1
|
Wyart C, Carbo-Tano M, Cantaut-Belarif Y, Orts-Del'Immagine A, Böhm UL. Cerebrospinal fluid-contacting neurons: multimodal cells with diverse roles in the CNS. Nat Rev Neurosci 2023; 24:540-556. [PMID: 37558908 DOI: 10.1038/s41583-023-00723-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 08/11/2023]
Abstract
The cerebrospinal fluid (CSF) is a complex solution that circulates around the CNS, and whose composition changes as a function of an animal's physiological state. Ciliated neurons that are bathed in the CSF - and thus referred to as CSF-contacting neurons (CSF-cNs) - are unusual polymodal interoceptive neurons. As chemoreceptors, CSF-cNs respond to variations in pH and osmolarity and to bacterial metabolites in the CSF. Their activation during infections of the CNS results in secretion of compounds to enhance host survival. As mechanosensory neurons, CSF-cNs operate together with an extracellular proteinaceous polymer known as the Reissner fibre to detect compression during spinal curvature. Once activated, CSF-cNs inhibit motor neurons, premotor excitatory neurons and command neurons to enhance movement speed and stabilize posture. At longer timescales, CSF-cNs instruct morphogenesis throughout life via the release of neuropeptides that act over long distances on skeletal muscle. Finally, recent evidence suggests that mouse CSF-cNs may act as neural stem cells in the spinal cord, inspiring new paths of investigation for repair after injury.
Collapse
Affiliation(s)
- Claire Wyart
- Institut du Cerveau (ICM), INSERM U1127, UMR CNRS 7225 Paris, Sorbonne Université, Paris, France.
| | - Martin Carbo-Tano
- Institut du Cerveau (ICM), INSERM U1127, UMR CNRS 7225 Paris, Sorbonne Université, Paris, France
| | - Yasmine Cantaut-Belarif
- Institut du Cerveau (ICM), INSERM U1127, UMR CNRS 7225 Paris, Sorbonne Université, Paris, France
| | | | - Urs L Böhm
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
2
|
Ng LCT, Harris BJ, Larmore M, Ta MC, Vien TN, Tokars VL, Yarov‐Yarovoy V, DeCaen PG. Energetic landscape of polycystin channel gating. EMBO Rep 2023; 24:e56783. [PMID: 37158562 PMCID: PMC10328073 DOI: 10.15252/embr.202356783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/06/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023] Open
Abstract
Members of the polycystin family (PKD2 and PKD2L1) of transient receptor potential (TRP) channels conduct Ca2+ and depolarizing monovalent cations. Variants in PKD2 cause autosomal dominant polycystic kidney disease (ADPKD) in humans, whereas loss of PKD2L1 expression causes seizure susceptibility in mice. Understanding structural and functional regulation of these channels will provide the basis for interpreting their molecular dysregulation in disease states. However, the complete structures of polycystins are unresolved, as are the conformational changes regulating their conductive states. To provide a holistic understanding of the polycystin gating cycle, we use computational prediction tools to model missing PKD2L1 structural motifs and evaluate more than 150 mutations in an unbiased mutagenic functional screen of the entire pore module. Our results provide an energetic landscape of the polycystin pore, which enumerates gating sensitive sites and interactions required for opening, inactivation, and subsequent desensitization. These findings identify the external pore helices and specific cross-domain interactions as critical structural regulators controlling the polycystin ion channel conductive and nonconductive states.
Collapse
Affiliation(s)
- Leo CT Ng
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Brandon J Harris
- Department of Physiology and Membrane BiologyUniversity of California, DavisDavisCAUSA
- Biophysics Graduate GroupUniversity of California, DavisDavisCAUSA
| | - Megan Larmore
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - My C Ta
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Thuy N Vien
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Valerie L Tokars
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Vladimir Yarov‐Yarovoy
- Department of Physiology and Membrane BiologyUniversity of California, DavisDavisCAUSA
- Department of Anesthesiology and Pain MedicineUniversity of California, DavisDavisCAUSA
| | - Paul G DeCaen
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| |
Collapse
|
3
|
Vien T, Ta M, Kimura L, Onay T, DeCaen P. Primary cilia TRP channel regulates hippocampal excitability. Proc Natl Acad Sci U S A 2023; 120:e2219686120. [PMID: 37216541 PMCID: PMC10235993 DOI: 10.1073/pnas.2219686120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Polycystins (PKD2, PKD2L1, and PKD2L2) are members of the transient receptor potential family, which form ciliary ion channels. Most notably, PKD2 dysregulation in the kidney nephron cilia is associated with polycystic kidney disease, but the function of PKD2L1 in neurons is undefined. In this report, we develop animal models to track the expression and subcellular localization of PKD2L1 in the brain. We discover that PKD2L1 localizes and functions as a Ca2+ channel in the primary cilia of hippocampal neurons that apically radiate from the soma. Loss of PKD2L1 expression ablates primary ciliary maturation and attenuates neuronal high-frequency excitability, which precipitates seizure susceptibility and autism spectrum disorder-like behavior in mice. The disproportionate impairment of interneuron excitability suggests that circuit disinhibition underlies the neurophenotypic features of these mice. Our results identify PKD2L1 channels as regulators of hippocampal excitability and the neuronal primary cilia as organelle mediators of brain electrical signaling.
