1
|
Skerjanz J, Bauernhofer L, Lenk K, Emmerstorfer-Augustin A, Leitinger G, Reichmann F, Stockner T, Groschner K, Tiapko O. TRPC1: The housekeeper of the hippocampus. Cell Calcium 2024; 123:102933. [PMID: 39116710 DOI: 10.1016/j.ceca.2024.102933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
The non-selective cation channel TRPC1 is highly expressed in the brain. Recent research shows that neuronal TRPC1 forms heteromeric complexes with TRPC4 and TRPC5, with a small portion existing as homotetramers, primarily in the ER. Given that most studies have focused on the role of heteromeric TRPC1/4/5 complexes, it is crucial to investigate the specific role of homomeric TRPC1 in maintaining brain homeostasis. This review highlights recent findings on TRPC1 in the brain, with a focus on the hippocampus, and compiles the latest data on modulators and their binding sites within the TRPC1/4/5 subfamily to stimulate new research on more selective TRPC1 ligands.
Collapse
Affiliation(s)
- Julia Skerjanz
- Gottfried Schatz Research Center, Division of Medical Physics and Biophysics, Medical University of Graz, Austria
| | - Lena Bauernhofer
- Biophysics Division, Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria; BioTechMed-Graz, Austria
| | - Kerstin Lenk
- Institute of Neural Engineering, Graz University of Technology, Austria; BioTechMed-Graz, Austria
| | | | - Gerd Leitinger
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Austria; BioTechMed-Graz, Austria; MEFOgraz, Austria
| | - Florian Reichmann
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria; BioTechMed-Graz, Austria
| | - Thomas Stockner
- Department of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus Groschner
- Gottfried Schatz Research Center, Division of Medical Physics and Biophysics, Medical University of Graz, Austria
| | - Oleksandra Tiapko
- Gottfried Schatz Research Center, Division of Medical Physics and Biophysics, Medical University of Graz, Austria; BioTechMed-Graz, Austria; MEFOgraz, Austria.
| |
Collapse
|
2
|
Seo JY, Jo HR, Lee SH, Kim DG, Lee H, Kim YL, Choi YI, Jung SJ, Son H. TRPC4 deletion elicits behavioral defects in sociability by dysregulating expression of microRNA-138-2. iScience 2024; 27:108617. [PMID: 38188509 PMCID: PMC10770719 DOI: 10.1016/j.isci.2023.108617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/08/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
To investigate whether the defects in transient receptor potential canonical 4 (TRPC4), which is strongly expressed in the hippocampus, are implicated in ASD, we examined the social behaviors of mice in which Trpc4 was deleted (Trpc4-/-). Trpc4-/- mice displayed the core symptoms of ASD, namely, social disability and repetitive behaviors. In microarray analysis of the hippocampus, microRNA (miR)-138-2, the precursor of miR-138, was upregulated in Trpc4-/- mice. We also found that binding of Matrin3 (MATR3), a selective miR-138-2 binding nuclear protein, to miR-138-2 was prominently enhanced, resulting in the downregulation of miR-138 in Trpc4-/- mice. Some parameters of the social defects and repetitive behaviors in the Trpc4-/- mice were rescued by increased miR-138 levels following miR-138-2 infusion in the hippocampus. Together, these results suggest that Trpc4 regulates some signaling components that oppose the development of social behavioral deficits through miR-138 and provide a potential therapeutic strategy for ASD.
Collapse
Affiliation(s)
- Jee Young Seo
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Hye-Ryeong Jo
- Hanyang University Hospital for Rheumatic Diseases, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Seung Hoon Lee
- Hanyang University Hospital for Rheumatic Diseases, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Huiju Lee
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Ye Lim Kim
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Young In Choi
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Sung Jun Jung
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
- Department of Physiology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| | - Hyeon Son
- Graduate School of Biomedical Science and Engineering, Hanyang Biomedical Research Institute, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Korea
| |
Collapse
|
3
|
Hamad MIK, Emerald BS, Kumar KK, Ibrahim MF, Ali BR, Bataineh MF. Extracellular molecular signals shaping dendrite architecture during brain development. Front Cell Dev Biol 2023; 11:1254589. [PMID: 38155836 PMCID: PMC10754048 DOI: 10.3389/fcell.2023.1254589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Proper growth and branching of dendrites are crucial for adequate central nervous system (CNS) functioning. The neuronal dendritic geometry determines the mode and quality of information processing. Any defects in dendrite development will disrupt neuronal circuit formation, affecting brain function. Besides cell-intrinsic programmes, extrinsic factors regulate various aspects of dendritic development. Among these extrinsic factors are extracellular molecular signals which can shape the dendrite architecture during early development. This review will focus on extrinsic factors regulating dendritic growth during early neuronal development, including neurotransmitters, neurotrophins, extracellular matrix proteins, contact-mediated ligands, and secreted and diffusible cues. How these extracellular molecular signals contribute to dendritic growth has been investigated in developing nervous systems using different species, different areas within the CNS, and different neuronal types. The response of the dendritic tree to these extracellular molecular signals can result in growth-promoting or growth-limiting effects, and it depends on the receptor subtype, receptor quantity, receptor efficiency, the animal model used, the developmental time windows, and finally, the targeted signal cascade. This article reviews our current understanding of the role of various extracellular signals in the establishment of the architecture of the dendrites.
Collapse
Affiliation(s)
- Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kukkala K. Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Marwa F. Ibrahim
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mo’ath F. Bataineh
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
4
|
Su CL, Cheng CC, Yen PH, Huang JX, Ting YJ, Chiang PH. Wireless neuromodulation in vitro and in vivo by intrinsic TRPC-mediated magnetomechanical stimulation. Commun Biol 2022; 5:1166. [PMID: 36323817 PMCID: PMC9630493 DOI: 10.1038/s42003-022-04124-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Various magnetic deep brain stimulation (DBS) methods have been developing rapidly in the last decade for minimizing the invasiveness of DBS. However, current magnetic DBS methods, such as magnetothermal and magnetomechanical stimulation, require overexpressing exogeneous ion channels in the central nervous system (CNS). It is unclear whether magnetomechanical stimulation can modulate non-transgenic CNS neurons or not. Here, we reveal that the torque of magnetic nanodiscs with weak and slow alternative magnetic field (50 mT at 10 Hz) could activate neurons through the intrinsic transient receptor potential canonical channels (TRPC), which are mechanosensitive ion channels widely expressed in the brain. The immunostaining with c-fos shows the increasement of neuronal activity by wireless DBS with magnetomechanical approach in vivo. Overall, this research demonstrates a magnetic nanodiscs-based magnetomechanical approach that can be used for wireless neuronal stimulation in vitro and untethered DBS in vivo without implants or genetic manipulation.
