In Parkinson's disease (PD), many signs of neurodegeneration result from compromised mitochondrial function attributable to specific effects of toxins on the mitochondrial respiratory chain and/or to genetic mutations. Despite these effects being present in almost all cell types, a distinguishing feature of PD is the extreme selectivity of cell loss, which is restricted to the dopaminergic neurons in the ventral portion of the substantia nigra pars compacta. Many hypotheses have been proposed to explain such selectivity, but only recently it has been convincingly shown that the innate autonomous activity of these neurons, which is sustained by their specific Cav1.3 L-type channel pore-forming subunit, is responsible for the generation of basal metabolic stress that, under physiological conditions, is compensated by mitochondrial buffering. However, when mitochondria function becomes even partially compromised (because of aging, exposure to environmental factors or genetic mutations), the metabolic stress overwhelms the protective mechanisms, and the process of neurodegeneration is engaged. The characteristics of Ca(2+) handling in neurons of the substantia nigra pars compacta and the possible involvement of PD-related proteins in the control of Ca(2+) homeostasis will be discussed in this review.
Collapse
|
160
|
Hasreiter J, Goldnagl L, Böhm S, Kubista H. Cav1.2 and Cav1.3 L-type calcium channels operate in a similar voltage range but show different coupling to Ca(2+)-dependent conductances in hippocampal neurons. Am J Physiol Cell Physiol 2014; 306:C1200-13. [PMID: 24760982 DOI: 10.1152/ajpcell.00329.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In the central nervous system, L-type voltage-gated calcium channels (LTCCs) come in two isoforms, namely Cav1.2 and Cav1.3 channels. It has been shown previously that these channels differ in biophysical properties, in subcellular localization, and in the coupling to the gene transcription machinery. In previous work on rat hippocampal neurons we have identified an excitatory cation conductance and an inhibitory potassium conductance as important LTCC coupling partners. Notably, a stimulus-dependent interplay of LTCC-mediated Ca(2+) influx and activation of these Ca(2+)-dependent conductances was found to give rise to characteristic voltage responses. However, the contribution of Cav1.2 and Cav1.3 to these voltage responses remained unknown. Hence, the relative contribution of the LTCC isoforms therein was the focus of the current study on hippocampal neurons derived from genetically modified mice, which either lack a LTCC isoform (Cav1.3 knockout mice) or express a dihydropyridine-insensitive LTCC isoform (Cav1.2DHP(-)-knockin mice). We identified common and alternate ion channel couplings of Cav1.2 and Cav1.3 channels. Whereas hyperpolarizing Ca(2+)-dependent conductances were coupled to both Cav1.2 and Cav1.3 channels, an afterdepolarizing potential was only induced by the activity of Cav1.2 channels. Unexpectedly, the activity of Cav1.2 channels was found at relatively hyperpolarized membrane voltages. Our data add important information about the differences between Cav1.2 and Cav1.3 channels that furthers our understanding of the physiological and pathophysiological neuronal roles of these calcium channels. Moreover, our findings suggest that Cav1.3 knockout mice together with Cav1.2DHP(-)-knockin mice provide valuable models for future investigation of hippocampal LTCC-dependent afterdepolarizations.
Collapse
Affiliation(s)
- Julia Hasreiter
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lena Goldnagl
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Böhm
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
161
|
Uchitel OD, González Inchauspe C, Di Guilmi MN. Calcium channels and synaptic transmission in familial hemiplegic migraine type 1 animal models. Biophys Rev 2014; 6:15-26. [PMID: 28509957 DOI: 10.1007/s12551-013-0126-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 10/18/2013] [Indexed: 11/26/2022] Open
Abstract
One of the outstanding developments in clinical neurology has been the identification of ion channel mutations as the origin of a wide variety of inherited disorders like migraine, epilepsy, and ataxia. The study of several channelopathies has provided crucial insights into the molecular mechanisms, pathogenesis, and therapeutic approaches to complex neurological diseases. This review addresses the mutations underlying familial hemiplegic migraine (FHM) with particular interest in Cav2.1 (i.e., P/Q-type) voltage-activated Ca2+ channel FHM type-1 mutations (FHM1). Transgenic mice harboring the human pathogenic FHM1 mutation R192Q or S218L (KI) have been used as models to study neurotransmission at several central and peripheral synapses. FHM1 KI mice are a powerful tool to explore presynaptic regulation associated with expression of Cav2.1 channels. FHM1 Cav2.1 channels activate at more hyperpolarizing potentials and show an increased open probability. These biophysical alterations may lead to a gain-of-function on synaptic transmission depending upon factors such as action potential waveform and/or Cav2.1 splice variants and auxiliary subunits. Analysis of FHM knock-in mouse models has demonstrated a deficient regulation of the cortical excitation/inhibition (E/I) balance. The resulting excessive increases in cortical excitation may be the mechanisms that underlie abnormal sensory processing together with an increase in the susceptibility to cortical spreading depression (CSD). Increasing evidence from FHM KI animal studies support the idea that CSD, the underlying mechanism of aura, can activate trigeminal nociception, and thus trigger the headache mechanisms.
Collapse
Affiliation(s)
- Osvaldo D Uchitel
- Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET), Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, piso 2, Ciudad Universitaria, Buenos Aires, 1428, Argentina.
| | - Carlota González Inchauspe
- Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET), Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, piso 2, Ciudad Universitaria, Buenos Aires, 1428, Argentina
| | - Mariano N Di Guilmi
- Instituto de Fisiología, Biología Molecular y Neurociencias (CONICET), Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, piso 2, Ciudad Universitaria, Buenos Aires, 1428, Argentina
| |
Collapse
|
162
|
Allostery in Ca²⁺ channel modulation by calcium-binding proteins. Nat Chem Biol 2014; 10:231-8. [PMID: 24441587 DOI: 10.1038/nchembio.1436] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/25/2013] [Indexed: 12/17/2022]
Abstract
Distinguishing between allostery and competition among modulating ligands is challenging for large target molecules. Out of practical necessity, inferences are often drawn from in vitro assays on target fragments, but such inferences may belie actual mechanisms. One key example of such ambiguity concerns calcium-binding proteins (CaBPs) that tune signaling molecules regulated by calmodulin (CaM). As CaBPs resemble CaM, CaBPs are believed to competitively replace CaM on targets. Yet, brain CaM expression far surpasses that of CaBPs, raising questions as to whether CaBPs can exert appreciable biological actions. Here, we devise a live-cell, holomolecule approach that reveals an allosteric mechanism for calcium channels whose CaM-mediated inactivation is eliminated by CaBP4. Our strategy is to covalently link CaM and/or CaBP to holochannels, enabling live-cell fluorescence resonance energy transfer assays to resolve a cyclical allosteric binding scheme for CaM and CaBP4 to channels, thus explaining how trace CaBPs prevail. This approach may apply generally for discerning allostery in live cells.
Collapse
|
163
|
Depressed pacemaker activity of sinoatrial node myocytes contributes to the age-dependent decline in maximum heart rate. Proc Natl Acad Sci U S A 2013; 110:18011-6. [PMID: 24128759 DOI: 10.1073/pnas.1308477110] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An inexorable decline in maximum heart rate (mHR) progressively limits human aerobic capacity with advancing age. This decrease in mHR results from an age-dependent reduction in "intrinsic heart rate" (iHR), which is measured during autonomic blockade. The reduced iHR indicates, by definition, that pacemaker function of the sinoatrial node is compromised during aging. However, little is known about the properties of pacemaker myocytes in the aged sinoatrial node. Here, we show that depressed excitability of individual sinoatrial node myocytes (SAMs) contributes to reductions in heart rate with advancing age. We found that age-dependent declines in mHR and iHR in ECG recordings from mice were paralleled by declines in spontaneous action potential (AP) firing rates (FRs) in patch-clamp recordings from acutely isolated SAMs. The slower FR of aged SAMs resulted from changes in the AP waveform that were limited to hyperpolarization of the maximum diastolic potential and slowing of the early part of the diastolic depolarization. These AP waveform changes were associated with cellular hypertrophy, reduced current densities for L- and T-type Ca(2+) currents and the "funny current" (If), and a hyperpolarizing shift in the voltage dependence of If. The age-dependent reduction in sinoatrial node function was not associated with changes in β-adrenergic responsiveness, which was preserved during aging for heart rate, SAM FR, L- and T-type Ca(2+) currents, and If. Our results indicate that depressed excitability of individual SAMs due to altered ion channel activity contributes to the decline in mHR, and thus aerobic capacity, during normal aging.
