101
|
Ding J, Zhang JR, Wang Y, Li CL, Lu D, Guan SM, Chen J. Effects of a non-selective TRPC channel blocker, SKF-96365, on melittin-induced spontaneous persistent nociception and inflammatory pain hypersensitivity. Neurosci Bull 2012; 28:173-81. [PMID: 22466128 DOI: 10.1007/s12264-012-1213-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
OBJECTIVE Melittin is the main peptide in bee venom and causes both persistent spontaneous nociception and pain hypersensitivity. Our recent studies indicated that both transient receptor potential (TRP) vanilloid receptor 1 (TRPV1) and canonical TRPs (TRPCs) are involved in mediating the melittin-induced activation of different subpopulations of primary nociceptive cells. Here, we further determined whether TRPC channels are involved in melittin-induced inflammatory nociceptive responses in behavioral assays. METHODS The anti-nociceptive and anti-hyperalgesic effects of localized peripheral administration of three doses of the non-selective TRPC antagonist, SKF-96365 (1-{β-[3-(4-methoxyphenyl)propoxy]-4-methoxyphenyl}-1H-imidazole hydrochloride), were evaluated in melittin tests. Pain-related behaviors were rated by counting the number of paw flinches, and measuring paw withdrawal thermal latency (s) and paw withdrawl mechanical threshold (g), over a 1-h time-course. RESULTS Localized peripheral SKF-96365 given before melittin prevented, and given after melittin significantly suppressed, the melittin-evoked persistent spontaneous nociception. Pre-blockade and post-suppression of activation of primary nociceptive activity resulted in decreased hypersensitivity to both thermal and mechanical stimuli applied to the primary injury site of the ipsilateral hindpaw, despite dose-effect differences between thermal and mechanical hyperalgesia. However, local administration of SKF-96365 into the contralateral hindpaw had no significant effect on any pain-associated behaviors. In addition, SKF-96365 had no effect on baseline threshold for either thermal or mechanical sensitivity under normal conditions. CONCLUSION Besides TRPV1, SKF-96365-sensitive TRPC channels might also be involved in the pathophysiological processing of melittin-induced inflammatory pain and hypersensitivity. Therapeutically, SKF-96365 is equally effective in preventing primary thermal and mechanical hyperalgesia as well as persistent spontaneous nociception. However, this drug is likely to be more effective in the relief of thermal hyperalgesia than mechanical hyperalgesia when applied 5 min after establishment of primary afferent activation.
Collapse
Affiliation(s)
- Jing Ding
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | | | | | | | | | | | | |
Collapse
|
102
|
Nagy GA, Botond G, Borhegyi Z, Plummer NW, Freund TF, Hájos N. DAG-sensitive and Ca(2+) permeable TRPC6 channels are expressed in dentate granule cells and interneurons in the hippocampal formation. Hippocampus 2012. [PMID: 23193081 DOI: 10.1002/hipo.22081] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Members of the transient receptor potential (TRP) cation channel family play important roles in several neuronal functions. To understand the precise role of these channels in information processing, their presence on neuronal elements must be revealed. In this study, we investigated the localization of TRPC6 channels in the adult hippocampal formation. Immunostainings with a specific antibody, which was validated in Trpc6 knockout mice, showed that in the dentate gyrus, TRPC6 channels are strongly expressed in granule cells. Immunogold staining revealing the subcellular localization of TRPC6 channels clarified that these proteins were predominantly present on the membrane surface of the dendritic shafts of dentate granule cells, and also in their axons, often associated with intracellular membrane cisternae. In addition, TRPC6 channels could be observed in the dendrites of some interneurons. Double immunofluorescent staining showed that TRPC6 channels were present in the dendrites of hilar interneurons and hippocampal interneurons with horizontal dendrites in the stratum oriens expressing mGlu1a receptors, whereas parvalbumin immunoreactivity was revealed in TRPC6-expressing dendrites with radial appearance in the stratum radiatum. Electron microscopy showed that the immunogold particles depicting TRPC6 channels were located on the surface membranes of the interneuron dendrites. Our results suggest that TRPC6 channels are in a key position to alter the information entry into the trisynaptic loop of the hippocampal formation from the entorhinal cortex, and to control the function of both feed-forward and feed-back inhibitory circuits in this brain region. © 2012 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gergő A Nagy
- Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1450 Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
103
|
Li H, Huang J, Du W, Jia C, Yao H, Wang Y. TRPC6 inhibited NMDA receptor activities and protected neurons from ischemic excitotoxicity. J Neurochem 2012; 123:1010-8. [DOI: 10.1111/jnc.12045] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 09/26/2012] [Accepted: 09/27/2012] [Indexed: 11/27/2022]
Affiliation(s)
- Hongyu Li
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
- The Graduate School, Chinese Academy of Science; Shanghai China
| | - Junbo Huang
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
- The Graduate School, Chinese Academy of Science; Shanghai China
| | - Wanlu Du
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| | - Caixia Jia
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| | - Hailan Yao
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction; Institute of Neuroscience, SIBS, State Key Laboratory of Neuroscience; Shanghai China
| |
Collapse
|
104
|
TRPC5 channel is the mediator of neurotrophin-3 in regulating dendritic growth via CaMKIIα in rat hippocampal neurons. J Neurosci 2012; 32:9383-95. [PMID: 22764246 DOI: 10.1523/jneurosci.6363-11.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Neurotrophin-3 (NT-3) plays numerous important roles in the CNS and the elevation of intracellular Ca(2+) ([Ca(2+)](i)) is critical for these functions of NT-3. However, the mechanism by which NT-3 induces [Ca(2+)](i) elevation remains largely unknown. Here, we found that transient receptor potential canonical (TRPC) 5 protein and TrkC, the NT-3 receptor, exhibited a similar temporal expression in rat hippocampus and cellular colocalization in hippocampal neurons. Stimulation of the neurons by NT-3 induced a nonselective cation conductance and PLCγ-dependent [Ca(2+)](i) elevation, which were both blocked when TRPC5, but not TRPC6 channels, were inhibited. Moreover, the Ca(2+) influx through TRPC5 induced by NT-3 inhibited the neuronal dendritic growth through activation of calmodulin-dependent kinase (CaMK) IIα. In contrast, the Ca(2+) influx through TRPC6 induced by NT-4 promoted the dendritic growth. Thus, TRPC5 acts as a novel and specific mediator for NT-3 to regulate dendrite development through CaMKIIα.