Collapse
Affiliation(s)
- Thuy N. Vien
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - My C. Ta
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Louise F. Kimura
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Tuncer Onay
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL60911
| | - Paul G. DeCaen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| |
Collapse
|
4
|
Shimizu T, Fujii T, Hanita K, Shinozaki R, Takamura Y, Suzuki Y, Kageyama T, Kato M, Nishijo H, Tominaga M, Sakai H. Polycystic kidney disease 2-like 1 channel contributes to the bitter aftertaste perception of quinine. Sci Rep 2023; 13:4271. [PMID: 36922541 PMCID: PMC10017821 DOI: 10.1038/s41598-023-31322-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Bitterness is an important physiological function in the defense responses to avoid toxic foods. The taste receptor 2 family is well known to mediate bitter taste perception in Type II taste cells. Here, we report that the polycystic kidney disease 2-like 1 (PKD2L1) channel is a novel sensor for the bitter aftertaste in Type III taste cells. The PKD2L1 channel showed rebound activation after the washout of quinine, a bitter tastant, in electrophysiological whole-cell recordings of the PKD2L1-expressing HEK293T cells and Ca2+-imaging analysis of Type III taste cells isolated from wild-type PKD2L1 mice. In the short-term two-bottle preference and lick tests in vivo, the wild-type mice avoided normal water while the PKD2L1-knockout mice preferred normal water after they ingested the quinine-containing water. These results may explain the new mechanism of the quinine-triggered bitter aftertaste perception in Type III taste cells.
Collapse
Affiliation(s)
- Takahiro Shimizu
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| | - Takuto Fujii
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Keisuke Hanita
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Ryo Shinozaki
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Yusaku Takamura
- Department of System Emotional Science, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshiro Suzuki
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences (NIPS), Okazaki, 444-8787, Japan
| | - Teppei Kageyama
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Mizuki Kato
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Hisao Nishijo
- Department of System Emotional Science, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences (NIPS), Okazaki, 444-8787, Japan
| | - Hideki Sakai
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
5
|
Abstract
Polycystin subunits can form hetero- and homotetrameric ion channels in the membranes of various compartments of the cell. Homotetrameric polycystin channels are voltage- and calcium-modulated, whereas heterotetrameric versions are proposed to be ligand- or autoproteolytically regulated. Their importance is underscored by variants associated with autosomal dominant polycystic kidney disease and by vital roles in fertilization and embryonic development. The diversity in polycystin assembly and subcellular distribution allows for a multitude of sensory functions by this class of channels. In this review, we highlight their recent structural and functional characterization, which has provided a molecular blueprint to investigate the conformational changes required for channel opening in response to unique stimuli. We consider each polycystin channel type individually, discussing how they contribute to sensory cell biology, as well as their impact on the physiology of various tissues.
