1
|
Kang H, Kim J, Park CH, Jeong B, So I. Direct modulation of TRPC ion channels by Gα proteins. Front Physiol 2024; 15:1362987. [PMID: 38384797 PMCID: PMC10880550 DOI: 10.3389/fphys.2024.1362987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
GPCR-Gi protein pathways are involved in the regulation of vagus muscarinic pathway under physiological conditions and are closely associated with the regulation of internal visceral organs. The muscarinic receptor-operated cationic channel is important in GPCR-Gi protein signal transduction as it decreases heart rate and increases GI rhythm frequency. In the SA node of the heart, acetylcholine binds to the M2 receptor and the released Gβγ activates GIRK (I(K,ACh)) channel, inducing a negative chronotropic action. In gastric smooth muscle, there are two muscarinic acetylcholine receptor (mAChR) subtypes, M2 and M3. M2 receptor activates the muscarinic receptor-operated nonselective cationic current (mIcat, NSCC(ACh)) and induces positive chronotropic effect. Meanwhile, M3 receptor induces hydrolysis of PIP2 and releases DAG and IP3. This IP3 increases intracellular Ca2+ and then leads to contraction of GI smooth muscles. The activation of mIcat is inhibited by anti-Gi/o protein antibodies in GI smooth muscle, indicating the involvement of Gαi/o protein in the activation of mIcat. TRPC4 channel is a molecular candidate for mIcat and can be directly activated by constitutively active Gαi QL proteins. TRPC4 and TRPC5 belong to the same subfamily and both are activated by Gi/o proteins. Initial studies suggested that the binding sites for G protein exist at the rib helix or the CIRB domain of TRPC4/5 channels. However, recent cryo-EM structure showed that IYY58-60 amino acids at ARD of TRPC5 binds with Gi3 protein. Considering the expression of TRPC4/5 in the brain, the direct G protein activation on TRPC4/5 is important in terms of neurophysiology. TRPC4/5 channels are also suggested as a coincidence detector for Gi and Gq pathway as Gq pathway increases intracellular Ca2+ and the increased Ca2+ facilitates the activation of TRPC4/5 channels. More complicated situation would occur when GIRK, KCNQ2/3 (IM) and TRPC4/5 channels are co-activated by stimulation of muscarinic receptors at the acetylcholine-releasing nerve terminals. This review highlights the effects of GPCR-Gi protein pathway, including dopamine, μ-opioid, serotonin, glutamate, GABA, on various oragns, and it emphasizes the importance of considering TRPC4/5 channels as crucial players in the field of neuroscience.
Collapse
Affiliation(s)
- Hana Kang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jinhyeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Christine Haewon Park
- Department of Physiology, University of California, San Francisco, San Francisco, CA, United States
| | - Byeongseok Jeong
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
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
|
3
|
Park CH, Kim J, Lee JE, Kwak M, So I. Pore residues of transient receptor potential channels canonical 1 and 4 heteromer determine channel properties. Am J Physiol Cell Physiol 2023; 325:C42-C51. [PMID: 37212545 DOI: 10.1152/ajpcell.00488.2022] [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: 11/01/2022] [Revised: 04/26/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Transient receptor potential channels canonical 1 and 4 (TRPC1 and TRPC4) are proteins belonging to the same TRPC channel family, and the two are known to form a heterotetrameric channel. TRPC4 can form a homotetrameric, nonselective cation channel by itself, but the involvement of the TRPC1 subunit changes several major characteristics of the channel. In this study, we focused on the pore region (selectivity filter, pore helix, and S6 helix) of TRPC1 and TRPC4 as a determinant of the identity and characteristics of a heteromeric TRPC1/4 channel: decreased calcium permeability of the channel and outward-rectifying current-voltage (I-V) curve. Mutants and chimeras of the pore residues were created, and their currents were recorded using whole cell patch clamp. The lower gate mutants of TRPC4 exhibited diminished calcium permeability as measured by GCaMP6 fluorescence. Also, chimeric channels substituting the pore region of TRPC1 to TRPC4 were made to locate the pore region that is critical in the production of an outward-rectifying I-V curve characteristic of TRPC1/4 heteromeric channels.NEW & NOTEWORTHY Heteromer research has been a challenging field due to lack of structural studies. Using chimeras and single mutants, we present evidence that the pore region of TRPC1/4 heteromer contributes to determining the channel's characteristics such as calcium permeability, I-V curve, and conductance.
Collapse
Affiliation(s)
- Christine Haewon Park
- Department of Physiology, Seoul National University School of Medicine, Seoul, Korea
| | - Jinsung Kim
- Department of Physiology, Seoul National University School of Medicine, Seoul, Korea
| | - Jung Eun Lee
- Department of Physiology, Seoul National University School of Medicine, Seoul, Korea
| | - Misun Kwak
- Department of Physiology, Seoul National University School of Medicine, Seoul, Korea
| | - Insuk So
- Department of Physiology, Seoul National University School of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Hong DK, Kho AR, Lee SH, Kang BS, Park MK, Choi BY, Suh SW. Pathophysiological Roles of Transient Receptor Potential (Trp) Channels and Zinc Toxicity in Brain Disease. Int J Mol Sci 2023; 24:ijms24076665. [PMID: 37047637 PMCID: PMC10094935 DOI: 10.3390/ijms24076665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023] Open
Abstract
Maintaining the correct ionic gradient from extracellular to intracellular space via several membrane-bound transporters is critical for maintaining overall cellular homeostasis. One of these transporters is the transient receptor potential (TRP) channel family that consists of six putative transmembrane segments systemically expressed in mammalian tissues. Upon the activation of TRP channels by brain disease, several cations are translocated through TRP channels. Brain disease, especially ischemic stroke, epilepsy, and traumatic brain injury, triggers the dysregulation of ionic gradients and promotes the excessive release of neuro-transmitters and zinc. The divalent metal cation zinc is highly distributed in the brain and is specifically located in the pre-synaptic vesicles as free ions, usually existing in cytoplasm bound with metallothionein. Although adequate zinc is essential for regulating diverse physiological functions, the brain-disease-induced excessive release and translocation of zinc causes cell damage, including oxidative stress, apoptotic cascades, and disturbances in energy metabolism. Therefore, the regulation of zinc homeostasis following brain disease is critical for the prevention of brain damage. In this review, we summarize recent experimental research findings regarding how TRP channels (mainly TRPC and TRPM) and zinc are regulated in animal brain-disease models of global cerebral ischemia, epilepsy, and traumatic brain injury. The blockade of zinc translocation via the inhibition of TRPC and TRPM channels using known channel antagonists, was shown to be neuroprotective in brain disease. The regulation of both zinc and TRP channels may serve as targets for treating and preventing neuronal death.