Collapse
|
105
|
Fan Q, Huang WB, Zhang XL. TRPC6: an underlying target for human glaucoma. Int J Ophthalmol 2012; 5:523-6. [PMID: 22937518 DOI: 10.3980/j.issn.2222-3959.2012.04.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 07/20/2012] [Indexed: 11/02/2022] Open
Abstract
Glaucoma is one of the leading causes of visual impairment and blindness worldwide. Of known risk factors for glaucoma, an increased in intraocular pressure is most highly correlated with glaucomatous damage. Irrespective of the cause, apoptosis of the retinal ganglion cells is the eventual outcome. It is widely accepted that glaucoma is a neurodegenerative disease that is strongly correlated with central nervous system disorders, such as Alzheimer's disease. These two disorders also share some similarities in pathogenic mechanisms. Recent studies suggest that the transient receptor potential canonical 6 channel could work together with brain-derived neurotrophic factor to promote neuron survival in brain and retina. In this study, we propose that transient receptor potential canonical 6 may contribute to the pathogenesis of human glaucoma and become a potential therapeutic target.
Collapse
Affiliation(s)
- Qian Fan
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | | | | |
Collapse
|
106
|
Xu P, Xu J, Li Z, Yang Z. Expression of TRPC6 in renal cortex and hippocampus of mouse during postnatal development. PLoS One 2012; 7:e38503. [PMID: 22701654 PMCID: PMC3368853 DOI: 10.1371/journal.pone.0038503] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 05/10/2012] [Indexed: 11/21/2022] Open
Abstract
TRPC6, a member of the TRPC family, attracts much attention from the public because of its relationship with the disease. In both the brain and kidney, TRPC6 serves a variety of functions. The aim of the present study was to observe the expression and effects of TRPC6 in renal cortex and hippocampus during early postnatal development of the mouse. In the present study, immunohistochemistry and Western blotting were used to detect the expression of TRPC6 in the mouse kidney and hippocampus of postnatal day 1, 3, 5, 7, 14, 21, 28 and 49 (P1, P3, P5, P7, P14, P21, P28 and P49). Results showed that the expression of TRPC6 was increased in the mouse hippocampus, and there was a significant increase between P7 and P14 during the postnatal development. Meanwhile, the expression of TRPC6 was also detected in glomerulus and tubules, and a decreased expression was found during postnatal maturation of mouse renal cortex. From these in vivo experiments, we concluded that the expression of TRPC6 was active in the developing mouse kidney cortex, and followed a loss of expression with the development of kidney. Meanwhile, an increased expression was found in the hippocampus with the development. Together, these data suggested that the developmental changes in TRPC6 expression might be required for proper postnatal kidney cortex development, and played a critical role in the hippocampus during development, which formed the basis for understanding the nephrogenesis and neurogenesis in mice and provided a practically useful knowledge to the clinical and related research.
Collapse
Affiliation(s)
- Pengjuan Xu
- College of Medicine, Nankai University, Tianjin, China
| | - Jing Xu
- College of Medicine, Nankai University, Tianjin, China
| | - Zhigui Li
- College of Medicine, Nankai University, Tianjin, China
| | - Zhuo Yang
- College of Medicine, Nankai University, Tianjin, China
- * E-mail:
| |
Collapse
|
107
|
Kumar S, Chakraborty S, Barbosa C, Brustovetsky T, Brustovetsky N, Obukhov AG. Mechanisms controlling neurite outgrowth in a pheochromocytoma cell line: the role of TRPC channels. J Cell Physiol 2012; 227:1408-19. [PMID: 21618530 DOI: 10.1002/jcp.22855] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transient Receptor Potential Canonical (TRPC) channels are implicated in modulating neurite outgrowth. The expression pattern of TRPCs changes significantly during brain development, suggesting that fine-tuning TRPC expression may be important for orchestrating neuritogenesis. To study how alterations in the TRPC expression pattern affect neurite outgrowth, we used nerve growth factor (NGF)-differentiated rat pheochromocytoma 12 (PC12) cells, a model system for neuritogenesis. In PC12 cells, NGF markedly up-regulated TRPC1 and TRPC6 expression, but down-regulated TRPC5 expression while promoting neurite outgrowth. Overexpression of TRPC1 augmented, whereas TRPC5 overexpression decelerated NGF-induced neurite outgrowth. Conversely, shRNA-mediated knockdown of TRPC1 decreased, whereas shRNA-mediated knockdown of TRPC5 increased NGF-induced neurite extension. Endogenous TRPC1 attenuated the anti-neuritogenic effect of overexpressed TRPC5 in part by forming the heteromeric TRPC1-TRPC5 channels. Previous reports suggested that TRPC6 may facilitate neurite outgrowth. However, we found that TRPC6 overexpression slowed down neuritogenesis, whereas dominant negative TRPC6 (DN-TRPC6) facilitated neurite outgrowth in NGF-differentiated PC12 cells. Consistent with these findings, hyperforin, a neurite outgrowth promoting factor, decreased TRPC6 expression in NGF-differentiated PC12 cells. Using pharmacological and molecular biological approaches, we determined that NGF up-regulated TRPC1 and TRPC6 expression via a p75(NTR)-IKK(2)-dependent pathway that did not involve TrkA receptor signaling in PC12 cells. Similarly, NGF up-regulated TRPC1 and TRPC6 via an IKK(2) dependent pathway in primary cultured hippocampal neurons. Thus, our data suggest that a balance of TRPC1, TRPC5, and TRPC6 expression determines neurite extension rate in neural cells, with TRPC6 emerging as an NGF-dependent "molecular damper" maintaining a submaximal velocity of neurite extension.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Cellular and Integrative Physiology, IUPUI-Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|
108
|