Collapse
Affiliation(s)
- Chih-Lun Su
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan, R.O.C
| | - Chao-Chun Cheng
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan, R.O.C
| | - Ping-Hsiang Yen
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan, R.O.C
| | - Jun-Xuan Huang
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan, R.O.C
| | - Yen-Jing Ting
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan, R.O.C
| | - Po-Han Chiang
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan, R.O.C..
| |
Collapse
|
5
|
Ahmed SR, Liu E, Yip A, Lin Y, Balaban E, Pompeiano M. Novel localizations of TRPC5 channels suggest novel and unexplored roles: A study in the chick embryo brain. Dev Neurobiol 2021; 82:41-63. [PMID: 34705331 DOI: 10.1002/dneu.22857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/16/2021] [Accepted: 10/14/2021] [Indexed: 11/06/2022]
Abstract
Mammalian TRPC5 channels are predominantly expressed in the brain, where they increase intracellular Ca2+ and induce depolarization. Because they augment presynaptic vesicle release, cause persistent neural activity, and show constitutive activity, TRPC5s could play a functional role in late developmental brain events. We used immunohistochemistry to examine TRPC5 in the chick embryo brain between 8 and 20 days of incubation, and provide the first detailed description of their distribution in birds and in the whole brain of any animal species. Stained areas substantially increased between E8 and E16, and staining intensity in many areas peaked at E16, a time when chick brains first show organized patterns of whole-brain metabolic activation like what is seen consistently after hatching. Areas showing cell soma staining match areas showing Trpc5 mRNA or protein in adult rodents (cerebral cortex, hippocampus, amygdala, cerebellar Purkinje cells). Chick embryos show protein staining in the optic tectum, cerebellar nuclei, and several brainstem nuclei; equivalent areas in the Allen Institute mouse maps express Trpc5 mRNA. The strongest cell soma staining was found in a dorsal hypothalamic area (matching a group of parvicellular arginine vasotocin neurons and a pallial amygdalohypothalamic cell corridor) and the vagal motor complex. Purkinje cells showed strong dendritic staining at E20. Unexpectedly, we also describe neurite staining in the septum, several hypothalamic nuclei, and a paramedian raphe area; the strongest neurite staining was in the median eminence. These novel localizations suggest new unexplored TRPC5 functions, and possible roles in late embryonic brain development.
Collapse
Affiliation(s)
- Sharifuddin Rifat Ahmed
- Department of Psychology, McGill University, Montreal, Quebec, Canada.,Faculté de médecine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Elise Liu
- Department of Psychology, McGill University, Montreal, Quebec, Canada.,Institute du Cerveau - ICM, Paris Brain Institute, Paris, 75013, France
| | - Alissa Yip
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Yuqi Lin
- Department of Psychology, McGill University, Montreal, Quebec, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Evan Balaban
- Department of Psychology, McGill University, Montreal, Quebec, Canada.,Department of Bioengineering and Aerospace Engineering, Carlo III University of Madrid, Avda. de la Universidad 30, Leganés, Madrid, E-28911, Spain
| | - Maria Pompeiano
- Department of Psychology, McGill University, Montreal, Quebec, Canada.,Department of Bioengineering and Aerospace Engineering, Carlo III University of Madrid, Avda. de la Universidad 30, Leganés, Madrid, E-28911, Spain
| |
Collapse
|
6
|
Nakao A, Matsunaga Y, Hayashida K, Takahashi N. Role of Oxidative Stress and Ca 2+ Signaling in Psychiatric Disorders. Front Cell Dev Biol 2021; 9:615569. [PMID: 33644051 PMCID: PMC7905097 DOI: 10.3389/fcell.2021.615569] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
Psychiatric disorders are caused by complex and diverse factors, and numerous mechanisms have been proposed for the pathogenesis of these disorders. Accumulating evidence suggests that oxidative stress is one of the general factors involved in the pathogenesis/pathophysiology of major psychiatric disorders, including bipolar disorder, depression, anxiety disorder, and schizophrenia. Indeed, some clinical trials have shown improvement of the symptoms of these disorders by antioxidant supplementation. However, the molecular basis for the relationship between oxidative stress and the pathogenesis of psychiatric disorders remains largely unknown. In general, Ca2+ channels play central roles in neuronal functions, including neuronal excitability, neurotransmitter release, synaptic plasticity, and gene regulation, and genes that encode Ca2+ channels have been found to be associated with psychiatric disorders. Notably, a class of Ca2+-permeable transient receptor potential (TRP) cation channels is activated by changes in cellular redox status, whereby these TRP channels can link oxidative stress to Ca2+ signals. Given the unique characteristic of redox-sensitive TRP channels, these channels could be a target for delineating the pathogenesis or pathophysiology of psychiatric disorders. In this review, we summarize the outcomes of clinical trials for antioxidant treatment in patients with psychiatric disorders and the current insights into the physiological/pathological significance of redox-sensitive TRP channels in the light of neural functions, including behavioral phenotypes, and discuss the potential role of TRP channels in the pathogenesis of psychiatric disorders. Investigation of redox-sensitive TRP channels may lead to the development of novel therapeutic strategies for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Akito Nakao
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yoshihiro Matsunaga
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Katsumi Hayashida
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Nobuaki Takahashi
- The Hakubi Center for Advanced Research, Kyoto University, Kyoto, Japan
| |
Collapse
|
7
|
Lavanderos B, Silva I, Cruz P, Orellana-Serradell O, Saldías MP, Cerda O. TRP Channels Regulation of Rho GTPases in Brain Context and Diseases. Front Cell Dev Biol 2020; 8:582975. [PMID: 33240883 PMCID: PMC7683514 DOI: 10.3389/fcell.2020.582975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological and neuropsychiatric disorders are mediated by several pathophysiological mechanisms, including developmental and degenerative abnormalities caused primarily by disturbances in cell migration, structural plasticity of the synapse, and blood-vessel barrier function. In this context, critical pathways involved in the pathogenesis of these diseases are related to structural, scaffolding, and enzymatic activity-bearing proteins, which participate in Ca2+- and Ras Homologs (Rho) GTPases-mediated signaling. Rho GTPases are GDP/GTP binding proteins that regulate the cytoskeletal structure, cellular protrusion, and migration. These proteins cycle between GTP-bound (active) and GDP-bound (inactive) states due to their intrinsic GTPase activity and their dynamic regulation by GEFs, GAPs, and GDIs. One of the most important upstream inputs that modulate Rho GTPases activity is Ca2+ signaling, positioning ion channels as pivotal molecular entities for Rho GTPases regulation. Multiple non-selective cationic channels belonging to the Transient Receptor Potential (TRP) family participate in cytoskeletal-dependent processes through Ca2+-mediated modulation of Rho GTPases. Moreover, these ion channels have a role in several neuropathological events such as neuronal cell death, brain tumor progression and strokes. Although Rho GTPases-dependent pathways have been extensively studied, how they converge with TRP channels in the development or progression of neuropathologies is poorly understood. Herein, we review recent evidence and insights that link TRP channels activity to downstream Rho GTPase signaling or modulation. Moreover, using the TRIP database, we establish associations between possible mediators of Rho GTPase signaling with TRP ion channels. As such, we propose mechanisms that might explain the TRP-dependent modulation of Rho GTPases as possible pathways participating in the emergence or maintenance of neuropathological conditions.