Collapse
Affiliation(s)
- Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - A Ra Kho
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, College of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Bo Young Choi
- Department of Physical Education, Hallym University, Chuncheon 24252, Republic of Korea
- Institute of Sport Science, Hallym University, Chuncheon 24252, Republic of Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
5
|
Jeong B, Sung TS, Jeon D, Park KJ, Jun JY, So I, Hong C. Inhibition of TRPC4 channel activity in colonic myocytes by tricyclic antidepressants disrupts colonic motility causing constipation. J Cell Mol Med 2022; 26:4911-4923. [PMID: 35560982 PMCID: PMC9549500 DOI: 10.1111/jcmm.17348] [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: 11/03/2021] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/27/2022] Open
Abstract
Tricyclic antidepressants (TCAs) have been used to treat depression and were recently approved for treating irritable bowel syndrome (IBS) patients with severe or refractory IBS symptoms. However, the molecular mechanism of TCA action in the gastrointestinal (GI) tract remains poorly understood. Transient receptor potential channel canonical type 4 (TRPC4), which is a Ca2+‐permeable nonselective cation channel, is a critical regulator of GI excitability. Herein, we investigated whether TCA modulates TRPC4 channel activity and which mechanism in colonic myocytes consequently causes constipation. To prove the clinical benefit in patients with diarrhoea caused by TCA treatment, we performed mechanical tension recording of repetitive motor pattern (RMP) in segment, electric field stimulation (EFS)‐induced and spontaneous contractions in isolated muscle strips. From these recordings, we observed that all TCA compounds significantly inhibited contractions of colonic motility in human. To determine the contribution of TRPC4 to colonic motility, we measured the electrical activity of heterologous or endogenous TRPC4 by TCAs using the patch clamp technique in HEK293 cells and murine colonic myocytes. In TRPC4‐overexpressed HEK cells, we observed TCA‐evoked direct inhibition of TRPC4. Compared with TRPC4‐knockout mice, we identified that muscarinic cationic current (mIcat) was suppressed through TRPC4 inhibition by TCA in isolated murine colonic myocytes. Collectively, we suggest that TCA action is responsible for the inhibition of TRPC4 channels in colonic myocytes, ultimately causing constipation. These findings provide clinical insights into abnormal intestinal motility and medical interventions aimed at IBS therapy.
Collapse
Affiliation(s)
- Byeongseok Jeong
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Tae Sik Sung
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Dongju Jeon
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Kyu Joo Park
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jae Yeoul Jun
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Insuk So
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Chansik Hong
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| |
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: 25] [Impact Index Per Article: 8.3] [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
|
Jeon J, Bu F, Sun G, Tian JB, Ting SM, Li J, Aronowski J, Birnbaumer L, Freichel M, Zhu MX. Contribution of TRPC Channels in Neuronal Excitotoxicity Associated With Neurodegenerative Disease and Ischemic Stroke. Front Cell Dev Biol 2021; 8:618663. [PMID: 33490083 PMCID: PMC7820370 DOI: 10.3389/fcell.2020.618663] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
The seven canonical members of transient receptor potential (TRPC) proteins form cation channels that evoke membrane depolarization and intracellular calcium concentration ([Ca2+] i ) rise, which are not only important for regulating cell function but their deregulation can also lead to cell damage. Recent studies have implicated complex roles of TRPC channels in neurodegenerative diseases including ischemic stroke. Brain ischemia reduces oxygen and glucose supply to neurons, i.e., Oxygen and Glucose Deprivation (OGD), resulting in [Ca2+] i elevation, ion dyshomeostasis, and excitotoxicity, which are also common in many forms of neurodegenerative diseases. Although ionotropic glutamate receptors, e.g., N-methyl-D-aspartate receptors, are well established to play roles in excitotoxicity, the contribution of metabotropic glutamate receptors and their downstream effectors, i.e., TRPC channels, should not be neglected. Here, we summarize the current findings about contributions of TRPC channels in neurodegenerative diseases, with a focus on OGD-induced neuronal death and rodent models of cerebral ischemia/reperfusion. TRPC channels play both detrimental and protective roles to neurodegeneration depending on the TRPC subtype and specific pathological conditions involved. When illustrated the mechanisms by which TRPC channels are involved in neuronal survival or death seem differ greatly, implicating diverse and complex regulation. We provide our own data showing that TRPC1/C4/C5, especially TRPC4, may be generally detrimental in OGD and cerebral ischemia/reperfusion. We propose that although TRPC channels significantly contribute to ischemic neuronal death, detailed mechanisms and specific roles of TRPC subtypes in brain injury at different stages of ischemia/reperfusion and in different brain regions need to be carefully and systematically investigated.
Collapse
Affiliation(s)
- Jaepyo Jeon
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Fan Bu
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Guanghua Sun
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jin-Bin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shun-Ming Ting
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jun Li
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Lutz Birnbaumer
- Institute for Biomedical Research (BIOMED UCA-CONICET), Buenos Aires, Argentina.,School of Medical Sciences, Catholic University of Argentina (UCA), Buenos Aires, Argentina.,Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Marc Freichel
- Department of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
8
|
Roles of TRP Channels in Neurological Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7289194. [PMID: 32963700 PMCID: PMC7492880 DOI: 10.1155/2020/7289194] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/02/2020] [Indexed: 11/17/2022]
Abstract
Transient receptor potential (TRP) proteins consist of a superfamily of cation channels that have been involved in diverse physiological processes in the brain as well as in the pathogenesis of neurological disease. TRP channels are widely expressed in the brain, including neurons and glial cells, as well as in the cerebral vascular endothelium and smooth muscle. Members of this channel superfamily show a wide variety of mechanisms ranging from ligand binding to voltage, physical, and chemical stimuli, implying the promising therapeutic potential of TRP in neurological diseases. In this review, we focus on the physiological functions of TRP channels in the brain and the pathological roles in neurological disorders to explore future potential neuroprotective strategies.
Collapse
|
9
|
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: 0.5] [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
|
10
|
Kepura F, Braun E, Dietrich A, Plant TD. TRPC1 Regulates the Activity of a Voltage-Dependent Nonselective Cation Current in Hippocampal CA1 Neurons. Cells 2020; 9:cells9020459. [PMID: 32085504 PMCID: PMC7072794 DOI: 10.3390/cells9020459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/26/2022] Open
Abstract
The cation channel subunit TRPC1 is strongly expressed in central neurons including neurons in the CA1 region of the hippocampus where it forms complexes with TRPC4 and TRPC5. To investigate the functional role of TRPC1 in these neurons and in channel function, we compared current responses to group I metabotropic glutamate receptor (mGluR I) activation and looked for major differences in dendritic morphology in neurons from TRPC1+/+ and TRPC1-/- mice. mGluR I stimulation resulted in the activation of a voltage-dependent nonselective cation current in both genotypes. Deletion of TRPC1 resulted in a modification of the shape of the current-voltage relationship, leading to an inward current increase. In current clamp recordings, the percentage of neurons that responded to depolarization in the presence of an mGluR I agonist with a plateau potential was increased in TRPC1-/- mice. There was also a small increase in the minor population of CA1 neurons that have more than one apical dendrite in TRPC1-/- mice. We conclude that TRPC1 has an inhibitory effect on receptor-operated nonselective cation channels in hippocampal CA1 neurons probably as a result of heterotetramer formation with other TRPC isoforms, and that TRPC1 deletion has only minor effects on dendritic morphology.