Roedding AS, Gao AF, Au-Yeung W, Scarcelli T, Li PP, Warsh JJ. Effect of oxidative stress on TRPM2 and TRPC3 channels in B lymphoblast cells in bipolar disorder. Bipolar Disord 2012; 14:151-61. [PMID: 22420591 DOI: 10.1111/j.1399-5618.2012.01003.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Recent findings implicate the calcium-permeable nonselective ion channels transient receptor potential (TRP) melastatin subtype 2 (TRPM2) and canonical subtype 3 (TRPC3) in the pathogenesis of bipolar disorder (BD). These channels are involved in calcium and oxidative stress signaling, both of which are disrupted in BD. Thus, we sought to determine if these channels are differentially affected by oxidative stress in cell lines of BD patient origin. METHODS B lymphoblast cell lines (BLCLs) from bipolar I disorder (BD-I) patients (n = 6) and healthy controls (n = 5) were challenged with the oxidative stressor rotenone (2.5 μM and 10 μM) or vehicle for acute (24 hours) and chronic (four days) intervals. Cell viability was measured using propidium iodide, while TRPM2- and TRPC3-mediated calcium fluxes were measured in the presence of their respective activators (H(2) O(2) and 1-oleoyl-2-acetyl-sn-glycerol) using Fluo-4. Changes in TRPM2 and TRPC3 expression levels were determined by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and Western blotting. RESULTS Cell viability decreased with increasing dose and duration of rotenone treatment, with BD-I patient BLCLs more susceptible than controls acutely (p < 0.001). A dose-dependent decrease in TRPC3 protein expression occurred after chronic (24%, p = 0.008) but not acute rotenone treatment. Interestingly, H(2) O(2) -provoked TRPM2-dependent calcium fluxes revealed an interaction between the effects of stressor addition and diagnostic subject group (p = 0.003). CONCLUSIONS These data support an important role for TRPM2 and TRPC3 in sensing and responding to oxidative stress and in transducing oxidative stress signaling to intracellular calcium homeostasis and cellular stress responses, all of which have been implicated in the pathophysiology of BD.
Collapse
Affiliation(s)
- Angela S Roedding
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
109
|
CRP1, a protein localized in filopodia of growth cones, is involved in dendritic growth. J Neurosci 2012; 31:16781-91. [PMID: 22090504 DOI: 10.1523/jneurosci.2595-11.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cysteine-rich protein (CRP) family is a subgroup of LIM domain proteins. CRP1, which cross-links actin filaments to make actin bundles, is the only CRP family member expressed in the CNS with little known about its function in nerve cells. Here, we report that CRP1 colocalizes with actin in the filopodia of growth cones in cultured rat hippocampal neurons. Knockdown of CRP1 expression by short hairpin RNA interference results in inhibition of filopodia formation and dendritic growth in neurons. Overexpression of CRP1 increases filopodia formation and neurite branching, which require its actin-bundling activity. Expression of CRP1 with a constitutively active form of Cdc42, a GTPase involved in filopodia formation, increases filopodia formation in COS-7 cells, suggesting cooperation between the two proteins. Moreover, we demonstrate that neuronal activity upregulates CRP1 expression in hippocampal neurons via Ca²⁺ influx after depolarization. Ca²⁺/calmodulin-dependent protein kinase IV (CaMKIV) and cAMP response element binding protein mediate the Ca²⁺-induced upregulation of CRP1 expression. Furthermore, CRP1 is required for the dendritic growth induced by Ca²⁺ influx or CaMKIV. Together, these data are the first to demonstrate a role for CRP1 in dendritic growth.
Collapse
|
110
|
Abstract
The Transient receptor potential (TRP) family of cation channels is a large protein family, which is mainly structurally uniform. Proteins consist typically of six transmembrane domains and mostly four subunits are necessary to form a functional channel. Apart from this, TRP channels display a wide variety of activation mechanisms (ligand binding, G-protein coupled receptor dependent, physical stimuli such as temperature, pressure, etc.) and ion selectivity profiles (from highly Ca(2+) selective to non-selective for cations). They have been described now in almost every tissue of the body, including peripheral and central neurons. Especially in the sensory nervous system the role of several TRP channels is already described on a detailed level. This review summarizes data that is currently available on their role in the central nervous system. TRP channels are involved in neurogenesis and brain development, synaptic transmission and they play a key role in the development of several neurological diseases.
Collapse
|
111
|
|
112
|
Nagendran T, Hardy LR. Calcium/calmodulin-dependent protein kinase IV mediates distinct features of basal and activity-dependent dendrite complexity. Neuroscience 2011; 199:548-62. [PMID: 21989476 DOI: 10.1016/j.neuroscience.2011.09.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/22/2011] [Accepted: 09/23/2011] [Indexed: 10/17/2022]
Abstract
Intracellular signaling mechanisms translate extracellular signals, such as neuronal activity, into effects on dendrite complexity. Deciphering these mechanisms has considerable impact on understanding how the brain develops and what can go wrong in developmental disorders. How neurons regulate intracellular signaling to control their dendrite morphology remains poorly understood and is likely to be determined at the level of individual neuronal types. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) is a signaling mechanism involved in the regulation of gene expression and dendrite growth. Expression of CaMKIV is developmentally regulated in the cerebral cortex, with highest expression occurring concomitant with the period of extensive dendrite growth and elaboration. Interestingly, cortical neurons heterogeneously expressed CaMKIV in postnatal rat cortices and cortical neurons in vitro. We tested if this differential CaMKIV expression mediated distinct arborization patterns in the dendrites of pyramidal and non-pyramidal neurons. In fact, CaMKIV mediated dendrite complexity via regulation of specific morphological features of the dendrite arbor: branching and elongation, but not primary dendrite formation. We found that small interfering RNA (siRNA) knockdown of CaMKIV decreased basal dendrite complexity indicating that endogenously expressed CaMKIV mediated dendrite complexity. CaMKIV was also required for activity-induced dendrite elaboration. Active CaMKIV expression in cortical neurons increased dendrite elaboration indicating that enzymatic activity was involved. These data indicated neuronal CaMKIV expression was required for basal and activity-induced dendrite complexity. Further, the data presented in this study indicate CaMKIV contributes to the diversity of dendrite arbors via restricted expression and regulation of distinct modes of dendrite elaboration.