Collapse
Affiliation(s)
- Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Octavio Orellana-Serradell
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
8
|
Effects of Transient Receptor Potential Cation 5 (TRPC5) Inhibitor, NU6027, on Hippocampal Neuronal Death after Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21218256. [PMID: 33158109 PMCID: PMC7662546 DOI: 10.3390/ijms21218256] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) can cause physical, cognitive, social, and behavioral changes that can lead to permanent disability or death. After primary brain injury, translocated free zinc can accumulate in neurons and lead to secondary events such as oxidative stress, inflammation, edema, swelling, and cognitive impairment. Under pathological conditions, such as ischemia and TBI, excessive zinc release, and accumulation occurs in neurons. Based on previous research, it hypothesized that calcium as well as zinc would be influx into the TRPC5 channel. Therefore, we hypothesized that the suppression of TRPC5 would prevent neuronal cell death by reducing the influx of zinc and calcium. To test our hypothesis, we used a TBI animal model. After the TBI, we immediately injected NU6027 (1 mg/kg, intraperitoneal), TRPC5 inhibitor, and then sacrificed animals 24 h later. We conducted Fluoro-Jade B (FJB) staining to confirm the presence of degenerating neurons in the hippocampal cornus ammonis 3 (CA3). After the TBI, the degenerating neuronal cell count was decreased in the NU6027-treated group compared with the vehicle-treated group. Our findings suggest that the suppression of TRPC5 can open a new therapeutic window for a reduction of the neuronal death that may occur after TBI.
Collapse
|
9
|
Abstract
Neurotrophin-3 (NT-3) belongs to a family of growth factors called neurotrophins whose actions are centered in the nervous system. NT-3 is structurally related to other neurotrophins like brain-derived neurotrophic factor. The expression of NT-3 starts with the onset of neurogenesis and continues throughout life. A wealth of information links NT-3 to the growth, differentiation, and survival of hippocampal cells as well as sympathetic and sensory neurons. These studies have described the distribution of NT-3 and its receptors throughout development and in the mature nervous system. Prior works has begun to cell-type specific impact of NT-3 as well as identify the signaling pathways involved. However, much less is known about how NT-3 regulates synaptic transmission. This chapter focuses role of NT-3 in the modulation of synaptic transmission.
Collapse
|
10
|
Maria-Ferreira D, de Oliveira NMT, da Silva LCM, Fernandes ES. Evidence of a Role for the TRPC Subfamily in Mediating Oxidative Stress in Parkinson's Disease. Front Physiol 2020; 11:332. [PMID: 32457638 PMCID: PMC7225354 DOI: 10.3389/fphys.2020.00332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) represents one of the most common multifactorial neurodegenerative disorders affecting the elderly population. It is associated with the aggregation of α-synuclein protein and the loss of dopaminergic neurons in the substantia nigra pars compacta of the brain. The disease is mainly represented by motor symptoms, such as resting tremors, postural instability, rigidity, and bradykinesia, that develop slowly over time. Parkinson's disease can also manifest as disturbances in non-motor functions. Although the pathology of PD has not yet been fully understood, it has been suggested that the disruption of the cellular redox status may contribute to cellular oxidative stress and, thus, to cell death. The generation of reactive oxygen species and reactive nitrogen intermediates, as well as the dysfunction of dopamine metabolism, play important roles in the degeneration of dopaminergic neurons. In this context, the transient receptor potential channel canonical (TRPC) sub-family plays an important role in neuronal degeneration. Additionally, PD gene products, including DJ-1, SNCA, UCH-L1, PINK-1, and Parkin, also interfere with mitochondrial function leading to reactive oxygen species production and dopaminergic neuronal vulnerability to oxidative stress. Herein, we discuss the interplay between these various biochemical and molecular events that ultimately lead to dopaminergic signaling disruption, highlighting the recently identified roles of TRPC in PD.
Collapse
Affiliation(s)
- Daniele Maria-Ferreira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Liziane Cristine Malaquias da Silva
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Elizabeth Soares Fernandes
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| |
Collapse
|
11
|
Roach KM, Bradding P. Ca 2+ signalling in fibroblasts and the therapeutic potential of K Ca3.1 channel blockers in fibrotic diseases. Br J Pharmacol 2020; 177:1003-1024. [PMID: 31758702 DOI: 10.1111/bph.14939] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
The role of Ca2+ signalling in fibroblasts is of great interest in fibrosis-related diseases. Intracellular free Ca2+ ([Ca2+ ]i ) is a ubiquitous secondary messenger, regulating a number of cellular functions such as secretion, metabolism, differentiation, proliferation and contraction. The intermediate conductance Ca2+ -activated K+ channel KCa 3.1 is pivotal in Ca2+ signalling and plays a central role in fibroblast processes including cell activation, migration and proliferation through the regulation of cell membrane potential. Evidence from a number of approaches demonstrates that KCa 3.1 plays an important role in the development of many fibrotic diseases, including idiopathic pulmonary, renal tubulointerstitial fibrosis and cardiovascular disease. The KCa 3.1 selective blocker senicapoc was well tolerated in clinical trials for sickle cell disease, raising the possibility of rapid translation to the clinic for people suffering from pathological fibrosis. This review after analysing all the data, concludes that targeting KCa 3.1 should be a high priority for human fibrotic disease.
Collapse
Affiliation(s)
- Katy M Roach
- Institute for Lung Health, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Peter Bradding
- Institute for Lung Health, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
12
|
Gq-Coupled Muscarinic Receptor Enhancement of KCNQ2/3 Channels and Activation of TRPC Channels in Multimodal Control of Excitability in Dentate Gyrus Granule Cells. J Neurosci 2018; 39:1566-1587. [PMID: 30593498 DOI: 10.1523/jneurosci.1781-18.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/21/2022] Open
Abstract
KCNQ (Kv7, "M-type") K+ channels and TRPC (transient receptor potential, "canonical") cation channels are coupled to neuronal discharge properties and are regulated via Gq/11-protein-mediated signals. Stimulation of Gq/11-coupled receptors both consumes phosphatidylinositol 4,5-bisphosphate (PIP2) via phosphalipase Cβ hydrolysis and stimulates PIP2 synthesis via rises in Ca2+ i and other signals. Using brain-slice electrophysiology and Ca2+ imaging from male and female mice, we characterized threshold K+ currents in dentate gyrus granule cells (DGGCs) and CA1 pyramidal cells, the effects of Gq/11-coupled muscarinic M1 acetylcholine (M1R) stimulation on M current and on neuronal discharge properties, and elucidated the intracellular signaling mechanisms involved. We observed disparate signaling cascades between DGGCs and CA1 neurons. DGGCs displayed M1R enhancement of M-current, rather than suppression, due to stimulation of PIP2 synthesis, which was paralleled by increased PIP2-gated G-protein coupled inwardly rectifying K+ currents as well. Deficiency of KCNQ2-containing M-channels ablated the M1R-induced enhancement of M-current in DGGCs. Simultaneously, M1R stimulation in DGGCs induced robust increases in [Ca2+]i, mostly due to TRPC currents, consistent with, and contributing to, neuronal depolarization and hyperexcitability. CA1 neurons did not display such multimodal signaling, but rather M current was suppressed by M1R stimulation in these cells, similar to the previously described actions of M1R stimulation on M-current in peripheral ganglia that mostly involves PIP2 depletion. Therefore, these results point to a pleiotropic network of cholinergic signals that direct cell-type-specific, precise control of hippocampal function with strong implications for hyperexcitability and epilepsy.SIGNIFICANCE STATEMENT At the neuronal membrane, protein signaling cascades consisting of ion channels and metabotropic receptors govern the electrical properties and neurotransmission of neuronal networks. Muscarinic acetylcholine receptors are G-protein-coupled metabotropic receptors that control the excitability of neurons through regulating ion channels, intracellular Ca2+ signals, and other second-messenger cascades. We have illuminated previously unknown actions of muscarinic stimulation on the excitability of hippocampal principal neurons that include M channels, TRPC (transient receptor potential, "canonical") cation channels, and powerful regulation of lipid metabolism. Our results show that these signaling pathways, and mechanisms of excitability, are starkly distinct between peripheral ganglia and brain, and even between different principal neurons in the hippocampus.