Collapse
Affiliation(s)
- Frauke Kepura
- Pharmakologisches Institut, BPC-Marburg, Fachbereich Medizin, Philipps-Universität Marburg, Karl-von-Frisch-Straße 2, 35043 Marburg, Germany; (F.K.); (E.B.); (A.D.)
| | - Eva Braun
- Pharmakologisches Institut, BPC-Marburg, Fachbereich Medizin, Philipps-Universität Marburg, Karl-von-Frisch-Straße 2, 35043 Marburg, Germany; (F.K.); (E.B.); (A.D.)
| | - Alexander Dietrich
- Pharmakologisches Institut, BPC-Marburg, Fachbereich Medizin, Philipps-Universität Marburg, Karl-von-Frisch-Straße 2, 35043 Marburg, Germany; (F.K.); (E.B.); (A.D.)
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 München, Germany
| | - Tim D. Plant
- Pharmakologisches Institut, BPC-Marburg, Fachbereich Medizin, Philipps-Universität Marburg, Karl-von-Frisch-Straße 2, 35043 Marburg, Germany; (F.K.); (E.B.); (A.D.)
- Center for Mind, Brain and Behavior, Philipps-Universität Marburg, 35032 Marburg, Germany
- Correspondence: ; Tel.: +49-6421-28-65038
| |
Collapse
|
11
|
Structure-Function Relationship and Physiological Roles of Transient Receptor Potential Canonical (TRPC) 4 and 5 Channels. Cells 2019; 9:cells9010073. [PMID: 31892199 PMCID: PMC7017149 DOI: 10.3390/cells9010073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/24/2019] [Accepted: 12/24/2019] [Indexed: 12/11/2022] Open
Abstract
The study of the structure–function relationship of ion channels has been one of the most challenging goals in contemporary physiology. Revelation of the three-dimensional (3D) structure of ion channels has facilitated our understanding of many of the submolecular mechanisms inside ion channels, such as selective permeability, voltage dependency, agonist binding, and inter-subunit multimerization. Identifying the structure–function relationship of the ion channels is clinically important as well since only such knowledge can imbue potential therapeutics with practical possibilities. In a sense, recent advances in the understanding of the structure–relationship of transient receptor potential canonical (TRPC) channels look promising since human TRPC channels are calcium-permeable, non-selective cation channels expressed in many tissues such as the gastrointestinal (GI) tract, kidney, heart, vasculature, and brain. TRPC channels are known to regulate GI contractility and motility, pulmonary hypertension, right ventricular hypertrophy, podocyte injury, seizure, fear, anxiety-like behavior, and many others. In this article, we tried to elaborate recent findings of Cryo-EM (cryogenic-electron microscopy) based structural information of TRPC 4 and 5 channels and domain-specific functions of the channel, such as G-protein mediated activation mechanism, extracellular modification of the channel, homo/hetero-tetramerization, and pharmacological gating mechanisms.
Collapse
|
12
|
Kim J, Ko J, Myeong J, Kwak M, Hong C, So I. TRPC1 as a negative regulator for TRPC4 and TRPC5 channels. Pflugers Arch 2019; 471:1045-1053. [PMID: 31222490 DOI: 10.1007/s00424-019-02289-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/17/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
Abstract
Transient receptor potential canonical (TRPC) channels are calcium permeable, non-selective cation channels with wide tissue-specific distribution. Among 7 TRPC channels, TRPC 1/4/5 and TRPC3/6/7 are subdivided based on amino acid sequence homology. TRPC4 and TRPC5 channels exhibit cationic current with homotetrameric form, but they also form heterotetrameric channel such as TRPC1/4 or TRPC1/5 once TRPC1 is incorporated. The expression of TRPC1 is ubiquitous whereas the expressions of TRPC4 and TRPC5 are rather focused in nervous system. With the help of conditional knock-out of TPRC1, 4 and/or 5 genes, TRPC channels made of these constituents are reported to be involved in various pathophysiological functions such as seizure, anxiety-like behaviour, fear, Huntington's disease, Parkinson's disease and many others. In heterologous expression system, many issues such as activation mechanism, stoichiometry and relative cation permeabilites of homomeric or heteromeric channels have been addressed. In this review, we discussed the role of TRPC1 channel per se in plasma membrane, role of TRPC1 in heterotetrameric conformation (TRPC1/4 or TRPC1/5) and relationship between TRPC1/4/5 channels, calcium influx and voltage-gated calcium channels.
Collapse
Affiliation(s)
- Jinsung Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Juyeon Ko
- Department of Physiology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jongyun Myeong
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Misun Kwak
- Department of Physiology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Chansik Hong
- Department of Physiology, College of Medicine, Chosun University, Kwangju, South Korea
| | - Insuk So
- Department of Physiology, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
13
|
Jeong S, Ko J, Kim M, Park KC, Park EYJ, Kim J, Baik Y, Wie J, Cho AE, Jeon JH, So I. Englerin A-sensing charged residues for transient receptor potential canonical 5 channel activation. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2019; 23:191-201. [PMID: 31080350 PMCID: PMC6488704 DOI: 10.4196/kjpp.2019.23.3.191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 11/17/2022]
Abstract
The transient receptor potential canonical (TRPC) 5 channel, known as a nonselective cation channel, has a crucial role in calcium influx. TRPC5 has been reported to be activated by muscarinic receptor activation and extracellular pH change and inhibited by the protein kinase C pathway. Recent studies have also suggested that TRPC5 is extracellularly activated by englerin A (EA), but the mechanism remains unclear. The purpose of this study is to identify the EA-interaction sites in TRPC5 and thereby clarify the mechanism of TRPC5 activation. TRPC5 channels are over-expressed in human embryonic kidney (HEK293) cells. TRPC5 mutants were generated by site-directed mutagenesis. The whole-cell patch-clamp configuration was used to record TRPC5 currents. Western analysis was also performed to observe the expression of TRPC5 mutants. To identify the EA-interaction site in TRPC5, we first generated pore mutants. When screening the mutants with EA, we observed the EA-induced current increases of TRPC5 abolished in K554N, H594N, and E598Q mutants. The current increases of other mutants were reduced in different levels. We also examined the functional intactness of the mutants that had no effect by EA with TRPC5 agonists, such as carbachol or GTPγS. Our results suggest that the three residues, Lys-554, His-594, and Glu-598, in TRPC5 might be responsible for direct interaction with EA, inducing the channel activation. We also suggest that although other pore residues are not critical, they could partly contribute to the EA-induced channel activation.