Collapse
Affiliation(s)
- T Nagendran
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Health Sciences University, 1120 15th Street, Augusta, GA 30912, USA
| | | |
Collapse
|
113
|
Wu TT, Zhao ZJ, Xu C, Zhang LC. Distribution of TRPC6 in the Cerebrospinal Fluid-Contacting Nucleus of Rat Brain Parenchyma and its Expression in Morphine Dependence and Withdrawal. Neurochem Res 2011; 36:2316-21. [DOI: 10.1007/s11064-011-0556-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2011] [Indexed: 02/02/2023]
|
114
|
Bollimuntha S, Selvaraj S, Singh BB. Emerging roles of canonical TRP channels in neuronal function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:573-93. [PMID: 21290317 DOI: 10.1007/978-94-007-0265-3_31] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ca(2+) signaling in neurons is intimately associated with the regulation of vital physiological processes including growth, survival and differentiation. In neurons, Ca(2+) elicits two major functions. First as a charge carrier, Ca(2+) reveals an indispensable role in information relay via membrane depolarization, exocytosis, and the release of neurotransmitters. Second on a global basis, Ca(2+) acts as a ubiquitous intracellular messenger to modulate neuronal function. Thus, to mediate Ca(2+)-dependent physiological events, neurons engage multiple mode of Ca(2+) entry through a variety of Ca(2+) permeable plasma membrane channels. Here we discuss a subset of specialized Ca(2+)-permeable non-selective TRPC channels and summarize their physiological and pathological role in the context of excitable cells. TRPC channels are predominately expressed in neuronal cells and are activated through complex mechanisms, including second messengers and store depletion. A growing body of evidence suggests a prime contribution of TRPC channels in regulating fundamental neuronal functions. TRPC channels have been shown to be associated with neuronal development, proliferation and differentiation. In addition, TRPC channels have also been suggested to have a potential role in regulating neurosecretion, long term potentiation, and synaptic plasticity. During the past years, numerous seminal discoveries relating TRPC channels to neurons have constantly emphasized on the significant contribution of this group of ion channels in regulating neuronal function. Here we review the major groundbreaking work that has uniquely placed TRPC channels in a pivotal position for governing neuronal Ca(2+) signaling and associated physiological responses.
Collapse
Affiliation(s)
- Sunitha Bollimuntha
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201, USA.
| | | | | |
Collapse
|
115
|
Functional regulation of transient receptor potential canonical 7 by cGMP-dependent protein kinase Iα. Cell Signal 2011; 23:1179-87. [PMID: 21402151 DOI: 10.1016/j.cellsig.2011.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Accepted: 03/06/2011] [Indexed: 11/23/2022]
Abstract
The cGMP/cGMP-dependent protein kinase (cGK) signaling pathway is implicated in the functional regulation of intracellular calcium levels. In the present study, we investigated the regulation of transient receptor potential canonical 7 (TRPC7) by the cGMP/cGK-I pathway. TRPC7 contains three putative cGK phosphorylation sites (Arg-Arg/Lys-Xaa-Ser/Thr). However, the role of cGK-I in the regulation of TRPC7 activity remains unclear. In vitro and in vivo kinase assays have revealed that cGK-Iα phosphorylates mouse TRPC7 but not mouse TRPC3. Site-directed mutagenesis analysis revealed that TRPC7 was phosphorylated by cGK-Iα at threonine 15. Phosphorylation of TRPC7 significantly suppressed carbachol-induced calcium influx and CREB phosphorylation. Furthermore, co-immunoprecipitation assay demonstrated that cGK-Iα interacted with the ankyrin repeat domain in the N terminus of TRPC7. cGK-Iβ also bound to TRPC7, while the type II regulatory subunit of cAMP-dependent protein kinase did not bind. These data indicate that cGK-Iα interacts with and phosphorylates TRPC7, contributing to the quick and accurate regulation of calcium influx and CREB phosphorylation.
Collapse
|
116
|
Transient receptor proteins illuminated: Current views on TRPs and disease. Vet J 2011; 187:153-64. [DOI: 10.1016/j.tvjl.2010.01.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 01/21/2010] [Accepted: 01/25/2010] [Indexed: 11/23/2022]
|
117
|
TRP Channels and Psychiatric Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:987-1009. [DOI: 10.1007/978-94-007-0265-3_51] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
118
|
Evidence for a supportive role of classical transient receptor potential 6 (TRPC6) in the exploration behavior of mice. Physiol Behav 2010; 102:245-50. [PMID: 21059368 DOI: 10.1016/j.physbeh.2010.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 10/11/2010] [Accepted: 11/01/2010] [Indexed: 11/21/2022]
Abstract
Non-selective classical transient receptor potential (TRPC) cation channels share important roles in processes of neuronal development and function. To test the influence of TRPC6 activity on behavior, we developed a TRPC6-deficient (TRPC6(-/-)) mouse model in a BALB/c genetic background. Both, TRPC6(-/-) and wild-type (WT) mice were analyzed first for their general health and reflex status (modified SHIRPA protocol) and then in three different behavioral tests (marble-burying test, square open field and elevated star maze). No abnormalities were detected in the SHIRPA protocol. Most interestingly, TRPC6(-/-) mice showed no significant differences in anxiety in a marble-burying test, but demonstrated reduced exploration in the square open field and the elevated star maze. Therefore, TRPC6 channel activity may play a yet unknown role for exploration behavior.