Collapse
|
164
|
Bazzazi H, Ben Johny M, Adams PJ, Soong TW, Yue DT. Continuously tunable Ca(2+) regulation of RNA-edited CaV1.3 channels. Cell Rep 2013; 5:367-77. [PMID: 24120865 DOI: 10.1016/j.celrep.2013.09.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/02/2013] [Accepted: 09/05/2013] [Indexed: 11/18/2022] Open
Abstract
CaV1.3 ion channels are dominant Ca(2+) portals into pacemaking neurons, residing at the epicenter of brain rhythmicity and neurodegeneration. Negative Ca(2+) feedback regulation of CaV1.3 channels (CDI) is therefore critical for Ca(2+) homeostasis. Intriguingly, nearly half the CaV1.3 transcripts in the brain are RNA edited to reduce CDI and influence oscillatory activity. It is then mechanistically remarkable that this editing occurs precisely within an IQ domain, whose interaction with Ca(2+)-bound calmodulin (Ca(2+)/CaM) is believed to induce CDI. Here, we sought the mechanism underlying the altered CDI of edited channels. Unexpectedly, editing failed to attenuate Ca(2+)/CaM binding. Instead, editing weakened the prebinding of Ca(2+)-free CaM (apoCaM) to channels, which proves essential for CDI. Thus, editing might render CDI continuously tunable by fluctuations in ambient CaM, a prominent effect we substantiate in substantia nigral neurons. This adjustability of Ca(2+) regulation by CaM now looms as a key element of CNS Ca(2+) homeostasis.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Calcium Signals Laboratory, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, Ross Building, Room 713, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
165
|
Chen R, Zeng X, Zhang R, Huang J, Kuang X, Yang J, Liu J, Tawfik O, Thrasher JB, Li B. Cav1.3 channel α1D protein is overexpressed and modulates androgen receptor transactivation in prostate cancers. Urol Oncol 2013; 32:524-36. [PMID: 24054868 DOI: 10.1016/j.urolonc.2013.05.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 05/20/2013] [Accepted: 05/26/2013] [Indexed: 01/10/2023]
Abstract
Widespread use of L-type calcium channel blockers for treating hypertension has led to multiple epidemiologic studies to assess the risk of prostate cancer incidence. These studies revealed a reverse correlation between the likelihood of prostate cancer risk and the use of L-type calcium channel blockers among men without family history but the mechanism was not clear. In this study, we examined the expression profiles of multiple L-type calcium channel genes in prostate cancers and determined their functional roles in androgen receptor (AR) transactivation and cell growth. By reanalyzing the ONCOMINE database, we found that L-type calcium channel CACNA1D gene expression levels in cancer tissues were significantly higher than noncancer tissues in 14 of 15 published complementary deoxyribonucleic acid microarray data sets, of which 9 data sets showed an increase of 2- to 17-folds. Quantitative polymerase chain reaction and immunostaining experiments revealed that CACNA1D gene and its coding protein α1D were highly expressed in prostate cancers, especially in castration-resistant diseases, compared with benign prostate tissues. Consistent with the notion of CACNA1D as an ERG-regulated gene, CACNA1D gene expression levels were significantly higher in prostate cancers with TMPRSS2-ERG gene fusion compared with the cases without this gene fusion. Blocking L-type channel's function or knocking down CACNA1D gene expression significantly suppressed androgen-stimulated Ca(2+) influx, AR transactivation, and cell growth in prostate cancer cells. Taken together, these data suggest that CACNA1D gene overexpression is associated with prostate cancer progression and might play an important role in Ca(2+) influx, AR activation, and cell growth in prostate cancer cells.
Collapse
Affiliation(s)
- Ruibao Chen
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS; Department of Urology, Tongji Hospital, Huanzhong University of Science & Technology, Wuhan, China
| | - Xing Zeng
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS; Department of Urology, Tongji Hospital, Huanzhong University of Science & Technology, Wuhan, China
| | - Ruitao Zhang
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS; Department of Anatomy and Pharmacology, Three Gorges University College of Medicine, Yichang, China
| | - Jiaoti Huang
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Xiangxing Kuang
- Department of Urology, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China
| | - Jun Yang
- Department of Urology, Tongji Hospital, Huanzhong University of Science & Technology, Wuhan, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Huanzhong University of Science & Technology, Wuhan, China
| | - Ossama Tawfik
- Department of Pathology & Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS
| | | | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS; Department of Anatomy and Pharmacology, Three Gorges University College of Medicine, Yichang, China; Department of Urology, The Affiliated Hospital, Guangdong Medical College, Zhanjiang, China.
| |
Collapse
|
166
|
Abstract
Local Ca(2+) signals through voltage-gated Ca(2+) channels (CaVs) drive synaptic transmission, neural plasticity, and cardiac contraction. Despite the importance of these events, the fundamental relationship between flux through a single CaV channel and the Ca(2+) signaling concentration within nanometers of its pore has resisted empirical determination, owing to limitations in the spatial resolution and specificity of fluorescence-based Ca(2+) measurements. Here, we exploited Ca(2+)-dependent inactivation of CaV channels as a nanometer-range Ca(2+) indicator specific to active channels. We observed an unexpected and dramatic boost in nanodomain Ca(2+) amplitude, ten-fold higher than predicted on theoretical grounds. Our results uncover a striking feature of CaV nanodomains, as diffusion-restricted environments that amplify small Ca(2+) fluxes into enormous local Ca(2+) concentrations. This Ca(2+) tuning by the physical composition of the nanodomain may represent an energy-efficient means of local amplification that maximizes information signaling capacity, while minimizing global Ca(2+) load.
Collapse
|
167
|
Meza U, Beqollari D, Romberg CF, Papadopoulos S, Bannister RA. Potent inhibition of L-type Ca2+ currents by a Rad variant associated with congestive heart failure. Biochem Biophys Res Commun 2013; 439:270-4. [PMID: 23973784 DOI: 10.1016/j.bbrc.2013.08.044] [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: 08/11/2013] [Accepted: 08/13/2013] [Indexed: 10/26/2022]
Abstract
Ca(2+) influx via L-type voltage-gated Ca(2+) channels supports the plateau phase of ventricular action potentials and is the trigger for excitation-contraction (EC) coupling in the myocardium. Rad, a member of the RGK (Rem, Rem2, Rad, Gem/Kir) family of monomeric G proteins, regulates ventricular action potential duration and EC coupling gain through its ability to inhibit cardiac L-type channel activity. In this study, we have investigated the potential dysfunction of a naturally occurring Rad variant (Q66P) that has been associated with congestive heart failure in humans. Specifically, we have tested whether Rad Q66P limits, or even eliminates, the inhibitory actions of Rad on CaV1.2 and CaV1.3, the two L-type channel isoforms known to be expressed in the heart. We have found that mouse Rad Q65P (the murine equivalent of human Rad Q66P) inhibits L-type currents conducted by CaV1.2 or CaV1.3 channels as potently as wild-type Rad (>95% inhibition of both channels). In addition, Rad Q65P attenuates the gating movement of both channels as effectively as wild-type Rad, indicating that the Q65P substitution does not differentially impair any of the three described modes of L-type channel inhibition by RGK proteins. Thus, we conclude that if Rad Q66P contributes to cardiomyopathy, it does so via a mechanism that is not related to its ability to inhibit L-type channel-dependent processes per se. However, our results do not rule out the possibility that decreased expression, mistargeting or altered regulation of Rad Q66P may reduce the RGK protein's efficacy in vivo.
Collapse
Affiliation(s)
- U Meza
- Department of Medicine-Cardiology Division, University of Colorado Denver-Anschutz Medical Campus, 12700 East 19th Avenue, P15-8006, B-139, Aurora, CO 80045, USA; Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Venustiano Carranza #2405, San Luis Potosí, SLP 78210, México.
| | | | | | | | | |
Collapse
|
168
|
Huang H, Yu D, Soong TW. C-Terminal Alternative Splicing of CaV1.3 Channels Distinctively Modulates Their Dihydropyridine Sensitivity. Mol Pharmacol 2013; 84:643-53. [DOI: 10.1124/mol.113.087155] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
169
|
Mathur BN, Tanahira C, Tamamaki N, Lovinger DM. Voltage drives diverse endocannabinoid signals to mediate striatal microcircuit-specific plasticity. Nat Neurosci 2013; 16:1275-83. [PMID: 23892554 PMCID: PMC3758434 DOI: 10.1038/nn.3478] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 06/20/2013] [Indexed: 11/09/2022]
Abstract
The dorsolateral striatum and cannabinoid type 1 receptor (CB1) signaling mediate habitual action learning, which is thought to require a balance of activity in the direct and indirect striatal output pathways. However, very little is known about how the high CB1-expressing striatal inhibitory microcircuitry might contribute to long-term plasticity capable of sculpting direct/indirect pathway output. Using optogenetic and molecular interrogation of striatal GABAergic microcircuits, we describe novel mechanisms of voltage-dependent long-term depression of inhibitory synapses (iLTD) onto mouse and rat medium spiny projection neurons (MSNs). This iLTD involves recruitment of different endocannabinoid types and shows both presynaptic and postsynaptic selectivity for MSN subtypes, ultimately resulting in a powerful disinhibition of direct pathway MSNs. These results indicate a new role for voltage states in gating circuit-specific forms of synaptic plasticity and illuminate possible circuit dynamics underlying action control.