Collapse
Affiliation(s)
- SeungJoo Jeong
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Juyeon Ko
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Minji Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Ki Chul Park
- Department of Bioinformatics, Korea University, Sejong 30019, Korea
| | - Eunice Yon June Park
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jinsung Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Youngjoo Baik
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jinhong Wie
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Art E Cho
- Department of Bioinformatics, Korea University, Sejong 30019, Korea
| | - Ju-Hong Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
14
|
Garcia C, Maurel-Ribes A, Nauze M, N'Guyen D, Martinez LO, Payrastre B, Sénard JM, Galés C, Pons V. Deciphering biased inverse agonism of cangrelor and ticagrelor at P2Y 12 receptor. Cell Mol Life Sci 2019; 76:561-576. [PMID: 30406277 PMCID: PMC11105710 DOI: 10.1007/s00018-018-2960-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 10/15/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
P2Y12 receptor (P2Y12-R) is one of the major targets for drug inhibiting platelet aggregation in the treatment/prevention of arterial thrombosis. However, the clinical use of P2Y12-R antagonists faces some limitations, such as a delayed onset of action (clopidogrel) or adverse effect profile (ticagrelor, cangrelor), justifying the development of a new generation of P2Y12-R antagonists with a better clinical benefit-risk balance. Although the recent concept of biased agonism offers the possibility to alleviate undesirable adverse effects while preserving therapeutic outcomes, it has never been explored at P2Y12-R. For the first time, using highly sensitive BRET2-based probes, we accurately delineated biased ligand efficacy at P2Y12-R in living HEK293T cells on G protein activation and downstream effectors. We demonstrated that P2Y12-R displayed constitutive Gi/o-dependent signaling that is impaired by the R122C mutation, previously associated with a bleeding disorder. More importantly, we reported the biased inverse agonist efficacy of cangrelor and ticagrelor that could underlie their clinical efficacy. Our study points out that constitutive P2Y12-R signaling is a normal feature of the receptor that might be essential for platelets to respond faster to a vessel injury. From a therapeutic standpoint, our data suggest that the beneficial advantages of antiplatelet drugs might be more related to inverse agonism at P2Y12-R than to antagonism of ADP-mediated signaling. In the future, deciphering P2Y12-R constitutive activity should allow the discovery of more selective biased P2Y12-R blockers demonstrating therapeutic advantages over classical antiplatelet drugs by improving therapeutic outcomes and concomitantly relieving undesirable adverse effects.
Collapse
Affiliation(s)
- Cédric Garcia
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, 31000, Toulouse, France
| | - Agnès Maurel-Ribes
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, 31000, Toulouse, France
| | - Michel Nauze
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 04, France
| | - Du N'Guyen
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 04, France
| | - Laurent O Martinez
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 04, France
| | - Bernard Payrastre
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, 31000, Toulouse, France
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 04, France
| | - Jean-Michel Sénard
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 04, France
- Service de Pharmacologie Clinique, Centre Hospitalier Universitaire de Toulouse, Université de Toulouse, 31000, Toulouse, France
| | - Céline Galés
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 04, France
| | - Véronique Pons
- INSERM U1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, 1 Avenue Jean Poulhès, BP 84225, 31432, Toulouse Cedex 04, France.
| |
Collapse
|
15
|
Toktanis G, Kaya-Sezginer E, Yilmaz-Oral D, Gur S. Potential therapeutic value of transient receptor potential channels in male urogenital system. Pflugers Arch 2018; 470:1583-1596. [PMID: 30194638 DOI: 10.1007/s00424-018-2188-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/11/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022]
Abstract
Transient receptor potential (TRP) channels comprise a family of cation channels implicated in a variety of cellular processes including light, mechanical or chemical stimuli, temperature, pH, or osmolarity. TRP channel proteins are a diverse family of proteins that are expressed in many tissues. We debated our recent knowledge about the expression, function, and regulation of TRP channels in the different parts of the male urogenital system in health and disease. Emerging evidence suggests that dysfunction of TRP channels significantly contributes to the pathophysiology of urogenital diseases. So far, there are many efforts underway to determine if these channels can be used as drug targets to reverse declines in male urogenital function. Furthermore, developing safe and efficacious TRP channel modulators is warranted for male urogenital disorders in a clinical setting.
Collapse
Affiliation(s)
| | - Ecem Kaya-Sezginer
- Faculty of Pharmacy, Department of Biochemistry and Pharmacology, Ankara University, Tandogan, 06100, Ankara, Turkey
| | - Didem Yilmaz-Oral
- Faculty of Pharmacy, Department of Biochemistry and Pharmacology, Ankara University, Tandogan, 06100, Ankara, Turkey.,Faculty of Pharmacy, Department of Pharmacology, Cukurova University, Adana, Turkey
| | - Serap Gur
- Faculty of Pharmacy, Department of Biochemistry and Pharmacology, Ankara University, Tandogan, 06100, Ankara, Turkey.
| |
Collapse
|
16
|
Zheng W, Hu R, Cai R, Hofmann L, Hu Q, Fatehi M, Long W, Kong T, Tang J, Light P, Flockerzi V, Cao Y, Chen X. Identification and characterization of hydrophobic gate residues in TRP channels. FASEB J 2018; 32:639-653. [DOI: 10.1096/fj.201700599rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Wang Zheng
- Institute of Biomedical and Pharmaceutical SciencesKey Laboratory of Fermentation Engineering of Ministry of EducationCollege of BioengineeringHubei University of TechnologyWuhanChina
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Ruikun Hu
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Ruiqi Cai
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Laura Hofmann
- Experimentelle und Klinische Pharmakologie und ToxikologieUniversität des SaarlandesHomburgGermany
| | - Qiaolin Hu
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Mohammad Fatehi
- Department of PharmacologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Wentong Long
- Department of PharmacologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Tim Kong
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| | - Jingfeng Tang
- Institute of Biomedical and Pharmaceutical SciencesKey Laboratory of Fermentation Engineering of Ministry of EducationCollege of BioengineeringHubei University of TechnologyWuhanChina
| | - Peter Light
- Department of PharmacologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und ToxikologieUniversität des SaarlandesHomburgGermany
| | - Ying Cao
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xing‐Zhen Chen
- Membrane Protein Disease Research GroupDepartment of PhysiologyTongji UniversityShanghaiChina
| |
Collapse
|
17
|
Bröker-Lai J, Kollewe A, Schindeldecker B, Pohle J, Nguyen Chi V, Mathar I, Guzman R, Schwarz Y, Lai A, Weißgerber P, Schwegler H, Dietrich A, Both M, Sprengel R, Draguhn A, Köhr G, Fakler B, Flockerzi V, Bruns D, Freichel M. Heteromeric channels formed by TRPC1, TRPC4 and TRPC5 define hippocampal synaptic transmission and working memory. EMBO J 2017; 36:2770-2789. [PMID: 28790178 DOI: 10.15252/embj.201696369] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 07/01/2017] [Accepted: 07/07/2017] [Indexed: 12/30/2022] Open
Abstract
Canonical transient receptor potential (TRPC) channels influence various neuronal functions. Using quantitative high-resolution mass spectrometry, we demonstrate that TRPC1, TRPC4, and TRPC5 assemble into heteromultimers with each other, but not with other TRP family members in the mouse brain and hippocampus. In hippocampal neurons from Trpc1/Trpc4/Trpc5-triple-knockout (Trpc1/4/5-/-) mice, lacking any TRPC1-, TRPC4-, or TRPC5-containing channels, action potential-triggered excitatory postsynaptic currents (EPSCs) were significantly reduced, whereas frequency, amplitude, and kinetics of quantal miniature EPSC signaling remained unchanged. Likewise, evoked postsynaptic responses in hippocampal slice recordings and transient potentiation after tetanic stimulation were decreased. In vivo, Trpc1/4/5-/- mice displayed impaired cross-frequency coupling in hippocampal networks and deficits in spatial working memory, while spatial reference memory was unaltered. Trpc1/4/5-/- animals also exhibited deficiencies in adapting to a new challenge in a relearning task. Our results indicate the contribution of heteromultimeric channels from TRPC1, TRPC4, and TRPC5 subunits to the regulation of mechanisms underlying spatial working memory and flexible relearning by facilitating proper synaptic transmission in hippocampal neurons.