Collapse
|
13
|
Tang Y, Liu S, Shu H, Xing Y, Tao F. AMPA receptor GluA1 Ser831 phosphorylation is critical for nitroglycerin-induced migraine-like pain. Neuropharmacology 2018; 133:462-469. [PMID: 29486167 PMCID: PMC5858972 DOI: 10.1016/j.neuropharm.2018.02.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/13/2018] [Accepted: 02/23/2018] [Indexed: 12/13/2022]
Abstract
Migraine is the third most common disease worldwide; however, the mechanisms underlying migraine headache are still not fully understood. Previous studies have demonstrated that α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor phosphorylation plays an important role in central sensitization of pain transmission. In the present study, we observed that AMPA receptor GluA1 Ser831 phosphorylation was enhanced in the spinal trigeminal nucleus caudalis (Sp5C) after intraperitoneal injection of nitroglycerin (NTG). The NTG injection induced acute migraine-like pain including photophobia and mechanical hypersensitivity as reported previously. Interestingly, targeted mutation of GluA1 Ser831 site to prevent phosphorylation significantly inhibited NTG-induced migraine-like pain. Moreover, NTG incubation caused a robust Ca2+ influx in cultured brainstem neurons, which was dramatically inhibited by GluA1 S831A (serine at the 831 site of GluA1 is mutated to alanine) phospho-deficient mutation, and treatment with 1-naphthyl acetyl spermine (NASPM), a selective Ca2+-permeable AMPA receptor channel blocker, dose-dependently blocked the NTG-evoked increase of Ca2+ influx in the cultured neurons. We further found that intra-Sp5C injection of NASPM significantly inhibited NTG-produced mechanical hypersensitivity. These results suggest that AMPA receptor phosphorylation at the Ser831 site in the Sp5C is critical for NTG-induced migraine-like pain.
Collapse
Affiliation(s)
- Yuanyuan Tang
- Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, Henan, China; Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Sufang Liu
- Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, Henan, China; Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Hui Shu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Ying Xing
- Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, Henan, China.
| | - Feng Tao
- Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, Henan, China; Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA; Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, TX, USA.
| |
Collapse
|
14
|
Qu Z, Wang Y, Li X, Wu L, Wang Y. TRPC6 expression in neurons is differentially regulated by NR2A- and NR2B-containing NMDA receptors. J Neurochem 2017; 143:282-293. [PMID: 28902407 DOI: 10.1111/jnc.14215] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/30/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
The expression of transient receptor potential canonical 6 (TRPC6) in central nervous system (CNS) is important for neuronal functions and certain neural disorders. However, the regulatory mechanism of TRPC6 expression in neurons is still obscure. In this study, we show that TRPC6 expression in the primary cultured cortical neurons is bidirectionally regulated by glutamate. Activation of NR2A-containing NMDARs induces TRPC6 transcription through a calcineurin-dependent pathway. In contrast, activation of NR2B-containing NMDARs causes TRPC6 degradation through calpain. Thus, TRPC6 expression in neurons is regulated by glutamate in a bidirectional manner that is dependent on NR2A and NR2B.
Collapse
Affiliation(s)
- Zhongwei Qu
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,The Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuqing Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,The Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Xia Li
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| | - Lin Wu
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Maddox JW, Khorsandi N, Gleason E. TRPC5 is required for the NO-dependent increase in dendritic Ca 2+ and GABA release from chick retinal amacrine cells. J Neurophysiol 2017; 119:262-273. [PMID: 28978766 DOI: 10.1152/jn.00500.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
GABAergic signaling from amacrine cells (ACs) is a fundamental aspect of visual signal processing in the inner retina. We have previously shown that nitric oxide (NO) can elicit release of GABA independently from activation of voltage-gated Ca2+ channels in cultured retinal ACs. This voltage-independent quantal GABA release relies on a Ca2+ influx mechanism with pharmacological characteristics consistent with the involvement of the transient receptor potential canonical (TRPC) channels TRPC4 and/or TRPC5. To determine the identity of these channels, we evaluated the ability of NO to elevate dendritic Ca2+ and to stimulate GABA release from cultured ACs under conditions known to alter the function of TRPC4 and 5. We found that these effects of NO are phospholipase C dependent, have a biphasic dependence on La3+, and are unaffected by moderate concentrations of the TRPC4-selective antagonist ML204. Together, these results suggest that NO promotes GABA release by activating TRPC5 channels in AC dendrites. To confirm a role for TRPC5, we knocked down the expression of TRPC5 using CRISPR/Cas9-mediated gene knockdown and found that both the NO-dependent Ca2+ elevations and increase in GABA release are dependent on the expression of TRPC5. These results demonstrate a novel NO-dependent mechanism for regulating neurotransmitter output from retinal ACs. NEW & NOTEWORTHY Elucidating the mechanisms regulating GABAergic synaptic transmission in the inner retina is key to understanding the flexibility of retinal ganglion cell output. Here, we demonstrate that nitric oxide (NO) can activate a transient receptor potential canonical 5 (TRPC5)-mediated Ca2+ influx, which is sufficient to drive vesicular GABA release from retinal amacrine cells. This NO-dependent mechanism can bypass the need for depolarization and may have an important role in processing the visual signal by enhancing retinal amacrine cell GABAergic inhibitory output.