Collapse
Affiliation(s)
- Brian N Mathur
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
170
|
Kang S, Cooper G, Dunne SF, Luan CH, James Surmeier D, Silverman RB. Antagonism of L-type Ca2+ channels CaV1.3 and CaV1.2 by 1,4-dihydropyrimidines and 4H-pyrans as dihydropyridine mimics. Bioorg Med Chem 2013; 21:4365-73. [DOI: 10.1016/j.bmc.2013.04.054] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/12/2013] [Accepted: 04/22/2013] [Indexed: 10/26/2022]
|
171
|
Gurkoff G, Shahlaie K, Lyeth B, Berman R. Voltage-gated calcium channel antagonists and traumatic brain injury. Pharmaceuticals (Basel) 2013; 6:788-812. [PMID: 24276315 PMCID: PMC3816709 DOI: 10.3390/ph6070788] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 01/17/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States. Despite more than 30 years of research, no pharmacological agents have been identified that improve neurological function following TBI. However, several lines of research described in this review provide support for further development of voltage gated calcium channel (VGCC) antagonists as potential therapeutic agents. Following TBI, neurons and astrocytes experience a rapid and sometimes enduring increase in intracellular calcium ([Ca2+]i). These fluxes in [Ca2+]i drive not only apoptotic and necrotic cell death, but also can lead to long-term cell dysfunction in surviving cells. In a limited number of in vitro experiments, both L-type and N-type VGCC antagonists successfully reduced calcium loads as well as neuronal and astrocytic cell death following mechanical injury. In rodent models of TBI, administration of VGCC antagonists reduced cell death and improved cognitive function. It is clear that there is a critical need to find effective therapeutics and rational drug delivery strategies for the management and treatment of TBI, and we believe that further investigation of VGCC antagonists should be pursued before ruling out the possibility of successful translation to the clinic.
Collapse
Affiliation(s)
- Gene Gurkoff
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
- NSF Center for Biophotonics Science and Technology, Suite 2700 Stockton Blvd, Suite 1400, Sacramento, CA, 95817, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-530-754-7501; Fax: +1-530-754-5125
| | - Kiarash Shahlaie
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| | - Bruce Lyeth
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| | - Robert Berman
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| |
Collapse
|
172
|
CaV1.3-selective L-type calcium channel antagonists as potential new therapeutics for Parkinson's disease. Nat Commun 2013; 3:1146. [PMID: 23093183 DOI: 10.1038/ncomms2149] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 09/20/2012] [Indexed: 12/29/2022] Open
Abstract
L-type calcium channels expressed in the brain are heterogeneous. The predominant class of L-type calcium channels has a Ca(V)1.2 pore-forming subunit. L-type calcium channels with a Ca(V)1.3 pore-forming subunit are much less abundant, but have been implicated in the generation of mitochondrial oxidant stress underlying pathogenesis in Parkinson's disease. Thus, selectively antagonizing Ca(V)1.3 L-type calcium channels could provide a means of diminishing cell loss in Parkinson's disease without producing side effects accompanying general antagonism of L-type calcium channels. However, there are no known selective antagonists of Ca(V)1.3 L-type calcium channel. Here we report high-throughput screening of commercial and 'in-house' chemical libraries and modification of promising hits. Pyrimidine-2,4,6-triones were identified as a potential scaffold; structure-activity relationship-based modification of this scaffold led to 1-(3-chlorophenethyl)-3-cyclopentylpyrimidine-2,4,6-(1H,3H,5H)-trione (8), a potent and highly selective Ca(V)1.3 L-type calcium channel antagonist. The biological relevance was confirmed by whole-cell patch-clamp electrophysiology. These studies describe the first highly selective Ca(V)1.3 L-type calcium channel antagonist and point to a novel therapeutic strategy for Parkinson's disease.
Collapse
|
173
|
Raised activity of L-type calcium channels renders neurons prone to form paroxysmal depolarization shifts. Neuromolecular Med 2013; 15:476-92. [PMID: 23695859 PMCID: PMC3732764 DOI: 10.1007/s12017-013-8234-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 05/08/2013] [Indexed: 12/31/2022]
Abstract
Neuronal L-type voltage-gated calcium channels (LTCCs) are involved in several physiological functions, but increased activity of LTCCs has been linked to pathology. Due to the coupling of LTCC-mediated Ca2+ influx to Ca2+-dependent conductances, such as KCa or non-specific cation channels, LTCCs act as important regulators of neuronal excitability. Augmentation of after-hyperpolarizations may be one mechanism that shows how elevated LTCC activity can lead to neurological malfunctions. However, little is known about other impacts on electrical discharge activity. We used pharmacological up-regulation of LTCCs to address this issue on primary rat hippocampal neurons. Potentiation of LTCCs with Bay K8644 enhanced excitatory postsynaptic potentials to various degrees and eventually resulted in paroxysmal depolarization shifts (PDS). Under conditions of disturbed Ca2+ homeostasis, PDS were evoked frequently upon LTCC potentiation. Exposing the neurons to oxidative stress using hydrogen peroxide also induced LTCC-dependent PDS. Hence, raising LTCC activity had unidirectional effects on brief electrical signals and increased the likeliness of epileptiform events. However, long-lasting seizure-like activity induced by various pharmacological means was affected by Bay K8644 in a bimodal manner, with increases in one group of neurons and decreases in another group. In each group, isradipine exerted the opposite effect. This suggests that therapeutic reduction in LTCC activity may have little beneficial or even adverse effects on long-lasting abnormal discharge activities. However, our data identify enhanced activity of LTCCs as one precipitating cause of PDS. Because evidence is continuously accumulating that PDS represent important elements in neuropathogenesis, LTCCs may provide valuable targets for neuroprophylactic therapy.
Collapse
|
174
|
Trippier PC, Labby KJ, Hawker DD, Mataka JJ, Silverman RB. Target- and mechanism-based therapeutics for neurodegenerative diseases: strength in numbers. J Med Chem 2013; 56:3121-47. [PMID: 23458846 PMCID: PMC3637880 DOI: 10.1021/jm3015926] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of new therapeutics for the treatment of neurodegenerative pathophysiologies currently stands at a crossroads. This presents an opportunity to transition future drug discovery efforts to target disease modification, an area in which much still remains unknown. In this Perspective we examine recent progress in the areas of neurodegenerative drug discovery, focusing on some of the most common targets and mechanisms: N-methyl-d-aspartic acid (NMDA) receptors, voltage gated calcium channels (VGCCs), neuronal nitric oxide synthase (nNOS), oxidative stress from reactive oxygen species, and protein aggregation. These represent the key players identified in neurodegeneration and are part of a complex, intertwined signaling cascade. The synergistic delivery of two or more compounds directed against these targets, along with the design of small molecules with multiple modes of action, should be explored in pursuit of more effective clinical treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul C. Trippier
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Kristin Jansen Labby
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Dustin D. Hawker
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Jan J. Mataka
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Richard B. Silverman
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA
| |
Collapse
|
175
|
Stecina K, Fedirchuk B, Hultborn H. Information to cerebellum on spinal motor networks mediated by the dorsal spinocerebellar tract. J Physiol 2013; 591:5433-43. [PMID: 23613538 DOI: 10.1113/jphysiol.2012.249110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The main objective of this review is to re-examine the type of information transmitted by the dorsal and ventral spinocerebellar tracts (DSCT and VSCT respectively) during rhythmic motor actions such as locomotion. Based on experiments in the 1960s and 1970s, the DSCT was viewed as a relay of peripheral sensory input to the cerebellum in general, and during rhythmic movements such as locomotion and scratch. In contrast, the VSCT was seen as conveying a copy of the output of spinal neuronal circuitry, including those circuits generating rhythmic motor activity (the spinal central pattern generator, CPG). Emerging anatomical and electrophysiological information on the putative subpopulations of DSCT and VSCT neurons suggest differentiated functions for some of the subpopulations. Multiple lines of evidence support the notion that sensory input is not the only source driving DSCT neurons and, overall, there is a greater similarity between DSCT and VSCT activity than previously acknowledged. Indeed the majority of DSCT cells can be driven by spinal CPGs for locomotion and scratch without phasic sensory input. It thus seems natural to propose the possibility that CPG input to some of these neurons may contribute to distinguishing sensory inputs that are a consequence of the active locomotion from those resulting from perturbations in the external world.