Collapse
|
119
|
Gees M, Colsoul B, Nilius B. The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol 2010; 2:a003962. [PMID: 20861159 DOI: 10.1101/cshperspect.a003962] [Citation(s) in RCA: 323] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The 28 mammalian members of the super-family of transient receptor potential (TRP) channels are cation channels, mostly permeable to both monovalent and divalent cations, and can be subdivided into six main subfamilies: the TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin), TRPML (mucolipin), and the TRPA (ankyrin) groups. TRP channels are widely expressed in a large number of different tissues and cell types, and their biological roles appear to be equally diverse. In general, considered as polymodal cell sensors, they play a much more diverse role than anticipated. Functionally, TRP channels, when activated, cause cell depolarization, which may trigger a plethora of voltage-dependent ion channels. Upon stimulation, Ca2+ permeable TRP channels generate changes in the intracellular Ca2+ concentration, [Ca2+]i, by Ca2+ entry via the plasma membrane. However, more and more evidence is arising that TRP channels are also located in intracellular organelles and serve as intracellular Ca2+ release channels. This review focuses on three major tasks of TRP channels: (1) the function of TRP channels as Ca2+ entry channels; (2) the electrogenic actions of TRPs; and (3) TRPs as Ca2+ release channels in intracellular organelles.
Collapse
Affiliation(s)
- Maarten Gees
- KU Leuven, Department of Molecular Cell Biology, Laboratory Ion Channel Research, Campus Gasthuisberg, Herestraat 49, bus 802, Leuven, Belgium
| | | | | |
Collapse
|
120
|
Di Stefano G, Casoli T, Platano D, Fattoretti P, Balietti M, Giorgetti B, Bertoni-Freddari C, Lattanzio F, Aicardi G. Differences in gene expression in the hippocampus of aged rats are associated with better long-term memory performance in a passive avoidance test. Rejuvenation Res 2010; 13:224-8. [PMID: 20426624 DOI: 10.1089/rej.2009.0965] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Microarray analysis was used to identify genes differentially expressed in the hippocampus of aged rats showing diverse long-term (3 and 6 h) spatial-associative memory performance in a single-trial inhibitory avoidance task. The transcription of 43 genes (including genes functionally linked to signal transduction, cell growth and differentiation, translation, energy metabolism, and nucleic acid processing) was significantly upregulated in good- versus bad-performing animals, whereas that of 18 genes (including genes functionally linked to transcription, cell growth and differentiation, apoptosis, and protein transport) was significantly downregulated in good- versus bad-performing animals. The differential expression of 14 of these genes was confirmed by real-time polymerase chain reaction.
Collapse
|
121
|
Louhivuori LM, Jansson L, Nordström T, Bart G, Näsman J, Åkerman KE. Selective interference with TRPC3/6 channels disrupts OX1 receptor signalling via NCX and reveals a distinct calcium influx pathway. Cell Calcium 2010; 48:114-23. [DOI: 10.1016/j.ceca.2010.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 07/20/2010] [Accepted: 07/23/2010] [Indexed: 11/15/2022]
|
122
|
Eder C. Ion channels in monocytes and microglia / brain macrophages: Promising therapeutic targets for neurological diseases. J Neuroimmunol 2010; 224:51-5. [DOI: 10.1016/j.jneuroim.2010.05.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 05/04/2010] [Indexed: 10/19/2022]
|
123
|
Ding X, He Z, Zhou K, Cheng J, Yao H, Lu D, Cai R, Jin Y, Dong B, Xu Y, Wang Y. Essential role of TRPC6 channels in G2/M phase transition and development of human glioma. J Natl Cancer Inst 2010; 102:1052-68. [PMID: 20554944 DOI: 10.1093/jnci/djq217] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Patients with glioblastoma multiforme, the most aggressive form of glioma, have a median survival of approximately 12 months. Calcium (Ca(2+)) signaling plays an important role in cell proliferation, and some members of the Ca(2+)-permeable transient receptor potential canonical (TRPC) family of channel proteins have demonstrated a role in the proliferation of many types of cancer cells. In this study, we investigated the role of TRPC6 in cell cycle progression and in the development of human glioma. METHODS TRPC6 protein and mRNA expression were assessed in glioma (n = 33) and normal (n = 17) brain tissues from patients and in human glioma cell lines U251, U87, and T98G. Activation of TRPC6 channels was tested by platelet-derived growth factor-induced Ca(2+) imaging. The effect of inhibiting TRPC6 activity or expression using the dominant-negative mutant TRPC6 (DNC6) or RNA interference, respectively, was tested on cell growth, cell cycle progression, radiosensitization of glioma cells, and development of xenografted human gliomas in a mouse model. The green fluorescent protein (GFP) and wild-type TRPC6 (WTC6) were used as controls. Survival of mice bearing xenografted tumors in the GFP, DNC6, and WTC6 groups (n = 13, 15, and 13, respectively) was compared using Kaplan-Meier analysis. All statistical tests were two-sided. RESULTS Functional TRPC6 was overexpressed in human glioma cells. Inhibition of TRPC6 activity or expression attenuated the increase in intracellular Ca(2+) by platelet-derived growth factor, suppressed cell growth and clonogenic ability, induced cell cycle arrest at the G2/M phase, and enhanced the antiproliferative effect of ionizing radiation. Cyclin-dependent kinase 1 activation and cell division cycle 25 homolog C expression regulated the cell cycle arrest. Inhibition of TRPC6 activity also reduced tumor volume in a subcutaneous mouse model of xenografted human tumors (P = .014 vs GFP; P < .001 vs WTC6) and increased mean survival in mice in an intracranial model (P < .001 vs GFP or WTC6). CONCLUSIONS In this preclinical model, TRPC6 channels were essential for glioma development via regulation of G2/M phase transition. This study suggests that TRPC6 might be a new target for therapeutic intervention of human glioma.