Collapse
Affiliation(s)
- Orhi Esarte Palomero
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA;
| | - Megan Larmore
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA;
| | - Paul G DeCaen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA;
| |
Collapse
|
6
|
Modulation of the long non-coding RNA Mir155hg by high, but not moderate, hydrostatic pressure in cartilage precursor cells. PLoS One 2022; 17:e0275682. [PMID: 36538560 PMCID: PMC9767356 DOI: 10.1371/journal.pone.0275682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 09/21/2022] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease in older adults and is characterized by a gradual degradation of articular cartilage due to decreased cartilage matrix gene expression and increased expression of genes involved in protein degradation, apoptosis and inflammation. Due to the high water content of cartilage, one of the main physical stimuli sensed by chondrocytes is hydrostatic pressure. We previously showed that high pressure above 20 MPa induced gene expression changes in chondrocyte precursor cells similar to what is observed in OA. Micro-RNAs are small non-coding RNAs essential to many physiological and pathological process including OA. As the micro-RNA miR-155 has been found increased in OA chondrocytes, we investigated the effects of high pressure on the expression of the miR-155 host gene Mir155hg. The chondrocyte progenitor cell line ATDC5 was pressurized under hydrostatic pressure up to 25 MPa and the expression of Mir155hg or the resulting micro-RNAs were measured; pharmacological inhibitors were used to identify the signaling pathways involved in the regulation of Mir155hg. We found that Mir155hg is strongly and rapidly up-regulated by high, but not moderate, pressure in chondrocyte progenitor cells. This up-regulation likely involves the membrane channel pannexin-1 and several intracellular signaling molecules including PKC and Src. MiR-155-5p and -3p were also up-regulated by pressure though somewhat later than Mir155hg, and a set of known miR-155-5p target genes, including Ikbke, Smarca4 and Ywhae, was affected by pressure, suggesting that Mir155hg may have important roles in cartilage physiology.
Collapse
|
7
|
TRPM7 is an essential regulator for volume-sensitive outwardly rectifying anion channel. Commun Biol 2021; 4:599. [PMID: 34017036 PMCID: PMC8137958 DOI: 10.1038/s42003-021-02127-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Animal cells can regulate their volume after swelling by the regulatory volume decrease (RVD) mechanism. In epithelial cells, RVD is attained through KCl release mediated via volume-sensitive outwardly rectifying Cl- channels (VSOR) and Ca2+-activated K+ channels. Swelling-induced activation of TRPM7 cation channels leads to Ca2+ influx, thereby stimulating the K+ channels. Here, we examined whether TRPM7 plays any role in VSOR activation. When TRPM7 was knocked down in human HeLa cells or knocked out in chicken DT40 cells, not only TRPM7 activity and RVD efficacy but also VSOR activity were suppressed. Heterologous expression of TRPM7 in TRPM7-deficient DT40 cells rescued both VSOR activity and RVD, accompanied by an increase in the expression of LRRC8A, a core molecule of VSOR. TRPM7 exerts the facilitating action on VSOR activity first by enhancing molecular expression of LRRC8A mRNA through the mediation of steady-state Ca2+ influx and second by stabilizing the plasmalemmal expression of LRRC8A protein through the interaction between LRRC8A and the C-terminal domain of TRPM7. Therefore, TRPM7 functions as an essential regulator of VSOR activity and LRRC8A expression.
Collapse
|
8
|
Involvement of pore helix in voltage-dependent inactivation of TRPM5 channel. Heliyon 2021; 7:e06102. [PMID: 33553759 PMCID: PMC7848652 DOI: 10.1016/j.heliyon.2021.e06102] [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: 02/08/2020] [Revised: 11/21/2020] [Accepted: 01/22/2021] [Indexed: 12/05/2022] Open
Abstract
The transient receptor potential melastatin 5 (TRPM5) channel is a monovalent-permeable cation channel that is activated by intracellular Ca2+. Expression of TRPM5 has been shown in taste cells, pancreas, brainstem and olfactory epithelium, and this channel is thought to be involved in controlling membrane potentials. In whole-cell patch-clamp recordings, TRPM5 exhibited voltage-dependent inactivation at negative membrane potentials and time constant of voltage-dependent inactivation of TRPM5 did not depend on the intracellular Ca2+ concentrations between 100 and 500 nM. Alanine substitution at Y913 and I916 in the pore helix of TRPM5 increased time constant of voltage-dependent inactivation. Meanwhile, voltage-dependent inactivation was reduced in TRPM5 mutants having glycine substitution at L901, Y913, Q915 and I916 in the pore helix. From these results, we conclude that the pore helix in the outer pore loop might play a role in voltage-dependent inactivation of TRPM5.