Collapse
Affiliation(s)
- Katinka Stecina
- K. Stecina: University of Copenhagen, Department of Neuroscience and Pharmacology, The Panum Institute, 33.3, Blegdamsvej 3, Copenhagen 2200, Denmark.
| | | | | |
Collapse
|
176
|
Gregory FD, Pangrsic T, Calin-Jageman IE, Moser T, Lee A. Harmonin enhances voltage-dependent facilitation of Cav1.3 channels and synchronous exocytosis in mouse inner hair cells. J Physiol 2013; 591:3253-69. [PMID: 23613530 DOI: 10.1113/jphysiol.2013.254367] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cav1.3 channels mediate Ca(2+) influx that triggers exocytosis of glutamate at cochlear inner hair cell (IHC) synapses. Harmonin is a PDZ-domain-containing protein that interacts with the C-terminus of the Cav1.3 α1 subunit (α11.3) and controls cell surface Cav1.3 levels by promoting ubiquitin-dependent proteosomal degradation. However, PDZ-domain-containing proteins have diverse functions and regulate other Cav1.3 properties, which could collectively influence presynaptic transmitter release. Here, we report that harmonin binding to the α11.3 distal C-terminus (dCT) enhances voltage-dependent facilitation (VDF) of Cav1.3 currents both in transfected HEK293T cells and in mouse inner hair cells. In HEK293T cells, this effect of harmonin was greater for Cav1.3 channels containing the auxiliary Cav β1 than with the β2 auxiliary subunit. Cav1.3 channels lacking the α11.3 dCT were insensitive to harmonin modulation. Moreover, the 'deaf-circler' dfcr mutant form of harmonin, which does not interact with the α11.3 dCT, did not promote VDF. In mature IHCs from mice expressing the dfcr harmonin mutant, Cav1.3 VDF was less than in control IHCs. This difference was not observed between control and dfcr IHCs prior to hearing onset. Membrane capacitance recordings from dfcr IHCs revealed a role for harmonin in synchronous exocytosis and in increasing the efficiency of Ca(2+) influx for triggering exocytosis. Collectively, our results indicate a multifaceted presynaptic role of harmonin in IHCs in regulating Cav1.3 Ca(2+) channels and exocytosis.
Collapse
Affiliation(s)
- Frederick D Gregory
- Department of Molecular Physiology and Biophysics, University of Iowa, 5-610 Bowen Science Building, 51 Newton Rd, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
177
|
Inagaki A, Lee A. Developmental alterations in the biophysical properties of Ca(v) 1.3 Ca(2+) channels in mouse inner hair cells. Channels (Austin) 2013; 7:171-81. [PMID: 23510940 PMCID: PMC3710344 DOI: 10.4161/chan.24104] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Prior to hearing onset, spontaneous action potentials activate voltage-gated Cav1.3 Ca2+ channels in mouse inner hair cells (IHCs), which triggers exocytosis of glutamate and excitation of afferent neurons. In mature IHCs, Cav1.3 channels open in response to evoked receptor potentials, causing graded changes in exocytosis required for accurate sound transmission. Developmental alterations in Cav1.3 properties may support distinct roles of Cav1.3 in IHCs in immature and mature IHCs, and have been reported in various species. It is not known whether such changes in Cav1.3 properties occur in mouse IHCs, but this knowledge is necessary for understanding the roles of Cav1.3 in developing and mature IHCs. Here, we describe age-dependent differences in the biophysical properties of Cav1.3 channels in mouse IHCs. In mature IHCs, Cav1.3 channels activate more rapidly and exhibit greater Ca2+-dependent inactivation (CDI) than in immature IHCs. Consistent with the properties of Cav1.3 channels in heterologous expression systems, CDI in mature IHCs is not affected by increasing intracellular Ca2+ buffering strength. However, CDI in immature IHCs is significantly reduced by strong intracellular Ca2+ buffering, which both slows the onset of, and accelerates recovery from, inactivation. These results signify a developmental decline in the sensitivity of CDI to global elevations in Ca2+, which restricts negative feedback regulation of Cav1.3 channels to incoming Ca2+ ions in mature IHCs. Together with faster Cav1.3 activation kinetics, increased reliance of Cav1.3 CDI on local Ca2+ may sharpen presynaptic Ca2+ signals and improve temporal aspects of sound coding in mature IHCs.
Collapse
Affiliation(s)
- Akira Inagaki
- Departments of Molecular Physiology and Biophysics, Otolaryngology-Head and Neck Surgery, and Neurology, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
178
|
Diminished vision in healthy aging is associated with increased retinal L-type voltage gated calcium channel ion influx. PLoS One 2013; 8:e56340. [PMID: 23457553 PMCID: PMC3572962 DOI: 10.1371/journal.pone.0056340] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/08/2013] [Indexed: 11/19/2022] Open
Abstract
Extensive evidence implicates an increase in hippocampal L-type voltage-gated calcium channel (L-VGCC) expression, and ion influx through these channels, in age-related cognitive declines. Here, we ask if this “calcium hypothesis" applies to the neuroretina: Is increased influx via L-VGCCs related to the well-documented but poorly-understood vision declines in healthy aging? In Long-Evans rats we find a significant age-related increase in ion flux through retinal L-VGCCs in vivo (manganese-enhanced MRI (MEMRI)) that are longitudinally linked with progressive vision declines (optokinetic tracking). Importantly, the degree of retinal Mn2+ uptake early in adulthood significantly predicted later visual contrast sensitivity declines. Furthermore, as in the aging hippocampus, retinal expression of a drug-insensitive L-VGCC isoform (α1D) increased – a pattern confirmed in vivo by an age-related decline in sensitivity to L-VGCC blockade. These data highlight mechanistic similarities between retinal and hippocampal aging, and raise the possibility of new treatment targets for minimizing vision loss during healthy aging.
Collapse
|
179
|
Bengtson CP, Kaiser M, Obermayer J, Bading H. Calcium responses to synaptically activated bursts of action potentials and their synapse-independent replay in cultured networks of hippocampal neurons. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1672-9. [PMID: 23360982 DOI: 10.1016/j.bbamcr.2013.01.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/15/2013] [Accepted: 01/18/2013] [Indexed: 11/29/2022]
Abstract
Both synaptic N-methyl-d-aspartate (NMDA) receptors and voltage-operated calcium channels (VOCCs) have been shown to be critical for nuclear calcium signals associated with transcriptional responses to bursts of synaptic input. However the direct contribution to nuclear calcium signals from calcium influx through NMDA receptors and VOCCs has been obscured by their concurrent roles in action potential generation and synaptic transmission. Here we compare calcium responses to synaptically induced bursts of action potentials with identical bursts devoid of any synaptic contribution generated using the pre-recorded burst as the voltage clamp command input to replay the burst in the presence of blockers of action potentials or ionotropic glutamate receptors. Synapse independent replays of bursts produced nuclear calcium responses with amplitudes around 70% of their original synaptically generated signals and were abolished by the L-type VOCC blocker, verapamil. These results identify a major direct source of nuclear calcium from local L-type VOCCs whose activation is boosted by NMDA receptor dependent depolarization. The residual component of synaptically induced nuclear calcium signals which was both VOCC independent and NMDA receptor dependent showed delayed kinetics consistent with a more distal source such as synaptic NMDA receptors or internal stores. The dual requirement of NMDA receptors and L-type VOCCs for synaptic activity-induced nuclear calcium dependent transcriptional responses most likely reflects a direct somatic calcium influx from VOCCs whose activation is amplified by synaptic NMDA receptor-mediated depolarization and whose calcium signal is boosted by a delayed input from distal calcium sources mostly likely entry through NMDA receptors and release from internal stores. This article is part of a Special Issue entitled: 12th European Symposium on Calcium.