Collapse
Affiliation(s)
- J Wesley Maddox
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Nikka Khorsandi
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| |
Collapse
|
16
|
Mukherjee S, Sheng W, Sun R, Janssen LJ. Ca2+/calmodulin-dependent protein kinase IIβ and IIδ mediate TGFβ-induced transduction of fibronectin and collagen in human pulmonary fibroblasts. Am J Physiol Lung Cell Mol Physiol 2017; 312:L510-L519. [DOI: 10.1152/ajplung.00084.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/20/2022] Open
Abstract
It is now clear that in addition to activating several complex kinase pathways (Smad, MAP kinase, PI3 kinase), TGFβ also acts by elevating cytosolic Ca2+ concentration within human pulmonary fibroblasts. Ca2+/calmodulin-dependent protein kinase II (CamK II) is also known to regulate gene expression in fibroblasts. In this study, we examined the interactions between calcium signaling, activation of CamK and other kinases, and extracellular matrix (ECM) gene expression. Human pulmonary fibroblasts were cultured and stimulated with artificially generated Ca2+ pulses in the absence of TGFβ, or with TGFβ (1 nM) or vehicle in the presence of various blockers of Ca2+ signaling. PCR and Western blotting were used to measure gene expression and protein levels, respectively. We found that Ca2+ pulses in the absence of TGFβ increased ECM gene expression in a pulse frequency-dependent manner, and that blocking Ca2+ signaling and the CamK II pathway significantly reduced TGFβ-mediated ECM gene expression, without having any effects on other kinase pathways (Smad, PI3 kinase, or MAP kinase). We also found that TGFβ elevated the expression of CamK IIβ and CamK IIδ, while siRNA silencing of those two subtypes significantly reduced TGFβ-mediated expression of collagen A1 and fibronectin 1. Our data suggest that TGFβ induces the expression of CamK IIβ and CamK IIδ, which in turn are activated by TGFβ-evoked Ca2+ waves in a frequency-dependent manner, leading to increased expression of ECM proteins.
Collapse
Affiliation(s)
- Subhendu Mukherjee
- Firestone Institute for Respiratory Health, St. Joseph’s Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Wei Sheng
- Firestone Institute for Respiratory Health, St. Joseph’s Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rui Sun
- Firestone Institute for Respiratory Health, St. Joseph’s Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Luke J. Janssen
- Firestone Institute for Respiratory Health, St. Joseph’s Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
17
|
Griesi-Oliveira K, Suzuki AM, Muotri AR. TRPC Channels and Mental Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:137-148. [PMID: 28508319 DOI: 10.1007/978-94-024-1088-4_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transient receptor potential canonical (TRPC) channels mediate the influx of different types of cations through the cell membrane and are involved in many functions of the organism. Evidences of involvement of TRPC channels in neuronal development suggest that this family of proteins might play a role in certain neurological disorders. As reported, knockout mice for different TRPC channels show alterations in neuronal morphological and functional parameters, with behavioral abnormalities, such as in exploratory and social behaviors. Although mutations in TRPC channels could be related to mental/neurological disorders, there are only a few cases reported in literature, indicating that this correlation should be further explored. Nonetheless, other functional evidences support the implication of these channels in neurological diseases. In this chapter, we summarize the main findings relating TRPC channels to neurological disorders, such as autism spectrum disorders, bipolar disorder, and intellectual disability among others.
Collapse
Affiliation(s)
| | - Angela May Suzuki
- Department of Genetics and Evolutionary Biology, Bioscience Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Alysson Renato Muotri
- Department of Pediatrics and Department of Cellular & Molecular Medicine, University of California San Diego School of Medicine, La Jolla, CA, USA. .,Rady Children's Hospital San Diego, San Diego, CA, USA. .,UCSD Stem Cell Program, Institute for Genomic Medicine, New York, NY, USA.
| |
Collapse
|
18
|
TRPC Channels and Neuron Development, Plasticity, and Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:95-110. [PMID: 28508316 DOI: 10.1007/978-94-024-1088-4_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this chapter, we mainly focus on the functions of TRPC channels in brain development, including neural progenitor proliferation, neurogenesis, neuron survival, axon guidance, dendritic morphology, synaptogenesis, and neural plasticity. We also notice emerging advances in understanding the functions of TRPC channels in periphery, especially their functions in sensation and nociception in dorsal root ganglion (DRG). Because TRPC channels are expressed in all major types of glial cells, which account for at least half of total cells in the brain, TRPC channels may act as modulators for glial functions as well. The future challenges for studying these channels could be (1) the detailed protein structures of these channels, (2) their cell type-specific functions, (3) requirement for their specific blockers or activators, and (4) change in the channel conformation in the brain.
Collapse
|
19
|
He Z. TRPC Channel Downstream Signaling Cascades. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:25-33. [PMID: 28508310 DOI: 10.1007/978-94-024-1088-4_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The family of TRP channel is comprised of a large group of cation-permeable channels, displaying as signaling integrators for sensing extracellular stimulus and initiating intracellular signaling cascades. This chapter offers a brief review of the signaling molecules related to TRPC channels, the first identified mammalian TRP family. Besides the signaling molecules involved in TRPC activation, I will focus on their upstream and downstream signaling cascades and the molecules involved in their intracellular trafficking.
Collapse
Affiliation(s)
- Zhuohao He
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
Calcium Entry Through Thermosensory Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:265-304. [PMID: 27161233 DOI: 10.1007/978-3-319-26974-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ThermoTRPs are unique channels that mediate Na(+) and Ca(2+) currents in response to changes in ambient temperature. In combination with their activation by other physical and chemical stimuli, they are considered key integrators of environmental cues into neuronal excitability. Furthermore, roles of thermoTRPs in non-neuronal tissues are currently emerging such as insulin secretion in pancreatic β-cells, and links to cancer. Calcium permeability through thermoTRPs appears a central hallmark for their physiological and pathological activities. Moreover, it is currently being proposed that beyond working as a second messenger, Ca(2+) can function locally by acting on protein complexes near the membrane. Interestingly, thermoTRPs can enhance and expand the inherent plasticity of signalplexes by conferring them temperature, pH and lipid regulation through Ca(2+) signalling. Thus, unveiling the local role of Ca(2+) fluxes induced by thermoTRPs on the dynamics of membrane-attached signalling complexes as well as their significance in cellular processes, are central issues that will expand the opportunities for therapeutic intervention in disorders involving dysfunction of thermoTRP channels.
Collapse
|
21
|
TRPC6 specifically interacts with APP to inhibit its cleavage by γ-secretase and reduce Aβ production. Nat Commun 2015; 6:8876. [PMID: 26581893 PMCID: PMC4696454 DOI: 10.1038/ncomms9876] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/12/2015] [Indexed: 01/12/2023] Open
Abstract
Generation of β-amyloid (Aβ) peptide in Alzheimer's disease involves cleavage of amyloid precursor protein (APP) by γ-secretase, a protease known to cleave several substrates, including Notch. Finding specific modulators for γ-secretase could be a potential avenue to treat the disease. Here, we report that transient receptor potential canonical (TRPC) 6 specifically interacts with APP leading to inhibition of its cleavage by γ-secretase and reduction in Aβ production. TRPC6 interacts with APP (C99), but not with Notch, and prevents C99 interaction with presenilin 1 (PS1). A fusion peptide derived from TRPC6 also reduces Aβ levels without effect on Notch cleavage. Crossing APP/PS1 mice with TRPC6 transgenic mice leads to a marked reduction in both plaque load and Aβ levels, and improvement in structural and behavioural impairment. Thus, TRPC6 specifically modulates γ-secretase cleavage of APP and preventing APP (C99) interaction with PS1 via TRPC6 could be a novel strategy to reduce Aβ formation. Attempts to treat Alzheimer's disease by targeting γ-secretase cleavage of APP into Aß have been unsuccessful, partially due to off-target effects. Here, the authors identify TRPC6 as a novel γ-secretase modulator, showing that it interacts with APP to regulate Aß levels while sparing Notch cleavage.