Collapse
|
9
|
Walsh S, Izquierdo-Serra M, Acosta S, Edo A, Lloret M, Moret R, Bosch E, Oliva B, Bertranpetit J, Fernández-Fernández JM. Adaptive selection drives TRPP3 loss-of-function in an Ethiopian population. Sci Rep 2020; 10:20999. [PMID: 33268808 PMCID: PMC7710729 DOI: 10.1038/s41598-020-78081-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 11/20/2020] [Indexed: 11/15/2022] Open
Abstract
TRPP3 (also called PKD2L1) is a nonselective, cation-permeable channel activated by multiple stimuli, including extracellular pH changes. TRPP3 had been considered a candidate for sour sensor in humans, due to its high expression in a subset of tongue receptor cells detecting sour, along with its membership to the TRP channel family known to function as sensory receptors. Here, we describe the functional consequences of two non-synonymous genetic variants (R278Q and R378W) found to be under strong positive selection in an Ethiopian population, the Gumuz. Electrophysiological studies and 3D modelling reveal TRPP3 loss-of-functions produced by both substitutions. R278Q impairs TRPP3 activation after alkalinisation by mislocation of H+ binding residues at the extracellular polycystin mucolipin domain. R378W dramatically reduces channel activity by altering conformation of the voltage sensor domain and hampering channel transition from closed to open state. Sour sensitivity tests in R278Q/R378W carriers argue against both any involvement of TRPP3 in sour detection and the role of such physiological process in the reported evolutionary positive selection past event.
Collapse
Affiliation(s)
- Sandra Walsh
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003, Barcelona, Catalonia, Spain
| | - Mercè Izquierdo-Serra
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Sandra Acosta
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003, Barcelona, Catalonia, Spain
| | - Albert Edo
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - María Lloret
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Roser Moret
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003, Barcelona, Catalonia, Spain
| | - Elena Bosch
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 43206, Reus, Spain
| | - Baldo Oliva
- Structural Bioinformatics Lab, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Jaume Bertranpetit
- Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003, Barcelona, Catalonia, Spain.
| | - José Manuel Fernández-Fernández
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain.
| |
Collapse
|
10
|
Di Bella DJ, Carcagno AL, Bartolomeu ML, Pardi MB, Löhr H, Siegel N, Hammerschmidt M, Marín-Burgin A, Lanuza GM. Ascl1 Balances Neuronal versus Ependymal Fate in the Spinal Cord Central Canal. Cell Rep 2019; 28:2264-2274.e3. [DOI: 10.1016/j.celrep.2019.07.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/07/2019] [Accepted: 07/23/2019] [Indexed: 01/04/2023] Open
|
11
|
Abstract
The opening of voltage-gated ion channels is initiated by transfer of gating charges that sense the electric field across the membrane. Although transient receptor potential ion channels (TRP) are members of this family, their opening is not intrinsically linked to membrane potential, and they are generally not considered voltage gated. Here we demonstrate that TRPP2, a member of the polycystin subfamily of TRP channels encoded by the PKD2L1 gene, is an exception to this rule. TRPP2 borrows a biophysical riff from canonical voltage-gated ion channels, using 2 gating charges found in its fourth transmembrane segment (S4) to control its conductive state. Rosetta structural prediction demonstrates that the S4 undergoes ∼3- to 5-Å transitional and lateral movements during depolarization, which are coupled to opening of the channel pore. Here both gating charges form state-dependent cation-π interactions within the voltage sensor domain (VSD) during membrane depolarization. Our data demonstrate that the transfer of a single gating charge per channel subunit is requisite for voltage, temperature, and osmotic swell polymodal gating of TRPP2. Taken together, we find that irrespective of stimuli, TRPP2 channel opening is dependent on activation of its VSDs.
Collapse
|
12
|
Gao P, Yan Z, Zhu Z. The role of adipose TRP channels in the pathogenesis of obesity. J Cell Physiol 2019; 234:12483-12497. [PMID: 30618095 DOI: 10.1002/jcp.28106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Peng Gao
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| |
Collapse
|
13
|
Lu Z, Cui Y, Wei X, Gao P, Zhang H, Wei X, Li Q, Sun F, Yan Z, Zheng H, Yang G, Liu D, Zhu Z. Deficiency of PKD2L1 (TRPP3) Exacerbates Pathological Cardiac Hypertrophy by Augmenting NCX1-Mediated Mitochondrial Calcium Overload. Cell Rep 2018; 24:1639-1652. [DOI: 10.1016/j.celrep.2018.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/19/2018] [Accepted: 07/06/2018] [Indexed: 10/28/2022] Open
|
14
|
Zheng W, Yang X, Hu R, Cai R, Hofmann L, Wang Z, Hu Q, Liu X, Bulkley D, Yu Y, Tang J, Flockerzi V, Cao Y, Cao E, Chen XZ. Hydrophobic pore gates regulate ion permeation in polycystic kidney disease 2 and 2L1 channels. Nat Commun 2018; 9:2302. [PMID: 29899465 PMCID: PMC5998024 DOI: 10.1038/s41467-018-04586-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 05/01/2018] [Indexed: 01/20/2023] Open
Abstract
PKD2 and PKD1 genes are mutated in human autosomal dominant polycystic kidney disease. PKD2 can form either a homomeric cation channel or a heteromeric complex with the PKD1 receptor, presumed to respond to ligand(s) and/or mechanical stimuli. Here, we identify a two-residue hydrophobic gate in PKD2L1, and a single-residue hydrophobic gate in PKD2. We find that a PKD2 gain-of-function gate mutant effectively rescues PKD2 knockdown-induced phenotypes in embryonic zebrafish. The structure of a PKD2 activating mutant F604P by cryo-electron microscopy reveals a π- to α-helix transition within the pore-lining helix S6 that leads to repositioning of the gate residue and channel activation. Overall the results identify hydrophobic gates and a gating mechanism of PKD2 and PKD2L1. Mutations in the cation channel PKD2 cause human autosomal dominant polycystic kidney disease but its channel function and gating mechanism are poorly understood. Here authors study PKD2 using electrophysiology and cryo-EM, which identifies hydrophobic gates and proposes a gating mechanism for PKD2.