Collapse
Affiliation(s)
- C Peter Bengtson
- Department of Neurobiology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | |
Collapse
|
180
|
Vandael DHF, Mahapatra S, Calorio C, Marcantoni A, Carbone E. Cav1.3 and Cav1.2 channels of adrenal chromaffin cells: emerging views on cAMP/cGMP-mediated phosphorylation and role in pacemaking. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1608-18. [PMID: 23159773 DOI: 10.1016/j.bbamem.2012.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 11/05/2012] [Accepted: 11/08/2012] [Indexed: 12/21/2022]
Abstract
Voltage-gated Ca²⁺ channels (VGCCs) are voltage sensors that convert membrane depolarizations into Ca²⁺ signals. In the chromaffin cells of the adrenal medulla, the Ca²⁺ signals driven by VGCCs regulate catecholamine secretion, vesicle retrievals, action potential shape and firing frequency. Among the VGCC-types expressed in these cells (N-, L-, P/Q-, R- and T-types), the two L-type isoforms, Ca(v)1.2 and Ca(v)1.3, control key activities due to their particular activation-inactivation gating and high-density of expression in rodents and humans. The two isoforms are also effectively modulated by G protein-coupled receptor pathways delimited in membrane micro-domains and by the cAMP/PKA and NO/cGMP/PKG phosphorylation pathways which induce prominent Ca²⁺ current changes if opposingly regulated. The two L-type isoforms shape the action potential and directly participate to vesicle exocytosis and endocytosis. The low-threshold of activation and slow rate of inactivation of Ca(v)1.3 confer to this channel the unique property of carrying sufficient inward current at subthreshold potentials able to activate BK and SK channels which set the resting potential, the action potential shape, the cell firing mode and the degree of spike frequency adaptation during spontaneous firing or sustained depolarizations. These properties help chromaffin cells to optimally adapt when switching from normal to stress-mimicking conditions. Here, we will review past and recent findings on cAMP- and cGMP-mediated modulations of Ca(v)1.2 and Ca(v)1.3 and the role that these channels play in the control of chromaffin cell firing. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- D H F Vandael
- Department of Drug Science, Laboratory of Cellular & Molecular Neuroscience, NIS Center, CNISM, University of Torino, Italy
| | | | | | | | | |
Collapse
|
181
|
Levels of Ca(V)1.2 L-Type Ca(2+) Channels Peak in the First Two Weeks in Rat Hippocampus Whereas Ca(V)1.3 Channels Steadily Increase through Development. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:597214. [PMID: 23097697 PMCID: PMC3477797 DOI: 10.1155/2012/597214] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 06/04/2012] [Indexed: 12/20/2022]
Abstract
Influx of calcium through voltage-dependent channels regulates processes throughout the nervous system. Specifically, influx through L-type channels plays a variety of roles in early neuronal development and is commonly modulated by G-protein-coupled receptors such as GABA(B) receptors. Of the four isoforms of L-type channels, only Ca(V)1.2 and Ca(V)1.3 are predominately expressed in the nervous system. Both isoforms are inhibited by the same pharmacological agents, so it has been difficult to determine the role of specific isoforms in physiological processes. In the present study, Western blot analysis and confocal microscopy were utilized to study developmental expression levels and patterns of Ca(V)1.2 and Ca(V)1.3 in the CA1 region of rat hippocampus. Steady-state expression of Ca(V)1.2 predominated during the early neonatal period decreasing by day 12. Steady-state expression of Ca(V)1.3 was low at birth and gradually rose to adult levels by postnatal day 15. In immunohistochemical studies, antibodies against Ca(V)1.2 and Ca(V)1.3 demonstrated the highest intensity of labeling in the proximal dendrites at all ages studied (P1-72). Immunohistochemical studies on one-week-old hippocampi demonstrated significantly more colocalization of GABA(B) receptors with Ca(V)1.2 than with Ca(V)1.3, suggesting that modulation of L-type calcium current in early development is mediated through Ca(V)1.2 channels.
Collapse
|
182
|
Christel CJ, Cardona N, Mesirca P, Herrmann S, Hofmann F, Striessnig J, Ludwig A, Mangoni ME, Lee A. Distinct localization and modulation of Cav1.2 and Cav1.3 L-type Ca2+ channels in mouse sinoatrial node. J Physiol 2012; 590:6327-42. [PMID: 23045342 DOI: 10.1113/jphysiol.2012.239954] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysregulation of L-type Ca(2+) currents in sinoatrial nodal (SAN) cells causes cardiac arrhythmia. Both Ca(v)1.2 and Ca(v)1.3 channels mediate sinoatrial L-type currents. Whether these channels exhibit differences in modulation and localization, which could affect their contribution to pacemaking, is unknown. In this study, we characterized voltage-dependent facilitation (VDF) and subcellular localization of Ca(v)1.2 and Ca(v)1.3 channels in mouse SAN cells and determined how these properties of Ca(v)1.3 affect sinoatrial pacemaking in a mathematical model. Whole cell Ba(2+) currents were recorded from SAN cells from mice carrying a point mutation that renders Ca(v)1.2 channels relatively insensitive to dihydropyridine antagonists. The Ca(v)1.2-mediated current was isolated in the presence of nimodipine (1 μm), which was subtracted from the total current to yield the Ca(v)1.3 component. With strong depolarizations (+80 mV), Ca(v)1.2 underwent significantly stronger inactivation than Ca(v)1.3. VDF of Ca(v)1.3 was evident during recovery from inactivation at a time when Ca(v)1.2 remained inactivated. By immunofluorescence, Ca(v)1.3 colocalized with ryanodine receptors in sarcomeric structures while Ca(v)1.2 was largely restricted to the delimiting plasma membrane. Ca(v)1.3 VDF enhanced recovery of pacemaker activity after pauses and positively regulated pacemaking during slow heart rate in a numerical model of mouse SAN automaticity, including preferential coupling of Ca(v)1.3 to ryanodine receptor-mediated Ca(2+) release. We conclude that strong VDF and colocalization with ryanodine receptors in mouse SAN cells are unique properties that may underlie a specific role for Ca(v)1.3 in opposing abnormal slowing of heart rate.
Collapse
Affiliation(s)
- Carl J Christel
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Alternative splicing: functional diversity among voltage-gated calcium channels and behavioral consequences. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1522-9. [PMID: 23022282 DOI: 10.1016/j.bbamem.2012.09.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 09/15/2012] [Accepted: 09/19/2012] [Indexed: 12/14/2022]
Abstract
Neuronal voltage-gated calcium channels generate rapid, transient intracellular calcium signals in response to membrane depolarization. Neuronal Ca(V) channels regulate a range of cellular functions and are implicated in a variety of neurological and psychiatric diseases including epilepsy, Parkinson's disease, chronic pain, schizophrenia, and bipolar disorder. Each mammalian Cacna1 gene has the potential to generate tens to thousands of Ca(V) channels by alternative pre-mRNA splicing, a process that adds fine granulation to the pool of Ca(V) channel structures and functions. The precise composition of Ca(V) channel splice isoform mRNAs expressed in each cell are controlled by cell-specific splicing factors. The activity of splicing factors are in turn regulated by molecules that encode various cellular features, including cell-type, activity, metabolic states, developmental state, and other factors. The cellular and behavioral consequences of individual sites of Ca(V) splice isoforms are being elucidated, as are the cell-specific splicing factors that control splice isoform selection. Altered patterns of alternative splicing of Ca(V) pre-mRNAs can alter behavior in subtle but measurable ways, with the potential to influence drug efficacy and disease severity. This article is part of a Special Issue entitled: Calcium channels.
Collapse
|
184
|
|
185
|
Mahapatra S, Marcantoni A, Zuccotti A, Carabelli V, Carbone E. Equal sensitivity of Cav1.2 and Cav1.3 channels to the opposing modulations of PKA and PKG in mouse chromaffin cells. J Physiol 2012; 590:5053-73. [PMID: 22826131 DOI: 10.1113/jphysiol.2012.236729] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mouse chromaffin cells (MCCs) express high densities of L-type Ca2+ channels (LTCCs), which control pacemaking activity and catecholamine secretion proportionally to their density of expression. In vivo phosphorylation of LTCCs by cAMP-PKA and cGMP–PKG, regulate LTCC gating in two opposing ways: the cAMP-PKA pathway potentiates while the cGMP–PKG cascade inhibits LTCCs. Despite this, no attempts have been made to answer three key questions related to the two Cav1 isoforms expressed in MCCs (Cav1.2 and Cav1.3): (i) how much are the two Cav1 channels basally modulated by PKA and PKG?, (ii) to what extent can Cav1.2 and Cav1.3 be further regulated by PKA or PKG activation?, and (iii) are the effects of both kinases cumulative when simultaneously active? Here, by comparing the size of L-type currents of wild-type (WT; Cav1.2+Cav1.3) and Cav1.3−/− KO (Cav1.2) MCCs, we provide new evidence that both PKA and PKG pathways affect Cav1.2 and Cav1.3 to the same extent either under basal conditions or induced stimulation. Inhibition of PKA by H89 (5 μM) reduced the L-type current in WT and KO MCCs by∼60%,while inhibition of PKG by KT 5823 (1 μM) increased by∼40% the same current in both cell types. Given that Cav1.2 and Cav1.3 carry the same quantity of Ca2+ currents, this suggests equal sensitivity of Cav1.2 and Cav1.3 to the two basal modulatory pathways. Maximal stimulation of cAMP–PKA by forskolin (100 μM) and activation of cGMP–PKG by pCPT-cGMP (1mM) uncovered a∼25% increase of L-type currents in the first case and∼65% inhibition in the second case in both WT and KO MCCs, suggesting equal sensitivity of Cav1.2 and Cav1.3 during maximal PKA or PKG stimulation. The effects of PKA and PKG were cumulative and most evident when one pathway was activated and the other was inhibited. The two extreme combinations(PKA activation–PKG inhibition vs. PKG activation-PKA inhibition) varied the size of L-type currents by one order of magnitude (from 180% to 18% of control size). Taken together our data suggest that: (i) Cav1.2 and Cav1.3 are equally sensitive to PKA and PKG action under both basal conditions and maximal stimulation, and (ii) PKA and PKG act independently on both Cav1.2 and Cav1.3, producing cumulative effects when opposingly activated. These extreme Cav1 channel modulations may occur either during high-frequency sympathetic stimulation to sustain prolonged catecholamine release (maximal L-type current) or following activation of the NO–cGMP–PKG signalling pathway (minimal L-type current) to limit the steady release of catecholamines.