Collapse
Affiliation(s)
- Xia Ding
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institute for Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Selvaraj S, Sun Y, Singh BB. TRPC channels and their implication in neurological diseases. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2010; 9:94-104. [PMID: 20201820 DOI: 10.2174/187152710790966650] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 08/07/2009] [Indexed: 11/22/2022]
Abstract
Calcium is an essential intracellular messenger and serves critical cellular functions in both excitable and non-excitable cells. Most of the physiological functions in these cells are uniquely regulated by changes in cytosolic Ca2+ levels ([Ca2+](i)), which are achieved via various mechanisms. One of these mechanism(s) is activated by the release of Ca2+ from the endoplasmic reticulum (ER), followed by Ca2+ influx across the plasma membrane (PM). Activation of PM Ca2+ channel is essential for not only refilling of the ER Ca2+ stores, but is also critical for maintaining [Ca2+](i) that regulates biological functions, such as neurosecretion, sensation, long term potentiation, synaptic plasticity, gene regulation, as well as cellular growth and differentiation. Alterations in Ca2+ homeostasis have been suggested in the onset/progression of neurological diseases, such as Parkinson's, Alzheimer's, bipolar disorder, and Huntington's. Available data on transient receptor potential conical (TRPC) protein indicate that these proteins initiate Ca2+ entry pathways and are essential in maintaining cytosolic, ER, and mitochondrial Ca2+ levels. A number of biological functions have been assigned to these TRPC proteins. Silencing of TRPC1 and TRPC3 has been shown to inhibit neuronal proliferation and loss of TRPC1 is implicated in neurodegeneration. Thus, TRPC channels not only contribute towards normal physiological processes, but are also implicated in several human pathological conditions. Overall, it is suggested that these channels could be used as potential therapeutic targets for many of these neurological diseases. Thus, in this review we have focused on the functional implication of TRPC channels in neuronal cells along with the elucidation of their role in neurodegeneration.
Collapse
Affiliation(s)
- Senthil Selvaraj
- Department of Biochemistry and Molecular Biology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201, USA
| | | | | |
Collapse
|
125
|
Avila ME, Sepúlveda FJ, Burgos CF, Moraga-Cid G, Parodi J, Moon RT, Aguayo LG, Opazo C, De Ferrari GV. Canonical Wnt3a modulates intracellular calcium and enhances excitatory neurotransmission in hippocampal neurons. J Biol Chem 2010; 285:18939-47. [PMID: 20404321 PMCID: PMC2881816 DOI: 10.1074/jbc.m110.103028] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 04/16/2010] [Indexed: 11/06/2022] Open
Abstract
A role for Wnt signal transduction in the development and maintenance of brain structures is widely acknowledged. Recent studies have suggested that Wnt signaling may be essential for synaptic plasticity and neurotransmission. However, the direct effect of a Wnt protein on synaptic transmission had not been demonstrated. Here we show that nanomolar concentrations of purified Wnt3a protein rapidly increase the frequency of miniature excitatory synaptic currents in embryonic rat hippocampal neurons through a mechanism involving a fast influx of calcium from the extracellular space, induction of post-translational modifications on the machinery involved in vesicle exocytosis in the presynaptic terminal leading to spontaneous Ca(2+) transients. Our results identify the Wnt3a protein and a member of its complex receptor at the membrane, the low density lipoprotein receptor-related protein 6 (LRP6) coreceptor, as key molecules in neurotransmission modulation and suggest cross-talk between canonical and Wnt/Ca(2+) signaling in central neurons.
Collapse
Affiliation(s)
- Miguel E. Avila
- From the Departments of Biochemistry and Molecular Biology and
| | - Fernando J. Sepúlveda
- Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción P.O. Box 4070386, Chile
| | | | - Gustavo Moraga-Cid
- Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción P.O. Box 4070386, Chile
| | - Jorge Parodi
- Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción P.O. Box 4070386, Chile
| | - Randall T. Moon
- Howard Hughes Medical Institute, Department of Pharmacology and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington 98195, and
| | - Luis G. Aguayo
- Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción P.O. Box 4070386, Chile
| | - Carlos Opazo
- Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción P.O. Box 4070386, Chile
| | - Giancarlo V. De Ferrari
- From the Departments of Biochemistry and Molecular Biology and
- the Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andrés Bello, Santiago P.O. Box 8370134, Chile
| |
Collapse
|
126
|
Ding X, He Z, Shi Y, Wang Q, Wang Y. Targeting TRPC6 channels in oesophageal carcinoma growth. Expert Opin Ther Targets 2010; 14:513-27. [PMID: 20235901 DOI: 10.1517/14728221003733602] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
127
|
Leuner K, Heiser JH, Derksen S, Mladenov MI, Fehske CJ, Schubert R, Gollasch M, Schneider G, Harteneck C, Chatterjee SS, Müller WE. Simple 2,4-diacylphloroglucinols as classic transient receptor potential-6 activators--identification of a novel pharmacophore. Mol Pharmacol 2010; 77:368-77. [PMID: 20008516 DOI: 10.1124/mol.109.057513] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The naturally occurring acylated phloroglucinol derivative hyperforin was recently identified as the first specific canonical transient receptor potential-6 (TRPC6) activator. Hyperforin is the major antidepressant component of St. John's wort, which mediates its antidepressant-like properties via TRPC6 channel activation. However, its pharmacophore moiety for activating TRPC6 channels is unknown. We hypothesized that the phloroglucinol moiety could be the essential pharmacophore of hyperforin and that its activity profile could be due to structural similarities with diacylglycerol (DAG), an endogenous nonselective activator of TRPC3, TRPC6, and TRPC7. Accordingly, a few 2-acyl and 2,4-diacylphloroglucinols were tested for their hyperforin-like activity profiles. We used a battery of experimental models to investigate all functional aspects of TRPC6 activation, including ion channel recordings, Ca(2+) imaging, neurite outgrowth, and inhibition of synaptosomal uptake. Phloroglucinol itself was inactive in all of our assays, which was also the case for 2-acylphloroglucinols. For TRPC6 activation, the presence of two symmetrically acyl-substitutions with appropriate alkyl chains in the phloroglucinol moiety seems to be an essential prerequisite. Potencies of these compounds in all assays were comparable with that of hyperforin for activating the TRPC6 channel. Finally, using structure-based modeling techniques, we suggest a binding mode for hyperforin to TRPC6. Based on this modeling approach, we propose that DAG is able to activate TRPC3, TRPC6, and TRPC7 because of higher flexibility within the chemical structure of DAG compared with the rather rigid structures of hyperforin and the 2,4-diacylphloroglucinol derivatives.