Collapse
Affiliation(s)
- Wang Zheng
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.,Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Xiaoyong Yang
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Ruikun Hu
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Ruiqi Cai
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Laura Hofmann
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, 66421, Germany
| | - Zhifei Wang
- Department of Biological Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qiaolin Hu
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Xiong Liu
- Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - David Bulkley
- Keck Advanced Microscopy Laboratory and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Yong Yu
- Department of Biological Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Veit Flockerzi
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, Homburg, 66421, Germany
| | - Ying Cao
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA.
| | - Xing-Zhen Chen
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China. .,Department of Physiology, Membrane Protein Disease Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
15
|
Su Q, Hu F, Liu Y, Ge X, Mei C, Yu S, Shen A, Zhou Q, Yan C, Lei J, Zhang Y, Liu X, Wang T. Cryo-EM structure of the polycystic kidney disease-like channel PKD2L1. Nat Commun 2018; 9:1192. [PMID: 29567962 PMCID: PMC5864754 DOI: 10.1038/s41467-018-03606-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 02/28/2018] [Indexed: 02/07/2023] Open
Abstract
PKD2L1, also termed TRPP3 from the TRPP subfamily (polycystic TRP channels), is involved in the sour sensation and other pH-dependent processes. PKD2L1 is believed to be a nonselective cation channel that can be regulated by voltage, protons, and calcium. Despite its considerable importance, the molecular mechanisms underlying PKD2L1 regulations are largely unknown. Here, we determine the PKD2L1 atomic structure at 3.38 Å resolution by cryo-electron microscopy, whereby side chains of nearly all residues are assigned. Unlike its ortholog PKD2, the pore helix (PH) and transmembrane segment 6 (S6) of PKD2L1, which are involved in upper and lower-gate opening, adopt an open conformation. Structural comparisons of PKD2L1 with a PKD2-based homologous model indicate that the pore domain dilation is coupled to conformational changes of voltage-sensing domains (VSDs) via a series of π-π interactions, suggesting a potential PKD2L1 gating mechanism.
Collapse
Affiliation(s)
- Qiang Su
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China
| | - Feizhuo Hu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China.,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China.,School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yuxia Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China.,X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering and McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China.,School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China, 102402
| | - Xiaofei Ge
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Changlin Mei
- Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Shengqiang Yu
- Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Aiwen Shen
- Department of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Qiang Zhou
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China.,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China.,School of Medicine, Tsinghua University, Beijing, 100084, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Chuangye Yan
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jianlin Lei
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China
| | - Yanqing Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China. .,School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China. .,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Xiaodong Liu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,School of Medicine, Tsinghua University, Beijing, 100084, China. .,X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering and McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China. .,School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China. .,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China, 102402.