Collapse
|
186
|
Huang H, Tan BZ, Shen Y, Tao J, Jiang F, Sung YY, Ng CK, Raida M, Köhr G, Higuchi M, Fatemi-Shariatpanahi H, Harden B, Yue DT, Soong TW. RNA editing of the IQ domain in Ca(v)1.3 channels modulates their Ca²⁺-dependent inactivation. Neuron 2012; 73:304-16. [PMID: 22284185 DOI: 10.1016/j.neuron.2011.11.022] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2011] [Indexed: 11/29/2022]
Abstract
Adenosine-to-inosine RNA editing is crucial for generating molecular diversity, and serves to regulate protein function through recoding of genomic information. Here, we discover editing within Ca(v)1.3 Ca²⁺ channels, renown for low-voltage Ca²⁺-influx and neuronal pacemaking. Significantly, editing occurs within the channel's IQ domain, a calmodulin-binding site mediating inhibitory Ca²⁺-feedback (CDI) on channels. The editing turns out to require RNA adenosine deaminase ADAR2, whose variable activity could underlie a spatially diverse pattern of Ca(v)1.3 editing seen across the brain. Edited Ca(v)1.3 protein is detected both in brain tissue and within the surface membrane of primary neurons. Functionally, edited Ca(v)1.3 channels exhibit strong reduction of CDI; in particular, neurons within the suprachiasmatic nucleus show diminished CDI, with higher frequencies of repetitive action-potential and calcium-spike activity, in wild-type versus ADAR2 knockout mice. Our study reveals a mechanism for fine-tuning Ca(v)1.3 channel properties in CNS, which likely impacts a broad spectrum of neurobiological functions.
Collapse
Affiliation(s)
- Hua Huang
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, 117597 Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Jurkovičová-Tarabová B, Griesemer D, Pirone A, Sinnegger-Brauns MJ, Striessnig J, Friauf E. Repertoire of high voltage-activated Ca2+ channels in the lateral superior olive: functional analysis in wild-type, Cav1.3−/−, and Cav1.2DHP−/− mice. J Neurophysiol 2012; 108:365-79. [DOI: 10.1152/jn.00948.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Voltage-gated Ca2+ (Cav)1.3 α-subunits of high voltage-activated Ca2+ channels (HVACCs) are essential for Ca2+ influx and transmitter release in cochlear inner hair cells and therefore for signal transmission into the central auditory pathway. Their absence leads to deafness and to striking structural changes in the auditory brain stem, particularly in the lateral superior olive (LSO). Here, we analyzed the contribution of various types of HVACCs to the total Ca2+ current ( ICa) in developing mouse LSO neurons to address several questions: do LSO neurons express functional Cav1.3 channels? What other types of HVACCs are expressed? Are there developmental changes? Do LSO neurons of Cav1.3−/− mice show any compensatory responses, namely, upregulation of other HVACCs? Our electrophysiological and pharmacological results showed the presence of functional Cav1.3 and Cav1.2 channels at both postnatal days 4 and 12. Aside from these L-type channels, LSO neurons also expressed functional P/Q-type, N-type, and, most likely, R-type channels. The relative contribution of the four different subtypes to ICa appeared to be 45%, 29%, 22%, and 4% at postnatal day 12, respectively. The physiological results were flanked and extended by quantitative RT-PCR data. Altogether, LSO neurons displayed a broad repertoire of HVACC subtypes. Genetic ablation of Cav1.3 resulted in functional reorganization of some other HVACCs but did not restore normal ICa properties. Together, our results suggest that several types of HVACCs are of functional relevance for the developing LSO. Whether on-site loss of Cav1.3, i.e., in LSO neurons, contributes to the recently described malformation of the LSO needs to be determined by using tissue-specific Cav1.3−/− animals.
Collapse
Affiliation(s)
| | - Désirée Griesemer
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Antonella Pirone
- Institute of Physiology II and Department of Otolaryngology, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany; and
| | - Martina J. Sinnegger-Brauns
- Institute of Pharmacy, Pharmacology and Toxicology, Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Jörg Striessnig
- Institute of Pharmacy, Pharmacology and Toxicology, Center of Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
188
|
Enríquez Denton M, Wienecke J, Zhang M, Hultborn H, Kirkwood PA. Voltage-dependent amplification of synaptic inputs in respiratory motoneurones. J Physiol 2012; 590:3067-90. [PMID: 22495582 PMCID: PMC3406391 DOI: 10.1113/jphysiol.2011.225789] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 04/05/2012] [Indexed: 11/08/2022] Open
Abstract
The role of persistent inward currents (PICs) in cat respiratory motoneurones (phrenic inspiratory and thoracic expiratory) was investigated by studying the voltage-dependent amplification of central respiratory drive potentials (CRDPs), recorded intracellularly, with action potentials blocked with the local anaesthetic derivative, QX-314. Decerebrate unanaesthetized or barbiturate-anaesthetized preparations were used. In expiratory motoneurones, plateau potentials were observed in the decerebrates, but not under anaesthesia. For phrenic motoneurones, no plateau potentials were observed in either state (except in one motoneurone after the abolition of the respiratory drive by means of a medullary lesion), but all motoneurones showed voltage-dependent amplification of the CRDPs, over a wide range of membrane potentials, too wide to result mainly from PIC activation. The measurements of the amplification were restricted to the phase of excitation, thus excluding the inhibitory phase. Amplification was found to be greatest for the smallest CRDPs in the lowest resistance motoneurones and was reduced or abolished following intracellular injection of the NMDA channel blocker, MK-801. Plateau potentials were readily evoked in non-phrenic cervical motoneurones in the same (decerebrate) preparations. We conclude that the voltage-dependent amplification of synaptic excitation in phrenic motoneurones is mainly the result of NMDA channel modulation rather than the activation of Ca2+ channel mediated PICs, despite phrenic motoneurones being strongly immunohistochemically labelled for CaV1.3 channels. The differential PIC activation in different motoneurones, all of which are CaV1.3 positive, leads us to postulate that the descending modulation of PICs is more selective than has hitherto been believed.
Collapse
Affiliation(s)
- M Enríquez Denton
- Sobell Department for Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | | | | | | |
Collapse
|
189
|
Lieb A, Scharinger A, Sartori S, Sinnegger-Brauns MJ, Striessnig J. Structural determinants of CaV1.3 L-type calcium channel gating. Channels (Austin) 2012; 6:197-205. [PMID: 22760075 PMCID: PMC3431584 DOI: 10.4161/chan.21002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A C-terminal modulatory domain (CTM) tightly regulates the biophysical properties of Ca(v)1.3 L-type Ca(2+) channels, in particular the voltage dependence of activation (V(0.5)) and Ca(2+) dependent inactivation (CDI). A functional CTM is present in the long C-terminus of human and mouse Ca(v)1.3 (Ca(v)1.3(L)), but not in a rat long cDNA clone isolated from superior cervical ganglia neurons (rCa(v)1.3(scg)). We therefore addressed the question if this represents a species-difference and compared the biophysical properties of rCa(v)1.3(scg) with a rat cDNA isolated from rat pancreas (rCa(v)1.3(L)). When expressed in tsA-201 cells under identical experimental conditions rCa(v)1.3(L) exhibited Ca(2+) current properties indistinguishable from human and mouse Ca(v)1.3(L), compatible with the presence of a functional CTM. In contrast, rCa(v)1.3(scg) showed gating properties similar to human short splice variants lacking a CTM. rCa(v)1.3(scg) differs from rCa(v)1.3(L) at three single amino acid (aa) positions, one alternative spliced exon (exon31), and a N-terminal polymethionine stretch with two additional lysines. Two aa (S244, A2075) in rCa(v)1.3(scg) explained most of the functional differences to rCa(v)1.3(L). Their mutation to the corresponding residues in rCa(v)1.3(L) (G244, V2075) revealed that both contributed to the more negative V 0.5, but caused opposite effects on CDI. A2075 (located within a region forming the CTM) additionally permitted higher channel open probability. The cooperative action in the double-mutant restored gating properties similar to rCa(v)1.3(L). We found no evidence for transcripts containing one of the single rCa(v)1.3(scg) mutations in rat superior cervical ganglion preparations. However, the rCa(v)1.3(scg) variant provided interesting insight into the structural machinery involved in Ca(v)1.3 gating.