Collapse
Affiliation(s)
- Jenny Bröker-Lai
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Astrid Kollewe
- Institute of Physiology, University of Freiburg, Freiburg, Germany
| | - Barbara Schindeldecker
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Jörg Pohle
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,Physiology of Neural Networks, Psychiatry/Psychopharmacology, Central Institute of Mental Health, J5, Heidelberg University, Mannheim, Germany
| | - Vivan Nguyen Chi
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Ilka Mathar
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Raul Guzman
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Yvonne Schwarz
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Alan Lai
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Petra Weißgerber
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | | | - Alexander Dietrich
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, München, Germany
| | - Martin Both
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Rolf Sprengel
- Max Planck Research Group of the Max Planck Institute for Medical Research at the Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Georg Köhr
- Physiology of Neural Networks, Psychiatry/Psychopharmacology, Central Institute of Mental Health, J5, Heidelberg University, Mannheim, Germany
| | - Bernd Fakler
- Institute of Physiology, University of Freiburg, Freiburg, Germany.,BIOSS, Center for Biological Signaling Studies, University of Freiburg, Freiburg, Germany‡
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Dieter Bruns
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
18
|
Wang LK, Chen X, Zhang CQ, Liang C, Wei YJ, Yue J, Liu SY, Yang H. Elevated Expression of TRPC4 in Cortical Lesions of Focal Cortical Dysplasia II and Tuberous Sclerosis Complex. J Mol Neurosci 2017; 62:222-231. [PMID: 28455787 DOI: 10.1007/s12031-017-0923-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/17/2017] [Indexed: 12/12/2022]
Abstract
Focal cortical dysplasia type II (FCD II) and tuberous sclerosis complex (TSC) are well-known causes of chronic refractory epilepsy in children. Canonical transient receptor potential channels (TRPCs) are non-selective cation channels that are commonly activated by phospholipase C (PLC) stimulation. Previous studies found that TRPC4 may participate in the process of epileptogenesis. This study aimed to examine the expression and distribution of TRPC4 in FCD II (n = 24) and TSC (n = 11) surgical specimens compared with that in age-matched autopsy control samples (n = 12). We found that the protein levels of TRPC4 and its upstream factor, PLC delta 1 (PLCD1), were elevated in FCD II and TSC samples compared to those of control samples. Immunohistochemistry assays revealed that TRPC4 staining was stronger in malformed cells, such as dysmorphic neurons, balloon cells and giant cells. Moderate-to-strong staining of the upstream factor PLCD1 was also identified in abnormal neurons. Moreover, double immunofluorescence staining revealed that TRPC4 was colocalised with glutamatergic and GABAergic neuron markers. Taken together, our results indicate that overexpression of TRPC4 protein may be involved in the epileptogenesis of FCD II and TSC.
Collapse
Affiliation(s)
- Lu-Kang Wang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of the People's Liberation Army Chengdu Military Region, Chengdu, Sichuan, 610083, China
| | - Chun-Qing Zhang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Chao Liang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Yu-Jia Wei
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Jiong Yue
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Shi-Yong Liu
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China
| | - Hui Yang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing, 400037, China.
| |
Collapse
|
19
|
The regulation of transient receptor potential canonical 4 (TRPC4) channel by phosphodiesterase 5 inhibitor via the cyclic guanosine 3'5'-monophosphate. Pflugers Arch 2017; 469:693-702. [PMID: 28124739 DOI: 10.1007/s00424-017-1937-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/28/2016] [Accepted: 01/16/2017] [Indexed: 02/07/2023]
Abstract
The transient receptor potential (TRP) protein superfamily consists of a diverse group of cation channels that bear structural similarities to the fruit fly Drosophila TRP. The TRP superfamily is distinct from other groups of ion channels in displaying a large diversity in ion selectivity, modes of activation, and physiological functions. Classical TRP (transient receptor potential canonical (TRPC)) channels are activated by stimulation of Gq-PLC-coupled receptors and modulated by phosphorylation. The cyclic guanosine monophosphate (cGMP)-PKG pathway is involved in the regulation of TRPC3 and TRPC6 channels. Phosphodiesterase (PDE) 5 inhibitor induced muscle relaxation in corporal smooth muscle cells and was used to treat erectile dysfunction by inhibiting cGMP degradation. Here, we report the functional relationship between TRPC4 and cGMP. In human embryonic kidney (HEK) 293 cells overexpressing TRPC4, cGMP selectively activated TRPC4 channels and increased cytosolic calcium level through TRPC4 channel. We investigated phosphorylation sites in TRPC4 channels and identified S688 as an important phosphorylation site for the cGMP-PKG pathway. Cyclic GMP also activated TRPC4-like current with doubly rectifying current-voltage relationship in prostate smooth muscle cell lines. Taken together, these results show that TRPC4 is phosphorylated by the cGMP-PKG pathway and might be an important target for modulating prostate function by PDE5 inhibitors.
Collapse
|
20
|
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: 0.6] [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
|
21
|
Ko J, Myeong J, Yang D, So I. Calcium permeability of transient receptor potential canonical (TRPC) 4 channels measured by TRPC4-GCaMP6s. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 21:133-140. [PMID: 28066150 PMCID: PMC5214905 DOI: 10.4196/kjpp.2017.21.1.133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 11/24/2016] [Accepted: 11/24/2016] [Indexed: 12/16/2022]
Abstract
Conflicting evidence has been obtained regarding whether transient receptor potential cation channels (TRPC) are store-operated channels (SOCs) or receptor-operated channels (ROCs). Moreover, the Ca/Na permeability ratio differs depending on whether the current-voltage (I-V) curve has a doubly rectifying shape or inward rectifying shape. To investigate the calcium permeability of TRPC4 channels, we attached GCaMP6s to TRPC4 and simultaneously measured the current and calcium signals. A TRPC4 specific activator, (–)-englerin A, induced both current and calcium fluorescence with the similar time course. Muscarinic receptor stimulator, carbachol, also induced both current and calcium fluorescence with the similar time course. By forming heteromers with TRPC4, TRPC1 significantly reduced the inward current with outward rectifying I-V curve, which also caused the decrease of calcium fluorescence intensity. These results suggest that GCaMP6s attached to TRPC4 can detect slight calcium changes near TRPC4 channels. Consequently, TRPC4-GCaMP6s can be a useful tool for testing the calcium permeability of TRPC4 channels.