| | - Tingliang Wang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua University, Beijing, 100084, China. .,Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, 100084, China. .,School of Medicine, Tsinghua University, Beijing, 100084, China. .,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
16
|
Park EYJ, Kwak M, Ha K, So I. Identification of clustered phosphorylation sites in PKD2L1: how PKD2L1 channel activation is regulated by cyclic adenosine monophosphate signaling pathway. Pflugers Arch 2017; 470:505-516. [PMID: 29230552 DOI: 10.1007/s00424-017-2095-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/21/2017] [Accepted: 12/01/2017] [Indexed: 01/01/2023]
Abstract
Polycystic kidney disease 2-like-1 (PKD2L1), or polycystin-L or TRPP2, formerly TRPP3, is a transient receptor potential (TRP) superfamily member. It is a calcium-permeable non-selective cation channel that regulates intracellular calcium concentration and thereby calcium signaling. PKD2L1 has been reported to take part in hedgehog signaling in renal primary cilia and sour tasting coupling with PKD1L3. In addition to the previous reports, PKD2L1 is recently found to play a crucial role in localization with β2-adrenergic receptor (β2AR) on the neuronal primary cilia. The disruption of PKD2L1 leads to the loss of β2AR on the primary cilia and reduction in intracellular concentration of cyclic adenosine monophosphate (cAMP). Since the role of cAMP and PKA is frequently mentioned in the studies of PKD diseases, we investigated on the mechanism of cAMP regulation in relation to the function of PKD2L1 channel. In this study, we observed the activity of PKD2L1 channel increased by the downstream cascades of β2AR and found the clustered phosphorylation sites, Ser-682, Ser-685, and Ser-686 that are significant in the channel regulation by phosphorylation.
Collapse
Affiliation(s)
- Eunice Yon June Park
- Department of Physiology, Seoul National University, College of Medicine, Biomedical Science Building 117, 103 Daehakro, Jongro-gu, Seoul, 110-799, South Korea
| | - Misun Kwak
- Department of Physiology, Seoul National University, College of Medicine, Biomedical Science Building 117, 103 Daehakro, Jongro-gu, Seoul, 110-799, South Korea
| | - Kotdaji Ha
- Department of Physiology, University of California, San Francisco, CA, 94158-2517, USA
| | - Insuk So
- Department of Physiology, Seoul National University, College of Medicine, Biomedical Science Building 117, 103 Daehakro, Jongro-gu, Seoul, 110-799, South Korea.
| |
Collapse
|
17
|
Integrative Approach with Electrophysiological and Theoretical Methods Reveals a New Role of S4 Positively Charged Residues in PKD2L1 Channel Voltage-Sensing. Sci Rep 2017; 7:9760. [PMID: 28852171 PMCID: PMC5575081 DOI: 10.1038/s41598-017-10357-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/02/2017] [Indexed: 01/15/2023] Open
Abstract
Numerical model-based simulations provide important insights into ion channel gating when experimental limitations exist. Here, a novel strategy combining numerical simulations with patch clamp experiments was used to investigate the net positive charges in the putative transmembrane segment 4 (S4) of the atypical, positively-shifted voltage-dependence of polycystic kidney disease 2-like 1 (PKD2L1) channel. Charge-neutralising mutations (K452Q, K455Q and K461Q) in S4 reduced gating charges, positively shifted the Boltzmann-type activation curve [i.e., open probability (Popen)-V curve] and altered the time-courses of activation/deactivation of PKD2L1, indicating that this region constitutes part of a voltage sensor. Numerical reconstruction of wild-type (WT) and mutant PKD2L1-mediated currents necessitated, besides their voltage-dependent gating parameters, a scaling factor that describes the voltage-dependence of maximal conductance, Gmax. Subsequent single-channel conductance (γ) measurements revealed that voltage-dependence of Gmax in WT can be explained by the inward-rectifying property of γ, which is greatly changed in PKD2L1 mutants. Homology modelling based on PKD2 and NaVAb structures suggest that such voltage dependence of Popen and γ in PKD2L1 could both reflect the charged state of the S4 domain. The present conjunctive experimental and theoretical approaches provide a framework to explore the undetermined mechanism(s) regulating TRP channels that possess non-classical voltage-dependent properties.
Collapse
|
18
|
Shimizu T, Higuchi T, Toba T, Ohno C, Fujii T, Nilius B, Sakai H. The asparagine 533 residue in the outer pore loop region of the mouse PKD2L1 channel is essential for its voltage-dependent inactivation. FEBS Open Bio 2017; 7:1392-1401. [PMID: 28904867 PMCID: PMC5586397 DOI: 10.1002/2211-5463.12273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 11/17/2022] Open
Abstract
Voltage‐dependent inactivation of ion channels contributes to the regulation of the membrane potential of excitable cells. Mouse polycystic kidney disease 2‐like 1 (PKD2L1) forms voltage‐dependent nonselective cation channels, which are activated but subsequently inactivated in response to membrane depolarization. Here, we found that the mutation of an asparagine 533 residue (N533Q) in the outer pore loop region of PKD2L1 caused a marked increase in outward currents induced by depolarization. In addition, the tail current analysis demonstrated that the N533Q mutants are activated during depolarization but the subsequent inactivation does not occur. Interestingly, the N533Q mutants lacked the channel activation triggered by the removal of stimuli such as extracellular alkalization and heating. Our findings suggest that the N533 residue in the outer pore loop region of PKD2L1 has a key role in the voltage‐dependent channel inactivation.