Collapse
|
22
|
Janssen LJ, Mukherjee S, Ask K. Calcium Homeostasis and Ionic Mechanisms in Pulmonary Fibroblasts. Am J Respir Cell Mol Biol 2015; 53:135-48. [PMID: 25785898 DOI: 10.1165/rcmb.2014-0269tr] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fibroblasts are key cellular mediators of many chronic interstitial lung diseases, including idiopathic pulmonary fibrosis, scleroderma, sarcoidosis, drug-induced interstitial lung disease, and interstitial lung disease in connective tissue disease. A great deal of effort has been expended to understand the signaling mechanisms underlying the various cellular functions of fibroblasts. Recently, it has been shown that Ca(2+) oscillations play a central role in the regulation of gene expression in human pulmonary fibroblasts. However, the mechanisms whereby cytosolic [Ca(2+)] are regulated and [Ca(2+)] oscillations transduced are both poorly understood. In this review, we present the general concepts of [Ca(2+)] homeostasis, of ionic mechanisms responsible for various Ca(2+) fluxes, and of regulation of gene expression by [Ca(2+)]. In each case, we then also summarize the original findings that pertain specifically to pulmonary fibroblasts. From these data, we propose an overall signaling cascade by which excitation of the fibroblasts triggers pulsatile release of internally sequestered Ca(2+), which, in turn, activates membrane conductances, including voltage-dependent Ca(2+) influx pathways. Collectively, these events produce recurring Ca(2+) oscillations, the frequency of which is transduced by Ca(2+)-dependent transcription factors, which, in turn, orchestrate a variety of cellular events, including proliferation, synthesis/secretion of extracellular matrix proteins, autoactivation (production of transforming growth factor-β), and transformation into myofibroblasts. That unifying hypothesis, in turn, allows us to highlight several specific cellular targets and therapeutic intervention strategies aimed at controlling unwanted pulmonary fibrosis. The relationships between Ca(2+) signaling events and the unfolded protein response and apoptosis are also explored.
Collapse
Affiliation(s)
- Luke J Janssen
- Firestone Institute for Respiratory Health, St. Joseph's Hospital, and Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Subhendu Mukherjee
- Firestone Institute for Respiratory Health, St. Joseph's Hospital, and Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health, St. Joseph's Hospital, and Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
23
|
Safina DR, Surin AM, Pinelis VG, Kostrov SV. Effect of neurotrophin-3 precursor on glutamate-induced calcium homeostasis deregulation in rat cerebellum granule cells. J Neurosci Res 2015; 93:1865-73. [DOI: 10.1002/jnr.23667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 08/19/2015] [Accepted: 08/25/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Dina R. Safina
- Laboratory of Protein Engineering; Institute of Molecular Genetics, Russian Academy of Sciences; Moscow Russia
| | - Alexander M. Surin
- Laboratory of Ionic Transport and Intracellular Signaling Pathology; Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences; Moscow Russia
- Laboratory of Molecular; Genetic, and Cell Biology, Scientific Center for Children's Health, Russian Academy of Medical Sciences; Moscow Russia
- Laboratory of Electrophysiology; Pirogov Russian National Research Medical University; Moscow Russia
| | - Vsevolod G. Pinelis
- Laboratory of Molecular; Genetic, and Cell Biology, Scientific Center for Children's Health, Russian Academy of Medical Sciences; Moscow Russia
| | - Sergey V. Kostrov
- Laboratory of Protein Engineering; Institute of Molecular Genetics, Russian Academy of Sciences; Moscow Russia
- Centre for Convergence of Nano-, Bio-, Information, and Cognitive Sciences and Technologies, National Research Centre “Kurchatov Institute,”; Moscow Russia
| |
Collapse
|
24
|
Guo W, Xie L, Zhao L, Zhao Y. mRNA and microRNA expression profiles of radioresistant NCI-H520 non-small cell lung cancer cells. Mol Med Rep 2015; 12:1857-67. [PMID: 25873351 PMCID: PMC4464398 DOI: 10.3892/mmr.2015.3600] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 03/09/2015] [Indexed: 01/05/2023] Open
Abstract
To elucidate the mechanism of radioresistance in non-small cell lung cancer (NSCLC) cells and to identify key molecules conferring radioresistance, the radioresistant subclone NCI-H520/R, derived from the NCI-H520 NSCLC cell line, was established with eight rounds of sublethal irradiation. The radioresistant features were subsequently assessed using a clonogenic assay, analysis of apoptosis and an MTT assay, the gene expression levels were examined using an Agilent Whole Human Genome 4×44 k Oligo microarray and Agilent Human miRCURY™ LNA array, and confirmed by reverse transcription-quantitative polymerase chain reaction. Pathway analysis and Gene Ontology (GO) analysis were performed to determine the biological functions of the subset of differentially expressed genes. miRNA-mRNA correlation analysis between the expression levels of each miRNA and all its predicted target genes was performed to further understand the radioresistance in the NCI-H520 cells. Following eight rounds of sublethal irradiation, a total of 2,862 mRNAs were significantly differentially expressed in the NCI-H520/R cells, including 893 upregulated genes and 1,969 downregulated genes. A total of 162 upregulated miRNAs and 274 downregulated miRNAs were significantly deregulated in the NCI-H520/R cells. Multiple core regulatory processes and signaling pathways were identified as being of likely relevance to radioresistance in NCI-H520/R cells, including the mitogen-activated protein kinase signaling pathway and neurotrophin signaling pathway. The expression of genes associated with radioresistance reflects the complex biological processes involved in clinical cancer cell eradication and requires further investigation for future enhancement of therapy.
Collapse
Affiliation(s)
- Wei Guo
- Ultrasound Diagnosis Department, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| | - Li Xie
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| | - Long Zhao
- Ultrasound Diagnosis Department, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| | - Yuehuan Zhao
- Ultrasound Diagnosis Department, Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
25
|
Ca(2+) signaling initiated by canonical transient receptor potential channels in dendritic development. Neurosci Bull 2015; 31:351-6. [PMID: 25732528 DOI: 10.1007/s12264-014-1511-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/22/2014] [Indexed: 12/25/2022] Open
Abstract
The spatial patterns of dendritic structures diverge in different types of neurons as adaptations to their unique functions. Although different intracellular mechanisms underlying dendritic morphogenesis have been suggested, it is evident that the elevation in intracellular Ca(2+) levels plays a major role in the process. Canonical transient receptor potential (TRPC) channels, known to be non-selective Ca(2+)-permeable cation channels, act as environmental detectors to sense and transduce extracellular signals into different intracellular responses, including the regulation of dendritic growth, via Ca(2+) influx. Here, we review recent advances in the understanding of Ca(2+) signaling, especially signals mediated by Ca(2+) influx via TRPC channels, and the underlying molecular events in dendritic development.