Collapse
Affiliation(s)
- Juyeon Ko
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jongyun Myeong
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Dongki Yang
- Department of Physiology, College of Medicine, Gachon University, Incheon 21936, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
22
|
Bissinger R, Lang E, Ghashghaeinia M, Singh Y, Zelenak C, Fehrenbacher B, Honisch S, Chen H, Fakhri H, Umbach AT, Liu G, Rexhepaj R, Liu G, Schaller M, Mack AF, Lupescu A, Birnbaumer L, Lang F, Qadri SM. Blunted apoptosis of erythrocytes in mice deficient in the heterotrimeric G-protein subunit Gαi2. Sci Rep 2016; 6:30925. [PMID: 27499046 PMCID: PMC4976336 DOI: 10.1038/srep30925] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/11/2016] [Indexed: 01/09/2023] Open
Abstract
Putative functions of the heterotrimeric G-protein subunit Gαi2-dependent signaling include ion channel regulation, cell differentiation, proliferation and apoptosis. Erythrocytes may, similar to apoptosis of nucleated cells, undergo eryptosis, characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine (PS) exposure. Eryptosis may be triggered by increased cytosolic Ca2+ activity and ceramide. In the present study, we show that Gαi2 is expressed in both murine and human erythrocytes and further examined the survival of erythrocytes drawn from Gαi2-deficient mice (Gαi2−/−) and corresponding wild-type mice (Gαi2+/+). Our data show that plasma erythropoietin levels, erythrocyte maturation markers, erythrocyte counts, hematocrit and hemoglobin concentration were similar in Gαi2−/− and Gαi2+/+ mice but the mean corpuscular volume was significantly larger in Gαi2−/− mice. Spontaneous PS exposure of circulating Gαi2−/− erythrocytes was significantly lower than that of circulating Gαi2+/+ erythrocytes. PS exposure was significantly lower in Gαi2−/− than in Gαi2+/+ erythrocytes following ex vivo exposure to hyperosmotic shock, bacterial sphingomyelinase or C6 ceramide. Erythrocyte Gαi2 deficiency further attenuated hyperosmotic shock-induced increase of cytosolic Ca2+ activity and cell shrinkage. Moreover, Gαi2−/− erythrocytes were more resistant to osmosensitive hemolysis as compared to Gαi2+/+ erythrocytes. In conclusion, Gαi2 deficiency in erythrocytes confers partial protection against suicidal cell death.
Collapse
Affiliation(s)
- Rosi Bissinger
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Elisabeth Lang
- Department of Gastroenterology, Hepatology and Infectious Diseases, University of Duesseldorf, Germany
| | - Mehrdad Ghashghaeinia
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Yogesh Singh
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Christine Zelenak
- Department of Internal Medicine, Charité Medical University, Berlin, Germany
| | | | - Sabina Honisch
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Hong Chen
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Hajar Fakhri
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Anja T Umbach
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Guilai Liu
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Rexhep Rexhepaj
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany.,Institute of Biochemistry and Molecular Biology, University of Bonn, Germany
| | - Guoxing Liu
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | | | | | - Adrian Lupescu
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Florian Lang
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany
| | - Syed M Qadri
- Institute of Cardiology, Vascular Medicine and Physiology, University of Tuebingen, Germany.,Institute of Biomedical Research (BIOMED), School of Medical Sciences, Catholic University of Argentina, Buenos Aires, Argentina.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
23
|
Galandrin S, Onfroy L, Poirot MC, Sénard JM, Galés C. Delineating biased ligand efficacy at 7TM receptors from an experimental perspective. Int J Biochem Cell Biol 2016; 77:251-63. [PMID: 27107932 DOI: 10.1016/j.biocel.2016.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 12/17/2022]
Abstract
During the last 10 years, the concept of "biased agonism" also called "functional selectivity" swamped the pharmacology of 7 transmembrane receptors and paved the way for developing signaling pathway-selective drugs with increased efficacy and less adverse effects. Initially thought to select the activation of only a subset of the signaling pathways by the reference agonist, bias ligands revealed higher complexity as they have been shown to stabilize variable receptor conformations that associate with distinct signaling events from the reference. Today, one major challenge relies on the in vitro determination of the bias and classification of these ligands, as a prerequisite for future in vivo and clinical translation. In this review, current experimental considerations for the bias evaluation related to choice of the cellular model, of the signaling pathway as well as of the assays are presented and discussed.
Collapse
Affiliation(s)
- Ségolène Galandrin
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France
| | - Lauriane Onfroy
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France
| | - Mathias Charles Poirot
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France
| | - Jean-Michel Sénard
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France; Service de Pharmacologie Clinique, Faculté de médecine, Centre Hospitalier Universitaire de Toulouse, Université de Toulouse, F-31000 Toulouse, France
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM, UMR 1048, Université Toulouse, F-31432 Toulouse, France.
| |
Collapse
|
24
|
Shin DM, Son A, Park S, Kim MS, Ahuja M, Muallem S. The TRPCs, Orais and STIMs in ER/PM Junctions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:47-66. [PMID: 27161224 DOI: 10.1007/978-3-319-26974-0_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The Ca(2+) second messenger is initiated at ER/PM junctions and propagates into the cell interior to convey the receptor information. The signal is maintained by Ca(2+) influx across the plasma membrane through the Orai and TRPC channels. These Ca(2+) influx channels form complexes at ER/PM junctions with the ER Ca(2+) sensor STIM1, which activates the channels. The function of STIM1 is modulated by other STIM isoforms like STIM1L, STIM2 and STIM2.1/STIM2β and by SARAF, which mediates the Ca(2+)-dependent inhibition of Orai channels. The ER/PM junctions are formed at membrane contact sites by tethering proteins that generate several types of ER/PM junctions, such as PI(4,5)P2-poor and PI(4,5)P2-rich domains. This chapter discusses several properties of the TRPC channels, the Orai channels and the STIMs, their key interacting proteins and how interaction of the STIMs with the channels gates their activity. The chapter closes by highlighting open questions and potential future directions in this field.