Collapse
Affiliation(s)
- Takahiro Shimizu
- Department of Pharmaceutical Physiology Graduate School of Medicine and Pharmaceutical Sciences University of Toyama Japan
| | - Taiga Higuchi
- Department of Pharmaceutical Physiology Graduate School of Medicine and Pharmaceutical Sciences University of Toyama Japan
| | - Toshihiro Toba
- Department of Pharmaceutical Physiology Graduate School of Medicine and Pharmaceutical Sciences University of Toyama Japan
| | - Chie Ohno
- Department of Pharmaceutical Physiology Graduate School of Medicine and Pharmaceutical Sciences University of Toyama Japan
| | - Takuto Fujii
- Department of Pharmaceutical Physiology Graduate School of Medicine and Pharmaceutical Sciences University of Toyama Japan
| | - Bernd Nilius
- Laboratory of Ion Channel Research Department of Cellular and Molecular Medicine KU Leuven Belgium
| | - Hideki Sakai
- Department of Pharmaceutical Physiology Graduate School of Medicine and Pharmaceutical Sciences University of Toyama Japan
| |
Collapse
|
19
|
Djenoune L, Wyart C. Light on a sensory interface linking the cerebrospinal fluid to motor circuits in vertebrates. J Neurogenet 2017; 31:113-127. [PMID: 28789587 DOI: 10.1080/01677063.2017.1359833] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cerebrospinal fluid (CSF) is circulating around the entire central nervous system (CNS). The main function of the CSF has been thought to insure the global homeostasis of the CNS. Recent evidence indicates that the CSF also dynamically conveys signals modulating the development and the activity of the nervous system. The later observation implies that cues from the CSF could act on neurons in the brain and the spinal cord via bordering receptor cells. Candidate neurons to enable such modulation are the cerebrospinal fluid-contacting neurons (CSF-cNs) that are located precisely at the interface between the CSF and neuronal circuits. The atypical apical extension of CSF-cNs bears a cluster of microvilli bathing in the CSF indicating putative sensory or secretory roles in relation with the CSF. In the brainstem and spinal cord, CSF-cNs have been described in over two hundred species by Kolmer and Agduhr, suggesting an important function within the spinal cord. However, the lack of specific markers and the difficulty to access CSF-cNs hampered their physiological investigation. The transient receptor potential channel PKD2L1 is a specific marker of spinal CSF-cNs in vertebrate species. The transparency of zebrafish at early stages eases the functional characterization of pkd2l1+ CSF-cNs. Recent studies demonstrate that spinal CSF-cNs detect spinal curvature via the channel PKD2L1 and modulate locomotion and posture by projecting onto spinal interneurons and motor neurons in vivo. In vitro recordings demonstrated that spinal CSF-cNs are sensing pH variations mainly through ASIC channels, in combination with PKD2L1. Altogether, neurons contacting the CSF appear as a novel sensory modality enabling the detection of mechanical and chemical stimuli from the CSF and modulating the excitability of spinal circuits underlying locomotion and posture.
Collapse
Affiliation(s)
- Lydia Djenoune
- a Institut du Cerveau et de la Moelle épinière (ICM) , Paris , France
| | - Claire Wyart
- a Institut du Cerveau et de la Moelle épinière (ICM) , Paris , France
| |
Collapse
|
20
|
The dual developmental origin of spinal cerebrospinal fluid-contacting neurons gives rise to distinct functional subtypes. Sci Rep 2017; 7:719. [PMID: 28389647 PMCID: PMC5428266 DOI: 10.1038/s41598-017-00350-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/30/2017] [Indexed: 11/30/2022] Open
Abstract
Chemical and mechanical cues from the cerebrospinal fluid (CSF) can affect the development and function of the central nervous system (CNS). How such cues are detected and relayed to the CNS remains elusive. Cerebrospinal fluid-contacting neurons (CSF-cNs) situated at the interface between the CSF and the CNS are ideally located to convey such information to local networks. In the spinal cord, these GABAergic neurons expressing the PKD2L1 channel extend an apical extension into the CSF and an ascending axon in the spinal cord. In zebrafish and mouse spinal CSF-cNs originate from two distinct progenitor domains characterized by distinct cascades of transcription factors. Here we ask whether these neurons with different developmental origins differentiate into cells types with different functional properties. We show in zebrafish larva that the expression of specific markers, the morphology of the apical extension and axonal projections, as well as the neuronal targets contacted by CSF-cN axons, distinguish the two CSF-cN subtypes. Altogether our study demonstrates that the developmental origins of spinal CSF-cNs give rise to two distinct functional populations of sensory neurons. This work opens novel avenues to understand how these subtypes may carry distinct functions related to development of the spinal cord, locomotion and posture.