Collapse
|
26
|
Li S, Wang J, Wei Y, Liu Y, Ding X, Dong B, Xu Y, Wang Y. Critical role of TRPC6 in maintaining the stability of HIF-1α in glioma cells under hypoxia. J Cell Sci 2015; 128:3317-29. [PMID: 26187851 DOI: 10.1242/jcs.173161] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/07/2015] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a key transcriptional factor responsible for the expression of a broad range of genes that facilitate acclimatization to hypoxia. Its stability is predominantly controlled by rapid hydroxylation of two prolines on its α subunit. However, how the rapid hydroxylation of HIF-1α is regulated is not fully understood. Here, we report that transient receptor potential canonical (TRPC) 6 channels control hydroxylation and stability of HIF-1α in human glioma cells under hypoxia. TRPC6 was rapidly activated by IGF-1R-PLCγ-IP3R pathway in hypoxia. Inhibition of TRPC6 enhanced the levels of α-ketoglutarate (α-KG) and promoted hydroxylation of HIF-1α to suppress HIF-1α accumulation without affecting its transcription or translation. Dimethyloxalylglycine N-(methoxyoxoacetyl)-glycine methyl ester (DMOG), an analog of α-KG, reversed the inhibition of HIF-1α accumulation. Moreover, TRPC6 regulated GLUT1 expression depending on HIF-1α accumulation to affect glucose uptake in hypoxia. Our results suggest that TRPC6 regulates metabolism to affect HIF-1α stability and consequent glucose metabolism in human glioma cells under hypoxia.
Collapse
Affiliation(s)
- Shanshan Li
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institutes of Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinkui Wang
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yi Wei
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yongjian Liu
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xia Ding
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institutes of Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Dong
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yinghui Xu
- Department of Neurosurgery, 1st Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Shanghai Institutes of Biological Sciences, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
27
|
Increased Expression of TRPC5 in Cortical Lesions of the Focal Cortical Dysplasia. J Mol Neurosci 2014; 55:561-9. [DOI: 10.1007/s12031-014-0390-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 07/22/2014] [Indexed: 11/25/2022]
|
28
|
Nilius B, Szallasi A. Transient receptor potential channels as drug targets: from the science of basic research to the art of medicine. Pharmacol Rev 2014; 66:676-814. [PMID: 24951385 DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The large Trp gene family encodes transient receptor potential (TRP) proteins that form novel cation-selective ion channels. In mammals, 28 Trp channel genes have been identified. TRP proteins exhibit diverse permeation and gating properties and are involved in a plethora of physiologic functions with a strong impact on cellular sensing and signaling pathways. Indeed, mutations in human genes encoding TRP channels, the so-called "TRP channelopathies," are responsible for a number of hereditary diseases that affect the musculoskeletal, cardiovascular, genitourinary, and nervous systems. This review gives an overview of the functional properties of mammalian TRP channels, describes their roles in acquired and hereditary diseases, and discusses their potential as drug targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bernd Nilius
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| | - Arpad Szallasi
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| |
Collapse
|
29
|
Mignon-Ravix C, Cacciagli P, Choucair N, Popovici C, Missirian C, Milh M, Mégarbané A, Busa T, Julia S, Girard N, Badens C, Sigaudy S, Philip N, Villard L. Intragenic rearrangements in X-linked intellectual deficiency: results of a-CGH in a series of 54 patients and identification of TRPC5 and KLHL15 as potential XLID genes. Am J Med Genet A 2014; 164A:1991-7. [PMID: 24817631 DOI: 10.1002/ajmg.a.36602] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/03/2014] [Indexed: 01/24/2023]
Abstract
High-resolution array comparative genomic hybridization (a-CGH) enables the detection of intragenic rearrangements, such as single exon deletion or duplication. This approach can lead to the identification of new disease genes. We report on the analysis of 54 male patients presenting with intellectual deficiency (ID) and a family history suggesting X-linked (XL) inheritance or maternal skewed X-chromosome inactivation (XCI), using a home-made X-chromosome-specific microarray covering the whole human X-chromosome at high resolution. The majority of patients had whole genome array-CGH prior to the selection and we did not include large rearrangements such as MECP2 and FMR1 duplications. We identified four rearrangements considered as causative or potentially pathogenic, corresponding to a detection rate of 8%. Two CNVs affected known XLID genes and were therefore considered as causative (IL1RAPL1 and OPHN1 intragenic deletions). Two new CNVs were considered as potentially pathogenic as they affected interesting candidates for ID. The first CNV is a deletion of the first exon of the TRPC5 gene, encoding a cation channel implicated in dendrite growth and patterning, in a child presenting with ID and an autism spectrum disorder (ASD). The second CNV is a partial deletion of KLHL15, in a patient with severe ID, epilepsy, and anomalies of cortical development. In both cases, in spite of strong arguments for clinical relevance, we were not able at this stage to confirm pathogenicity of the mutations, and the causality of the variants identified in XLID remains to be confirmed.
Collapse
Affiliation(s)
- Cécile Mignon-Ravix
- Inserm, UMR_S 910, Marseille, France; Aix Marseille Université, GMGF, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
The role of canonical transient receptor potential channels in seizure and excitotoxicity. Cells 2014; 3:288-303. [PMID: 24722470 PMCID: PMC4092853 DOI: 10.3390/cells3020288] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/29/2014] [Accepted: 04/02/2014] [Indexed: 12/11/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels are a family of polymodal cation channels with some degree of Ca2+ permeability. Although initially thought to be channels mediating store-operated Ca2+ influx, TRPC channels can be activated by stimulation of Gq-coupled G-protein coupled receptors, or by an increase in intracellular free Ca2+ concentration. Thus, activation of TRPC channels could be a common downstream event of many signaling pathways that contribute to seizure and excitotoxicity, such as N-methyl-D-aspartate (NMDA) receptor-mediated Ca2+ influx, or metabotropic glutamate receptor activation. Recent studies with genetic ablation of various TRPC family members have demonstrated that TRPC channels, in particular heteromeric TRPC1/4 channels and homomeric TRPC5 channels, play a critical role in both pilocarpine-induced acute seizures and neuronal cell death. However, exact underlying mechanisms remain to be fully elucidated, and selective TRPC modulators and antibodies with better specificity are urgently needed for future research.