Collapse
Affiliation(s)
- K Leuner
- Institute of Pharmacology, Goethe University, Biocenter N260, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Kojundzic SL, Puljak L, Hogan Q, Sapunar D. Depression of Ca(2+)/calmodulin-dependent protein kinase II in dorsal root ganglion neurons after spinal nerve ligation. J Comp Neurol 2010; 518:64-74. [PMID: 19882720 DOI: 10.1002/cne.22209] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The enzyme calcium/calmodulin-dependent protein kinase II (CaMKII) is associated with memory and its alpha isoform is critical for development of activity-induced synaptic changes. Therefore, we hypothesized that CaMKII is involved in altered function of dorsal root ganglion (DRG) neurons after neuronal injury. To test this hypothesis, Sprague-Dawley rats were made hyperalgesic by L5 and L6 spinal nerve ligation (SNL), and changes in total phosphorylated and unphosphorylated CaMKII (tCaMKII) and phosphorylated form of its alpha isoform (pCaMKIIalpha) were analyzed using immunochemistry in different subpopulations of DRG. SNL did not induce any changes in tCaMKII between experimental groups, while the overall percentage of pCaMKIIalpha-positive neurons in injured L5 DRG SNL (24.8%) decreased significantly when compared to control (41.7%). SNL did not change the percentage of pCaMKIIalpha/N52 colabeled neurons but decreased the percentage of N52-negative nonmyelinated neurons that expressed pCaMKIIalpha from 27% in control animals to 11% after axotomy. We also observed a significant decrease in the percentage of small nonpeptidergic neurons labeled with IB4 (37.6% in control vs. 4.0% in L5 SNL DRG), as well as a decrease in the percentage of pCaMKIIalpha/IB4 colabeled neurons in injured L5 DRGs (27% in control vs. 1% in L5 DRG of SNL group). Our results show that reduction in pCaMKIIalpha levels following peripheral injury is due to the loss of IB4-positive neurons. These results indicate that diminished afferent activity after axotomy may lead to decreased phosphorylation of CaMKIIalpha.
Collapse
Affiliation(s)
- Sanja Lovric Kojundzic
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia
| | | | | | | |
Collapse
|
129
|
Chen YQ, Xie X. Podophyllotoxin induces CREB phosphorylation and CRE-driven gene expression via PKA but not MAPKs. Mol Cells 2010; 29:41-50. [PMID: 20033853 DOI: 10.1007/s10059-010-0015-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 10/15/2009] [Accepted: 10/20/2009] [Indexed: 12/11/2022] Open
Abstract
CRE-driven luciferase reporter is commonly used in drug screening systems involving G protein-coupled receptors (GPCRs). In a screen campaign designed to search for melanocortin-4 receptor (MC4R) agonists, podophyllotoxin, a microtubules disruptor, was found to induce cAMP-responsive element (CRE)-driven reporter expression. MC4R was not involved because podophyllotoxin induced CREB activation and CRE-driven transcription in cells not expressing MC4R. Previous studies indicated that intracellular calcium, PKA, and MAPKs are involved in CREB phosphorylation and activation. Our studies revealed that podophyllotoxin did not affect intracellular calcium level and the phosphorylation state of p38. Podophyllotoxin induced JNK and ERK activation, but blockade of JNK and ERK activation with specific inhibitors had no effect on podophyllotoxin-induced CREB activation and CRE-regulated gene expression. Further experiments revealed that H89, a specific inhibitor of PKA, significantly inhibited podophyllotoxin-induced CREB activation. Podophyllotoxin itself did not alter intracellular cAMP level. Taken together, podophyllotoxin induces CREB activation and CRE-driven gene expression via PKA activation by a cAMP-independent mechanism.
Collapse
Affiliation(s)
- Ya Qiong Chen
- State Key Laboratory of Drug Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | | |
Collapse
|
130
|
Takemura M, Mishima T, Wang Y, Kasahara J, Fukunaga K, Ohashi K, Mizuno K. Ca2+/calmodulin-dependent protein kinase IV-mediated LIM kinase activation is critical for calcium signal-induced neurite outgrowth. J Biol Chem 2009; 284:28554-62. [PMID: 19696021 DOI: 10.1074/jbc.m109.006296] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Actin cytoskeletal remodeling is essential for neurite outgrowth. LIM kinase 1 (LIMK1) regulates actin cytoskeletal remodeling by phosphorylating and inactivating cofilin, an actin filament-disassembling factor. In this study, we investigated the role of LIMK1 in calcium signal-induced neurite outgrowth. The calcium ionophore ionomycin induced LIMK1 activation and cofilin phosphorylation in Neuro-2a neuroblastoma cells. Knockdown of LIMK1 or expression of a kinase-dead mutant of LIMK1 suppressed ionomycin-induced cofilin phosphorylation and neurite outgrowth in Neuro-2a cells. Ionomycin-induced cofilin phosphorylation and neurite outgrowth were also blocked by KN-93, an inhibitor of Ca(2+)/calmodulin-dependent protein kinases (CaMKs), and STO-609, an inhibitor of CaMK kinase. An active form of CaMKIV but not CaMKI enhanced Thr-508 phosphorylation of LIMK1 and increased the kinase activity of LIMK1. Moreover, the active form of CaMKIV induced cofilin phosphorylation and neurite outgrowth, and a dominant negative form of CaMKIV suppressed ionomycin-induced neurite outgrowth. Taken together, our results suggest that LIMK1-mediated cofilin phosphorylation is critical for ionomycin-induced neurite outgrowth and that CaMKIV mediates ionomycin-induced LIMK1 activation.