Collapse
Affiliation(s)
- Dong Min Shin
- Department of Oral Biology, BK 21 PLUS Project, Yonsei University College of Dentistry, Seoul, 120-752, South Korea.
| | - Aran Son
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, 20892, USA
| | - Seonghee Park
- Department of Physiology, School of Medicine, EwhaWomans University, 911-1 Mok-6-dong, Yang Chun-gu, Seoul, 158-710, South Korea
| | - Min Seuk Kim
- Department of Oral Physiology, School of Dentistry, Wonkwang University, Iksan City, Jeonbuk, South Korea
| | - Malini Ahuja
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, 20892, USA
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
25
|
Ross CL, Teli T, Harrison BS. Effect of electromagnetic field on cyclic adenosine monophosphate (cAMP) in a human mu-opioid receptor cell model. Electromagn Biol Med 2015; 35:206-13. [PMID: 26151161 DOI: 10.3109/15368378.2015.1043556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
During the cell communication process, endogenous and exogenous signaling affect normal as well as pathological developmental conditions. Exogenous influences such as extra-low-frequency electromagnetic field (EMF) have been shown to effect pain and inflammation by modulating G-protein receptors, down-regulating cyclooxygenase-2 activity, and affecting the calcium/calmodulin/nitric oxide pathway. Investigators have reported changes in opioid receptors and second messengers, such as cyclic adenosine monophosphate (cAMP), in opiate tolerance and dependence by showing how repeated exposure to morphine decreases adenylate cyclase activity causing cAMP to return to control levels in the tolerant state, and increase above control levels during withdrawal. Resonance responses to biological systems using exogenous EMF signals suggest that frequency response characteristics of the target can determine the EMF biological response. In our past research we found significant down regulation of inflammatory markers tumor necrosis factor alpha (TNF-α) and nuclear factor kappa B (NFκB) using 5 Hz EMF frequency. In this study cAMP was stimulated in Chinese Hamster Ovary (CHO) cells transfected with human mu-opioid receptors, then exposed to 5 Hz EMF, and outcomes were compared with morphine treatment. Results showed a 23% greater inhibition of cAMP-treating cells with EMF than with morphine. In order to test our results for frequency specific effects, we ran identical experiments using 13 Hz EMF, which produced results similar to controls. This study suggests the use of EMF as a complementary or alternative treatment to morphine that could both reduce pain and enhance patient quality of life without the side-effects of opiates.
Collapse
Affiliation(s)
- Christina L Ross
- a Wake Forest Institute for Regenerative Medicine , Wake Forest Baptist Health , Winston-Salem , NC , USA.,b Wake Forest Center for Integrative Medicine , Wake Forest Baptist Health , Winston-Salem , NC , USA
| | - Thaleia Teli
- a Wake Forest Institute for Regenerative Medicine , Wake Forest Baptist Health , Winston-Salem , NC , USA
| | - Benjamin S Harrison
- a Wake Forest Institute for Regenerative Medicine , Wake Forest Baptist Health , Winston-Salem , NC , USA
| |
Collapse
|
26
|
Wie J, Kim BJ, Myeong J, Ha K, Jeong SJ, Yang D, Kim E, Jeon JH, So I. The Roles of Rasd1 small G proteins and leptin in the activation of TRPC4 transient receptor potential channels. Channels (Austin) 2015; 9:186-95. [PMID: 26083271 PMCID: PMC4594510 DOI: 10.1080/19336950.2015.1058454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
TRPC4 is important regulators of electrical excitability in gastrointestinal myocytes, pancreatic β-cells and neurons. Much is known regarding the assembly and function of these channels including TRPC1 as a homotetramer or a heteromultimer and the roles that their interacting proteins play in controlling these events. Further, they are one of the best-studied targets of G protein-coupled receptors and growth factors in general and Gαi/o and Gαq protein coupled receptor or epidermal growth factor and leptin in particular. However, our understanding of the roles of small G proteins and leptin on TRPC4 channels is still rudimentary. We discuss potential roles for Rasd1 small G protein and leptin in channel activation in addition to their known role in cellular signaling.
Collapse
Affiliation(s)
- Jinhong Wie
- a Department of Physiology ; Seoul National University College of Medicine ; Seoul , Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Myeong J, Kwak M, Jeon JP, Hong C, Jeon JH, So I. Close spatio-association of the transient receptor potential canonical 4 (TRPC4) channel with Gαi in TRPC4 activation process. Am J Physiol Cell Physiol 2015; 308:C879-89. [PMID: 25788576 DOI: 10.1152/ajpcell.00374.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/12/2015] [Indexed: 01/03/2023]
Abstract
TPRC channels are Ca(2+)-permeable, nonselective cation channels that are activated by a wide variety of stimuli, including G protein-coupled receptors (GPCRs). TRPC4 is commonly assumed to be activated by Gq/phospholipase C-coupled receptors. However, the other molecular mechanisms by which Gα proteins regulate TRPC4 remain unclear. Here, we found that Gαi2 regulates TRPC4 activation by direct binding. To investigate this mechanism, we used whole patch clamp and fluorescence resonance energy transfer (FRET). We tagged an isoform of mTRPC4 and G protein with CFP and YFP, respectively, and transiently transfected cells with the FRET pair. The FRET efficiency between TRPC4β-CFP and the constitutively active mutant form of Gαi2 was nearly 15% and was greater than that observed with wild-type Gαi2 (nearly 5%). Gβγ and the TRPC4 channel showed a FRET efficiency lower than 6%. In HEK293 cells transfected with the M2 muscarinic receptor, the application of carbachol increased the FRET efficiency between TRPC4β-CFP and Gαi2(WT)-YFP from 4.7 ± 0.4% (n = 7) to 12.6 ± 1.4% (n = 7). We also found that the TRPC4 channel directly interacts with Gαi2, but not with Gαq, when the channel is open. We analyzed the calcium levels in HEK293 cells expressing the channels and Gαi2 or Gαq using the calcium indicator YC6.1 (Yellow Cameleon 6.1). In response to the muscarinic agonist carbachol, M2-, Gαi2-, and TRPC4-expressing cells showed a prolonged Ca(2+) influx compared with cells expressing only M2. Together, these data suggest that Gαi2 activates the TRPC4 channel by direct binding, which then induces Ca(2+) entry.
Collapse
Affiliation(s)
- JongYun Myeong
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea; and
| | - Misun Kwak
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea; and
| | - Jae-Pyo Jeon
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Chansik Hong
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea; and
| | - Ju-Hong Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea; and
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea; and
| |
Collapse
|
28
|
Wie J, Kim J, Ha K, Zhang YH, Jeon JH, So I. Dexamethasone activates transient receptor potential canonical 4 (TRPC4) channels via Rasd1 small GTPase pathway. Pflugers Arch 2014; 467:2081-91. [PMID: 25502319 DOI: 10.1007/s00424-014-1666-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/24/2014] [Accepted: 12/01/2014] [Indexed: 12/15/2022]
Abstract
Canonical transient receptor potential 4 (TRPC4) channels are calcium-permeable, nonselective cation channels that are widely distributed in mammalian cells. It is generally speculated that TRPC4 channels are activated by Gq/11-PLC pathway or directly activated by Gi/o proteins. Although many mechanistic studies regarding TRPC4 have dealt with heterotrimeric G proteins, here, we first report the functional relationship between TRPC4 and small GTPase, Rasd1. Rasd1 selectively activated TRPC4 channels, and it was the only Ras protein among Ras protein family that can activate TRPC4 channels. For this to occur, it was found that certain population of functional Gαi1 and Gαi3 proteins are essential. Meanwhile, dexamethasone, a synthetic glucocorticoid and anti-inflammatory drug was known to increase messenger RNA (mRNA) level of Rasd1 in pancreatic β-cells. We have found that dexamethasone triggers TRPC4-like cationic current in INS-1 cells via increasing protein expression level of Rasd1. This relationship among dexamethasone, Rasd1, and TRPC4 could suggest a new therapeutic agent for hospitalized diabetes mellitus (DM) patients with prolonged dexamethasone prescription.