Collapse
|
31
|
Abstract
Human canonical transient receptor potential channel 5 (TRPC5) has been cloned from the Xq23 region on chromosome X as a suspect in nonsyndromic mental retardation. TRPC5 is a Ca(2+)-permeable cation channel predominantly expressed in the CNS, including the hippocampus, cerebellum, amygdala, sensory neurons, and retina. It also shows more restricted expression in the periphery, notably in the kidney and cardiovascular system. Homotetrameric TRPC5 channels are primarily activated by receptors coupled to Gq and phospholipase C and/or Gi proteins, but TRPC5 channels may also gate in a store-dependent manner, which requires other partner proteins such TRPC1, STIM1, and Orai1. There is an impressive array of other activators of TRPC5 channels, such as nitric oxide, lysophospholipids, sphingosine-1-phosphate, reduced thioredoxin, protons, lanthanides, and calcium, and many can cause its direct activation. Moreover, TRPC5 shows constitutive activity, and it is responsive to membrane stretch and cold. Thus, TRPC5 channels have significant potential for synergistic activation and may serve as an important focal point in Ca(2+) signalling and electrogenesis. Moreover, TRPC5 functions in partnership with about 60 proteins, including TRPC1, TRPC4, calmodulin, IP3 receptors, NHERF, NCS-1, junctate, stathmin 2, Ca(2+)-binding protein 1, caveolin, and SESTD1, while its desensitisation is mediated by both protein kinases A and C. TRPC5 has a distinct voltage dependence shared only with its closest relative, TRPC4. Its unique N-shaped activation curve underlined by intracellular Mg(2+) block seems to be perfectly "shaped" to trigger action potential discharge, but not to grossly interfere with the action potential shape. The range of biological functions of TRPC5 channels is also impressive, from neurotransmission to control of axon guidance and vascular smooth muscle cell migration and contractility. Recent studies of Trpc5 gene knockouts begin to uncover its roles in fear, anxiety, seizures, and cold sensing.
Collapse
Affiliation(s)
- Alexander V Zholos
- Department of Biophysics, Educational and Scientific Centre "Institute of Biology", Taras Shevchenko Kiev National University, Kiev, 03022, Ukraine,
| |
Collapse
|
32
|
Su B, Ji YS, Sun XL, Liu XH, Chen ZY. Brain-derived neurotrophic factor (BDNF)-induced mitochondrial motility arrest and presynaptic docking contribute to BDNF-enhanced synaptic transmission. J Biol Chem 2013; 289:1213-26. [PMID: 24302729 DOI: 10.1074/jbc.m113.526129] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Appropriate mitochondrial transport and distribution are essential for neurons because of the high energy and Ca(2+) buffering requirements at synapses. Brain-derived neurotrophic factor (BDNF) plays an essential role in regulating synaptic transmission and plasticity. However, whether and how BDNF can regulate mitochondrial transport and distribution are still unclear. Here, we find that in cultured hippocampal neurons, application of BDNF for 15 min decreased the percentage of moving mitochondria in axons, a process dependent on the activation of the TrkB receptor and its downstream PI3K and phospholipase-Cγ signaling pathways. Moreover, the BDNF-induced mitochondrial stopping requires the activation of transient receptor potential canonical 3 and 6 (TRPC3 and TRPC6) channels and elevated intracellular Ca(2+) levels. The Ca(2+) sensor Miro1 plays an important role in this process. Finally, the BDNF-induced mitochondrial stopping leads to the accumulation of more mitochondria at presynaptic sites. Mutant Miro1 lacking the ability to bind Ca(2+) prevents BDNF-induced mitochondrial presynaptic accumulation and synaptic transmission, suggesting that Miro1-mediated mitochondrial motility is involved in BDNF-induced mitochondrial presynaptic docking and neurotransmission. Together, these data suggest that mitochondrial transport and distribution play essential roles in BDNF-mediated synaptic transmission.
Collapse
Affiliation(s)
- Bo Su
- From the Department of Neurobiology, Shandong Provincial Key Laboratory of Mental Disorders, School of Medicine, Shandong University, Jinan, Shandong 250012, China
| | | | | | | | | |
Collapse
|
33
|
Jeon JP, Roh SE, Wie J, Kim J, Kim H, Lee KP, Yang D, Jeon JH, Cho NH, Kim IG, Kang DE, Kim HJ, So I. Activation of TRPC4β by Gαi subunit increases Ca2+ selectivity and controls neurite morphogenesis in cultured hippocampal neuron. Cell Calcium 2013; 54:307-19. [PMID: 24011658 DOI: 10.1016/j.ceca.2013.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 07/27/2013] [Accepted: 07/29/2013] [Indexed: 11/16/2022]
Abstract
The ubiquitous transient receptor potential canonical (TRPC) channels function as non-selective, Ca(2+)-permeable channels. TRPC channels are activated by stimulation of Gαq-PLC-coupled receptors. Here, we report that TRPC4/TRPC5 can be activated by Gαi. We studied the essential role of Gαi subunits in TRPC4 activation and investigated changes in ion selectivity and pore dilation of the TRPC4 channel elicited by the Gαi2 subunit. Activation of TRPC4 by Gαi2 increased Ca2+ permeability and Ca2+ influx through TRPC4 channels. Co-expression of the muscarinic receptor (M2) and TRPC4 in HEK293 cells induced TRPC4-mediated Ca2+ influx. Moreover, both TRPC4β and the TRPC4β-Gαi2 signaling complex induced inhibition of neurite growth and arborization in cultured hippocampal neurons. Cells treated with KN-93, a CaMKII inhibitor, prevented TRPC4- and TRPC4-Gαi2(Q205L)-mediated inhibition of neurite branching and growth. These findings indicate an essential role of Gαi proteins in TRPC4 activation and extend our knowledge of the functional role of TRPC4 in hippocampal neurons.
Collapse
Affiliation(s)
- Jae-Pyo Jeon
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Pomrenze MB, Baratta MV, Rasmus KC, Cadle BA, Nakamura S, Birnbaumer L, Cooper DC. Cocaine self-administration in mice with forebrain knock-down of trpc5 ion channels. F1000Res 2013; 2:53. [PMID: 24358869 PMCID: PMC3790609 DOI: 10.12688/f1000research.2-53.v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/12/2013] [Indexed: 01/13/2023] Open
Abstract
Canonical transient receptor potential (TRPC) channels are a family of non-selective cation channels that play a crucial role in modulating neuronal excitability due to their involvement in intracellular Ca2+ regulation and dendritic growth. TRPC5 channels a) are one of the two most prevalent TRPC channels in the adult rodent brain; b) are densely expressed in deep layer pyramidal neurons of the prefrontal cortex (PFC); and c) modulate neuronal persistent activity necessary for working memory and attention. In order to evaluate the causal role of TRPC5 in motivation/reward-related behaviors, conditional forebrain TRPC5 knock-down (trpc5-KD) mice were generated and trained to nose-poke for intravenous cocaine. Here we present a data set containing the first 6 days of saline or cocaine self-administration in wild type (WT) and trpc5-KD mice. In addition, we also present a data set showing the dose-response to cocaine after both groups had achieved similar levels of cocaine self-administration. Compared to WT mice, trpc5-KD mice exhibited an apparent increase in self-administration on the first day of cocaine testing without prior operant training. There were no apparent differences between WT and trpc5-KD mice for saline responding on the first day of training. Both groups showed similar dose-response sensitivity to cocaine after several days of achieving similar levels of cocaine intake.
Collapse
Affiliation(s)
- Matthew B Pomrenze
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80309, USA
| | - Michael V Baratta
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80309, USA
| | - Kristin C Rasmus
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80309, USA
| | - Brian A Cadle
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80309, USA
| | - Shinya Nakamura
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80309, USA
| | - Lutz Birnbaumer
- National Institute of Environmental Health Science, National Institute of Health, Research Triangle Park, NC, 27709, USA
| | - Donald C Cooper
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80309, USA
| |
Collapse
|