Collapse
Affiliation(s)
- Miyohiko Takemura
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | | | | | | | | | | | | |
Collapse
|
131
|
Tu P, Kunert-Keil C, Lucke S, Brinkmeier H, Bouron A. Diacylglycerol analogues activate second messenger-operated calcium channels exhibiting TRPC-like properties in cortical neurons. J Neurochem 2009; 108:126-38. [PMID: 19094061 DOI: 10.1111/j.1471-4159.2008.05752.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The lipid diacylglycerol (DAG) analogue 1-oleoyl-2-acetyl-sn-glycerol (OAG) was used to verify the existence of DAG-sensitive channels in cortical neurons dissociated from E13 mouse embryos. Calcium imaging experiments showed that OAG increased the cytosolic concentration of Ca(2+) ([Ca(2+)]i) in nearly 35% of the KCl-responsive cells. These Ca(2+) responses disappeared in a Ca(2+)-free medium supplemented with EGTA. Mn(2+) quench experiments showed that OAG activated Ca(2+)-conducting channels that were also permeant to Ba(2+). The OAG-induced Ca(2+) responses were unaffected by nifedipine or omega-conotoxin GVIA (Sigma-Aldrich, Saint-Quentin Fallavier, France) but blocked by 1-[beta-(3-(4-Methoxyphenyl)propoxy)-4-methoxyphenethyl]-1H-imidazole hydrochloride (SKF)-96365 and Gd(3+). Replacing Na(+) ions with N-methyl-D-glucamine diminished the amplitude of the OAG-induced Ca(2+) responses showing that the Ca(2+) entry was mediated via Na(+)-dependent and Na(+)-independent mechanisms. Experiments carried out with the fluorescent Na(+) indicator CoroNa Green showed that OAG elevated [Na(+)]i. Like OAG, the DAG lipase inhibitor RHC80267 increased [Ca(2+)]i but not the protein kinase C activator phorbol 12-myristate 13-acetate. Moreover, the OAG-induced Ca(2+) responses were not regulated by protein kinase C activation or inhibition but they were augmented by flufenamic acid which increases currents through C-type transient receptor potential protein family (TRPC) 6 channels. In addition, application of hyperforin, a specific activator of TRPC6 channels, elevated [Ca(2+)]i. Whole-cell patch-clamp recordings showed that hyperforin activated non-selective cation channels. They were blocked by SKF-96365 but potentiated by flufenamic acid. Altogether, our data show the presence of hyperforin- and OAG-sensitive Ca(2+)-permeable channels displaying TRPC6-like properties. This is the first report revealing the existence of second messenger-operated channels in cortical neurons.
Collapse
Affiliation(s)
- Peng Tu
- CNRS UMR 5249, Grenoble, France
| | | | | | | | | |
Collapse
|
132
|
Abramowitz J, Birnbaumer L. Physiology and pathophysiology of canonical transient receptor potential channels. FASEB J 2009; 23:297-328. [PMID: 18940894 PMCID: PMC2630793 DOI: 10.1096/fj.08-119495] [Citation(s) in RCA: 253] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Accepted: 09/25/2008] [Indexed: 11/11/2022]
Abstract
The existence of a mammalian family of TRPC ion channels, direct homologues of TRP, the visual transduction channel of flies, was discovered during 1995-1996 as a consequence of research into the mechanism by which the stimulation of the receptor-Gq-phospholipase Cbeta signaling pathway leads to sustained increases in intracellular calcium. Mammalian TRPs, TRPCs, turned out to be nonselective, calcium-permeable cation channels, which cause both a collapse of the cell's membrane potential and entry of calcium. The family comprises 7 members and is widely expressed. Many cells and tissues express between 3 and 4 of the 7 TRPCs. Despite their recent discovery, a wealth of information has accumulated, showing that TRPCs have widespread roles in almost all cells studied, including cells from excitable and nonexcitable tissues, such as the nervous and cardiovascular systems, the kidney and the liver, and cells from endothelia, epithelia, and the bone marrow compartment. Disruption of TRPC function is at the root of some familial diseases. More often, TRPCs are contributing risk factors in complex diseases. The present article reviews what has been uncovered about physiological roles of mammalian TRPC channels since the time of their discovery. This analysis reveals TRPCs as major and unsuspected gates of Ca(2+) entry that contribute, depending on context, to activation of transcription factors, apoptosis, vascular contractility, platelet activation, and cardiac hypertrophy, as well as to normal and abnormal cell proliferation. TRPCs emerge as targets for a thus far nonexistent field of pharmacological intervention that may ameliorate complex diseases.
Collapse
Affiliation(s)
- Joel Abramowitz
- Transmembrane Signaling Group, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
133
|
Functional roles of TRPC channels in the developing brain. Pflugers Arch 2008; 458:283-9. [PMID: 19023589 DOI: 10.1007/s00424-008-0618-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 11/04/2008] [Indexed: 01/17/2023]
Abstract
Transient receptor potential canonical (TRPC) channels are Ca(2+)-permeable, nonselective cation channels formed by homomeric or heteromeric complexes of TRPC proteins that contain six transmembrane domains. These channels can be activated through a phospholipase-C-dependent mechanism, making them sensors for environmental cues. Their expression begins early in embryonic days and remains in adulthood. These channels have important roles in the processes of neuronal development, including neural stem cell proliferation, cerebellar granule cell survival, axon path finding, neuronal morphogenesis, and synaptogenesis. In this review, we will discuss functional implications of TRPC channels during brain development.
Collapse
|
134
|
Wayman GA, Lee YS, Tokumitsu H, Silva AJ, Silva A, Soderling TR. Calmodulin-kinases: modulators of neuronal development and plasticity. Neuron 2008; 59:914-31. [PMID: 18817731 DOI: 10.1016/j.neuron.2008.08.021] [Citation(s) in RCA: 443] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 08/18/2008] [Accepted: 08/19/2008] [Indexed: 11/26/2022]
Abstract
In the nervous system, many intracellular responses to elevated calcium are mediated by CaM kinases (CaMKs), a family of protein kinases whose activities are initially modulated by binding Ca(2+)/calmodulin and subsequently by protein phosphorylation. One member of this family, CaMKII, is well-established for its effects on modulating synaptic plasticity and learning and memory. However, recent studies indicate that some actions on neuronal development and function attributed to CaMKII may instead or in addition be mediated by other members of the CaMK cascade, such as CaMKK, CaMKI, and CaMKIV. This review summarizes key neuronal functions of the CaMK cascade in signal transduction, gene transcription, synaptic development and plasticity, and behavior. The technical challenges of mapping cellular protein kinase signaling pathways are also discussed.
Collapse
Affiliation(s)
- Gary A Wayman
- Vollum Institute, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | | | | | |
Collapse
|