Collapse
Affiliation(s)
- Andreas Lieb
- Institute of Pharmacy and Center for Molecular Biosciences, University of Innsbruck, Austria
| | | | | | | | | |
Collapse
|
190
|
Tuckwell HC. Quantitative aspects of L-type Ca2+ currents. Prog Neurobiol 2012; 96:1-31. [DOI: 10.1016/j.pneurobio.2011.09.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 09/16/2011] [Accepted: 09/23/2011] [Indexed: 12/24/2022]
|
191
|
Yamamoto S, Tanabe M, Ono H. N- and L-Type Voltage-Dependent Ca 2+ Channels Contribute to the Generation of After-Discharges in the Spinal Ventral Root After Cessation of Noxious Mechanical Stimulation. J Pharmacol Sci 2012; 119:82-90. [DOI: 10.1254/jphs.12035fp] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
192
|
Voltage-Gated Ca2+ Channel Mediated Ca2+ Influx in Epileptogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:1219-47. [DOI: 10.1007/978-94-007-2888-2_55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
193
|
Christel C, Lee A. Ca2+-dependent modulation of voltage-gated Ca2+ channels. Biochim Biophys Acta Gen Subj 2011; 1820:1243-52. [PMID: 22223119 DOI: 10.1016/j.bbagen.2011.12.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Revised: 12/15/2011] [Accepted: 12/16/2011] [Indexed: 01/06/2023]
Abstract
BACKGROUND Voltage-gated (Cav) Ca2+ channels are multi-subunit complexes that play diverse roles in a wide variety of tissues. A fundamental mechanism controlling Cav channel function involves the Ca2+ ions that permeate the channel pore. Ca2+ influx through Cav channels mediates feedback regulation to the channel that is both negative (Ca2+-dependent inactivation, CDI) and positive (Ca2+-dependent facilitation, CDF). SCOPE OF REVIEW This review highlights general mechanisms of CDI and CDF with an emphasis on how these processes have been studied electrophysiologically in native and heterologous expression systems. MAJOR CONCLUSIONS Electrophysiological analyses have led to detailed insights into the mechanisms and prevalence of CDI and CDF as Cav channel regulatory mechanisms. All Cav channel family members undergo some form of Ca2+-dependent feedback that relies on CaM or a related Ca2+ binding protein. Tremendous progress has been made in characterizing the role of CaM in CDI and CDF. Yet, what contributes to the heterogeneity of CDI/CDF in various cell-types and how Ca2+-dependent regulation of Cav channels controls Ca2+ signaling remain largely unexplored. GENERAL SIGNIFICANCE Ca2+ influx through Cav channels regulates diverse physiological events including excitation-contraction coupling in muscle, neurotransmitter and hormone release, and Ca2+-dependent gene transcription. Therefore, the mechanisms that regulate channels, such as CDI and CDF, can have a large impact on the signaling potential of excitable cells in various physiological contexts. This article is part of a Special Issue entitled Biochemical, biophysical and genetic approaches to intracellular calcium signaling.
Collapse
Affiliation(s)
- Carl Christel
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
194
|
Ryglewski S, Lance K, Levine RB, Duch C. Ca(v)2 channels mediate low and high voltage-activated calcium currents in Drosophila motoneurons. J Physiol 2011; 590:809-25. [PMID: 22183725 DOI: 10.1113/jphysiol.2011.222836] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Different blends of membrane currents underlie distinct functions of neurons in the brain. A major step towards understanding neuronal function, therefore, is to identify the genes that encode different ionic currents. This study combined in situ patch clamp recordings of somatodendritic calcium currents in an identified adult Drosophila motoneuron with targeted genetic manipulation. Voltage clamp recordings revealed transient low voltage-activated (LVA) currents with activation between –60 mV and –70 mV as well as high voltage-activated (HVA) current with an activation voltage around –30 mV. LVA could be fully inactivated by prepulses to –50 mV and was partially amiloride sensitive. Recordings from newly generated mutant flies demonstrated that DmαG (Ca(v)3 homolog) encoded the amiloride-sensitive portion of the transient LVA calcium current. We further demonstrated that the Ca(v)2 homolog, Dmca1A, mediated the amiloride-insensitive component of LVA current. This novel role of Ca(v)2 channels was substantiated by patch clamp recordings from conditional mutants, RNAi knock-downs, and following Dmca1A overexpression. In addition, we show that Dmca1A underlies the HVA somatodendritic calcium currents in vivo. Therefore, the Drosophila Ca(v)2 homolog, Dmca1A, underlies HVA and LVA somatodendritic calcium currents in the same neuron. Interestingly, DmαG is required for regulating LVA and HVA derived from Dmca1A in vivo. In summary, each vertebrate gene family for voltage-gated calcium channels is represented by a single gene in Drosophila, namely Dmca1D (Ca(v)1), Dmca1A (Ca(v)2) and DmαG (Ca(v)3), but the commonly held view that LVA calcium currents are usually mediated by Ca(v)3 rather than Ca(v)2 channels may require reconsideration.
Collapse
Affiliation(s)
- Stefanie Ryglewski
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.
| | | | | | | |
Collapse
|
195
|
Hurley MJ, Dexter DT. Voltage-gated calcium channels and Parkinson's disease. Pharmacol Ther 2011; 133:324-33. [PMID: 22133841 DOI: 10.1016/j.pharmthera.2011.11.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 11/15/2011] [Indexed: 12/27/2022]
Abstract
A complex interaction of environmental, genetic and epigenetic factors combine with ageing to cause the most prevalent of movement disorders Parkinson's disease. Current pharmacological treatments only tackle the symptoms and do not stop progression of the disease or reverse the neurodegenerative process. While some incidences of Parkinson's disease arise through heritable genetic defects, the cause of the majority of cases remains unknown. Likewise, why some neuronal populations are more susceptible to neurodegeneration than others is not clear, but as the molecular pathways responsible for the process of cell death are unravelled, it is increasingly apparent that disrupted cellular energy metabolism plays a central role. Precise control of cellular calcium concentrations is crucial for maintenance of energy homeostasis. Recently, differential cellular expression of neuronal voltage-gated calcium channel (Ca(V)) isoforms has been implicated in the susceptibility of vulnerable neurons to neurodegeneration in Parkinson's disease. Ca(V) channels are also involved in the synaptic plasticity response to the denervation that occurs in Parkinson's disease and following chronic treatment with anti-parkinsonian drugs. This review will examine the putative role neuronal Ca(V) channels have in the pathogenesis and treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Michael J Hurley
- Centre for Neuroscience, Department of Medicine, Imperial College, London W12 ONN, United Kingdom.
| | | |
Collapse
|
196
|
Wang J, Thio SS, Yang SS, Yu D, Yu CY, Wong YP, Liao P, Li S, Soong TW. Splice Variant Specific Modulation of Ca
V
1.2 Calcium Channel by Galectin-1 Regulates Arterial Constriction. Circ Res 2011; 109:1250-8. [DOI: 10.1161/circresaha.111.248849] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
Ca
V
1.2 channels are essential for excitation–contraction coupling in the cardiovascular system, and alternative splicing optimizes its role. Galectin-1 (Gal-1) has been reported to regulate vascular smooth muscle cell (VSMC) function and play a role in pulmonary hypertension. We have identified Gal-1 multiple times in yeast 2-hybrid assays using the Ca
V
1.2 I–II loop as bait.
Objective:
Our hypothesis is that Gal-1 interacts directly with Ca
V
1.2 channel at the I–II loop to affect arterial constriction.
Methods and Results:
Unexpectedly, Gal-1 was found to selectively bind to the I–II loop only in the absence of alternatively spliced exon 9*. We found that the current densities of Ca
V
1.2
Δ9*
channels were significantly inhibited as a result of decreased functional surface expression due to the binding of Gal-1 at the export signal located on the C-terminus of exon 9. Moreover, the suppression of Gal-1 expression by siRNA in rat A7r5 and isolated VSMCs produced the opposite effect of increased
I
Ca,L
. The physiological significance of Gal-1 mediated splice variant-specific inhibition of Ca
V
1.2 channels was demonstrated in organ bath culture where rat MAs were reversibly permeabilized with Gal-1 siRNA and the arterial wall exhibited increased K
+
-induced constriction.
Conclusion:
The above data indicated that Gal-1 regulates
I
Ca,L
via decreasing the functional surface expression of Ca
V
1.2 channels in a splice variant selective manner and such a mechanism may play a role in modulating vascular constriction.