Collapse
Affiliation(s)
- Jinhong Wie
- Department of Physiology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea
| | - Jinsung Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea.,Catholic University of Korea, College of Medicine, Seoul, 137-701, Republic of Korea
| | - Kotdaji Ha
- Department of Physiology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea
| | - Yin Hua Zhang
- Department of Physiology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea.
| |
Collapse
|
29
|
Kang HJ, Menlove K, Ma J, Wilkins A, Lichtarge O, Wensel TG. Selectivity and evolutionary divergence of metabotropic glutamate receptors for endogenous ligands and G proteins coupled to phospholipase C or TRP channels. J Biol Chem 2014; 289:29961-74. [PMID: 25193666 DOI: 10.1074/jbc.m114.574483] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
To define the upstream and downstream signaling specificities of metabotropic glutamate receptors (mGluR), we have examined the ability of representative mGluR of group I, II, and III to be activated by endogenous amino acids and catalyze activation of G proteins coupled to phospholipase C (PLC), or activation of G(i/o) proteins coupled to the ion channel TRPC4β. Fluorescence-based assays have allowed us to observe interactions not previously reported or clearly identified. We have found that the specificity for endogenous amino acids is remarkably stringent. Even at millimolar levels, structurally similar compounds do not elicit significant activation. As reported previously, the clear exception is L-serine-O-phosphate (L-SOP), which strongly activates group III mGluR, especially mGluR4,-6,-8 but not group I or II mGluR. Whereas L-SOP cannot activate mGluR1 or mGluR2, it acts as a weak antagonist for mGluR1 and a potent antagonist for mGluR2, suggesting that co-recognition of L-glutamate and L-SOP arose early in evolution, and was followed later by divergence of group I and group II mGluR versus group III in l-SOP responses. mGluR7 has low affinity and efficacy for activation by both L-glutamate and L-SOP. Molecular docking studies suggested that residue 74 corresponding to lysine in mGluR4 and asparagine in mGluR7 might play a key role, and, indeed, mutagenesis experiments demonstrated that mutating this residue to lysine in mGluR7 enhances the potency of L-SOP. Experiments with pertussis toxin and dominant-negative Gα(i/o) proteins revealed that mGluR1 couples strongly to TRPC4β through Gα(i/o), in addition to coupling to PLC through Gα(q/11).
Collapse
Affiliation(s)
- Hye Jin Kang
- From the Graduate Program in Structural and Computational Biology and Molecular Biophysics
| | - Kit Menlove
- From the Graduate Program in Structural and Computational Biology and Molecular Biophysics
| | - Jianpeng Ma
- From the Graduate Program in Structural and Computational Biology and Molecular Biophysics, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, and Department of Bioengineering, Rice University, Houston, Texas 77005
| | - Angela Wilkins
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, and
| | - Olivier Lichtarge
- From the Graduate Program in Structural and Computational Biology and Molecular Biophysics, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, and
| | - Theodore G Wensel
- From the Graduate Program in Structural and Computational Biology and Molecular Biophysics, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, and
| |
Collapse
|
30
|
Sung HH, Choo SH, Ko M, Kang SJ, Chae MR, Kam SC, Han DH, So I, Lee SW. Increased expression of TRPC4 channels associated with erectile dysfunction in diabetes. Andrology 2014; 2:550-8. [PMID: 24782410 DOI: 10.1111/j.2047-2927.2014.00214.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/09/2014] [Accepted: 03/15/2014] [Indexed: 11/27/2022]
Abstract
In recent reports, an association between altered TRPC channel function and the development of various diabetic complications has drawn the attention of many investigators. The aim of this study was to investigate the expression of TRPC4 channels of corpus smooth muscle (CSM) cells in diabetes, and to evaluate the association between erectile dysfunction (ED) and altered TRPC4 channel function. The expression of TRPC4 in the penile tissue of human, normal and diabetic rat was investigated using RT-PCR, western blotting and immunohistochemistry (IHC). In vivo gene transfer of dominant negative (DN) TRPC4 into the CSM of rat was conducted. In vivo pelvic nerve stimulation was performed to measure erectile function. Expression of TRPC1, TRPC3, TRPC4 and TRPC6 in human and rat CSM tissues was confirmed by RT-PCR, western blot and IHC. In the diabetic rat, the expression levels of mRNA and protein of the TRPC4, and TRPC6 were significantly increased compared to control rats (p < 0.05). The change in TRPC4 expression in the diabetic rats was higher than those of the other TRPC subunits (p < 0.05). The IHC showed that only TRPC4 expression had a higher intensity in the diabetes compared to normal rats (p < 0.05). Gene transfection with TRPC4(DN) into the diabetic rats restored erectile function to levels similar to that of normal controls. Gene expression of TRPC4(DN) in CSM tissue was confirmed by RT-PCR 2 weeks after transfection. This study demonstrated that TRPC4 channel expression increased in the penile CSM cells of diabetic rats. The down-regulation of TRPC4 with DN form restored erectile function in the diabetic rats. The alteration of TRPC4 channel is one of pathophysiology of ED and could be a target for drug development for ED.
Collapse
Affiliation(s)
- H H Sung
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Nilius B, Flockerzi V. What do we really know and what do we need to know: some controversies, perspectives, and surprises. Handb Exp Pharmacol 2014; 223:1239-80. [PMID: 24961986 DOI: 10.1007/978-3-319-05161-1_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TRP channels comprise one of the most rapid growing research topics in ion channel research, in fields related to ion channels including channelopathies and translational medicine. We provide here a critical survey on our current knowledge of TRP channels and highlight some of the still open or controversial questions. This comprises questions related to evolution of TRP channels; biophysics, i.e., permeation; pore properties and gating; modulation; the still-elusive 3D structure; and channel subunits but also their role as general sensory channels and in human diseases. We will conclude that our knowledge on TRP channels is still at the very beginning of an exciting research journey.
Collapse
Affiliation(s)
- Bernd Nilius
- Department Cell Mol Medicine, Laboratory Ion Channel Research, KU Leuven, Campus Gasthuisberg, O&N 1, Herestraat 49-Bus 802, 3000, Leuven, Belgium,
| | | |
Collapse
|