Collapse
Affiliation(s)
- Juejin Wang
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Sharon S.C. Thio
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Sophia S.H. Yang
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Dejie Yu
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Chye Yun Yu
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Yuk Peng Wong
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Ping Liao
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Shengnan Li
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| | - Tuck Wah Soong
- From the Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (J.W., D.Y., Y.P.W., T.W.S.); National Neuroscience Institute, Singapore (J.W., S.S.C.T., S.H.Y., C.Y.Y., P.L., T.W.S.); and Department of Pharmacology, Nanjing Medical University, Nanjing, China (J.W., S.L.)
| |
Collapse
|
197
|
Tan GMY, Yu D, Wang J, Soong TW. Alternative splicing at C terminus of Ca(V)1.4 calcium channel modulates calcium-dependent inactivation, activation potential, and current density. J Biol Chem 2011; 287:832-47. [PMID: 22069316 DOI: 10.1074/jbc.m111.268722] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Ca(V)1.4 voltage-gated calcium channel is predominantly expressed in the retina, and mutations to this channel have been associated with human congenital stationary night blindness type-2. The L-type Ca(V)1.4 channel displays distinct properties such as absence of calcium-dependent inactivation (CDI) and slow voltage-dependent inactivation (VDI) due to the presence of an autoinhibitory domain (inhibitor of CDI) in the distal C terminus. We hypothesized that native Ca(V)1.4 is subjected to extensive alternative splicing, much like the other voltage-gated calcium channels, and employed the transcript scanning method to identify alternatively spliced exons within the Ca(V)1.4 transcripts isolated from the human retina. In total, we identified 19 alternative splice variations, of which 16 variations have not been previously reported. Characterization of the C terminus alternatively spliced exons using whole-cell patch clamp electrophysiology revealed a splice variant that exhibits robust CDI. This splice variant arose from the splicing of a novel alternate exon (43*) that can be found in 13.6% of the full-length transcripts screened. Inclusion of exon 43* inserts a stop codon that truncates half the C terminus. The Ca(V)1.4 43* channel exhibited robust CDI, a larger current density, a hyperpolarized shift in activation potential by ∼10 mV, and a slower VDI. Through deletional experiments, we showed that the inhibitor of CDI was responsible for modulating channel activation and VDI, in addition to CDI. Calcium currents in the photoreceptors were observed to exhibit CDI and are more negatively activated as compared with currents elicited from heterologously expressed full-length Ca(V)1.4. Naturally occurring alternative splice variants may in part contribute to the properties of the native Ca(V)1.4 channels.
Collapse
Affiliation(s)
- Gregory Ming Yeong Tan
- Ion Channel and Transporter Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456
| | | | | | | |
Collapse
|
198
|
CaV2.1 voltage activated calcium channels and synaptic transmission in familial hemiplegic migraine pathogenesis. ACTA ACUST UNITED AC 2011; 106:12-22. [PMID: 22074995 DOI: 10.1016/j.jphysparis.2011.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 10/12/2011] [Accepted: 10/17/2011] [Indexed: 12/28/2022]
Abstract
Studies on the genetic forms of epilepsy, chronic pain, and migraine caused by mutations in ion channels have given crucial insights into the molecular mechanisms, pathogenesis, and therapeutic approaches to complex neurological disorders. In this review we focus on the role of mutated CaV2.1 (i.e., P/Q-type) voltage-activated Ca2+ channels, and on the ultimate consequences that mutations causing familial hemiplegic migraine type-1 (FHM1) have in neurotransmitter release. Transgenic mice harboring the human pathogenic FHM1 mutation R192Q or S218L (KI) have been used as models to study neurotransmission at several central and peripheral synapses. FHM1 KI mice are a powerful tool to explore presynaptic regulation associated with expression of CaV2.1 channels. Mutated CaV2.1 channels activate at more hyperpolarizing potentials and lead to a gain-of-function in synaptic transmission. This gain-of-function might underlie alterations in the excitatory/ inhibitory balance of synaptic transmission, favoring a persistent state of hyperexcitability in cortical neurons that would increase the susceptibility for cortical spreading depression (CSD), a mechanism believed to initiate the attacks of migraine with aura.
Collapse
|
199
|
Bock G, Gebhart M, Scharinger A, Jangsangthong W, Busquet P, Poggiani C, Sartori S, Mangoni ME, Sinnegger-Brauns MJ, Herzig S, Striessnig J, Koschak A. Functional properties of a newly identified C-terminal splice variant of Cav1.3 L-type Ca2+ channels. J Biol Chem 2011; 286:42736-42748. [PMID: 21998310 PMCID: PMC3234942 DOI: 10.1074/jbc.m111.269951] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
An intramolecular interaction between a distal (DCRD) and a proximal regulatory domain (PCRD) within the C terminus of long Ca(v)1.3 L-type Ca(2+) channels (Ca(v)1.3(L)) is a major determinant of their voltage- and Ca(2+)-dependent gating kinetics. Removal of these regulatory domains by alternative splicing generates Ca(v)1.3(42A) channels that activate at a more negative voltage range and exhibit more pronounced Ca(2+)-dependent inactivation. Here we describe the discovery of a novel short splice variant (Ca(v)1.3(43S)) that is expressed at high levels in the brain but not in the heart. It lacks the DCRD but, in contrast to Ca(v)1.3(42A), still contains PCRD. When expressed together with α2δ1 and β3 subunits in tsA-201 cells, Ca(v)1.3(43S) also activated at more negative voltages like Ca(v)1.3(42A) but Ca(2+)-dependent inactivation was less pronounced. Single channel recordings revealed much higher channel open probabilities for both short splice variants as compared with Ca(v)1.3(L). The presence of the proximal C terminus in Ca(v)1.3(43S) channels preserved their modulation by distal C terminus-containing Ca(v)1.3- and Ca(v)1.2-derived C-terminal peptides. Removal of the C-terminal modulation by alternative splicing also induced a faster decay of Ca(2+) influx during electrical activities mimicking trains of neuronal action potentials. Our findings extend the spectrum of functionally diverse Ca(v)1.3 L-type channels produced by tissue-specific alternative splicing. This diversity may help to fine tune Ca(2+) channel signaling and, in the case of short variants lacking a functional C-terminal modulation, prevent excessive Ca(2+) accumulation during burst firing in neurons. This may be especially important in neurons that are affected by Ca(2+)-induced neurodegenerative processes.
Collapse
Affiliation(s)
- Gabriella Bock
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | - Mathias Gebhart
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | - Anja Scharinger
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | - Wanchana Jangsangthong
- Department of Pharmacology and Center for Molecular Medicine, University of Cologne, Gleueler Strasse 24 and Robert-Koch-Strasse 21, D-50931 Cologne, Germany
| | - Perrine Busquet
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | - Chiara Poggiani
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | - Simone Sartori
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | - Matteo E Mangoni
- Département de Physiologie, CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France; INSERM, U661, F-34000 Montpellier, France; Universités de Montpellier 1 & 2, UMR-5203, F-34000 Montpellier, France; INSERM, U637, Montpellier, France
| | - Martina J Sinnegger-Brauns
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria
| | - Stefan Herzig
- Department of Pharmacology and Center for Molecular Medicine, University of Cologne, Gleueler Strasse 24 and Robert-Koch-Strasse 21, D-50931 Cologne, Germany
| | - Jörg Striessnig
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria.
| | - Alexandra Koschak
- Institute of Pharmacy, Pharmacology and Toxicology and Center of Molecular Biosciences Innsbruck, Peter-Mayr-Strasse 1/I, A-6020 Innsbruck, Austria.
| |
Collapse
|
200
|
Tan BZ, Jiang F, Tan MY, Yu D, Huang H, Shen Y, Soong TW. Functional characterization of alternative splicing in the C terminus of L-type CaV1.3 channels. J Biol Chem 2011; 286:42725-42735. [PMID: 21998309 DOI: 10.1074/jbc.m111.265207] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ca(V)1.3 channels are unique among the high voltage-activated Ca(2+) channel family because they activate at the most negative potentials and display very rapid calcium-dependent inactivation. Both properties are of crucial importance in neurons of the suprachiasmatic nucleus and substantia nigra, where the influx of Ca(2+) ions at subthreshold membrane voltages supports pacemaking function. Previously, alternative splicing in the Ca(V)1.3 C terminus gives rise to a long (Ca(V)1.3(42)) and a short form (Ca(V)1.3(42A)), resulting in a pronounced activation at more negative voltages and faster inactivation in the latter. It was further shown that the C-terminal modulator in the Ca(V)1.3(42) isoforms modulates calmodulin binding to the IQ domain. Using splice variant-specific antibodies, we determined that protein localization of both splice variants in different brain regions were similar. Using the transcript-scanning method, we further identified alternative splicing at four loci in the C terminus of Ca(V)1.3 channels. Alternative splicing of exon 41 removes the IQ motif, resulting in a truncated Ca(V)1.3 protein with diminished inactivation. Splicing of exon 43 causes a frameshift and exhibits a robust inactivation of similar intensity to Ca(V)1.3(42A). Alternative splicing of exons 44 and 48 are in-frame, altering interaction of the distal modulator with the IQ domain and tapering inactivation slightly. Thus, alternative splicing in the C terminus of Ca(V)1.3 channels modulates its electrophysiological properties, which could in turn alter neuronal firing properties and functions.
Collapse
Affiliation(s)
- Bao Zhen Tan
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore 117597; NUS Graduate School for Integrative Engineering and Science, National University of Singapore, Singapore 117597
| | - Fengli Jiang
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore 117597
| | - Ming Yeong Tan
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore 117597
| | - Dejie Yu
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore 117597
| | - Hua Huang
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore 117597; NUS Graduate School for Integrative Engineering and Science, National University of Singapore, Singapore 117597
| | - Yiru Shen
- National Neuroscience Institute, Singapore 308433
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore 117597; NUS Graduate School for Integrative Engineering and Science, National University of Singapore, Singapore 117597; National Neuroscience Institute, Singapore 308433.
| |
Collapse
|