Collapse
|
21
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
22
|
Elinder F, Liin SI. Actions and Mechanisms of Polyunsaturated Fatty Acids on Voltage-Gated Ion Channels. Front Physiol 2017; 8:43. [PMID: 28220076 PMCID: PMC5292575 DOI: 10.3389/fphys.2017.00043] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/16/2017] [Indexed: 01/29/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) act on most ion channels, thereby having significant physiological and pharmacological effects. In this review we summarize data from numerous PUFAs on voltage-gated ion channels containing one or several voltage-sensor domains, such as voltage-gated sodium (NaV), potassium (KV), calcium (CaV), and proton (HV) channels, as well as calcium-activated potassium (KCa), and transient receptor potential (TRP) channels. Some effects of fatty acids appear to be channel specific, whereas others seem to be more general. Common features for the fatty acids to act on the ion channels are at least two double bonds in cis geometry and a charged carboxyl group. In total we identify and label five different sites for the PUFAs. PUFA site 1: The intracellular cavity. Binding of PUFA reduces the current, sometimes as a time-dependent block, inducing an apparent inactivation. PUFA site 2: The extracellular entrance to the pore. Binding leads to a block of the channel. PUFA site 3: The intracellular gate. Binding to this site can bend the gate open and increase the current. PUFA site 4: The interface between the extracellular leaflet of the lipid bilayer and the voltage-sensor domain. Binding to this site leads to an opening of the channel via an electrostatic attraction between the negatively charged PUFA and the positively charged voltage sensor. PUFA site 5: The interface between the extracellular leaflet of the lipid bilayer and the pore domain. Binding to this site affects slow inactivation. This mapping of functional PUFA sites can form the basis for physiological and pharmacological modifications of voltage-gated ion channels.
Collapse
Affiliation(s)
- Fredrik Elinder
- Department of Clinical and Experimental Medicine, Linköping University Linköping, Sweden
| | - Sara I Liin
- Department of Clinical and Experimental Medicine, Linköping University Linköping, Sweden
| |
Collapse
|
23
|
Taruno A, Marunaka Y. Hypotonicity activates a voltage-dependent membrane conductance in N2a neuroblastoma cells. Biochem Biophys Res Commun 2017; 484:331-335. [PMID: 28130109 DOI: 10.1016/j.bbrc.2017.01.118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/22/2017] [Indexed: 01/26/2023]
Abstract
To maintain cellular and bodily homeostasis, cells respond to extracellular stimuli including osmotic stress by activating various ion channels, which have been implicated in many physiological and pathophysiological conditions. However, cellular osmosensory mechanisms remain elusive. Here, we report a novel voltage-dependent current in N2a cells activated by exposure to hypotonic stress. After a hypotonic challenge, N2a cells sequentially develop two distinct currents. The volume-regulated anion channel (VRAC) current emerges first and, after a delay, activation of a previously uncharacterized strongly outwardly rectifying current follows. The latter, delayed current (Id) is insensitive to NPPB, a nonspecific blocker of Cl- channels, and intracellular Mg2+, which inhibits VRAC and swelling-activated TRPM3 and TRPM7 channels. Replacement of extracellular Na+ with NMDG+ reduces inward tail currents, suggesting that Id is mediated by cations. Finally, Id shows voltage-dependent activation with slow activation kinetics and half-maximal activation at +76 mV. These pharmacological and biophysical characteristics of Id are distinct from those of known osmotic cell swelling-activated ion channels. In conclusion, our data identify and characterize a novel osmotically-activated, voltage-dependent ion channel in N2a cells.
Collapse
Affiliation(s)
- Akiyuki Taruno
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, 465 Kajiicho Kamigyo-ward, Kyoto 602-8566, Japan.
| | - Yoshinori Marunaka
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, 465 Kajiicho Kamigyo-ward, Kyoto 602-8566, Japan; Department of Bio-Ionomics, Kyoto Prefectural University of Medicine, 465 Kajiicho Kamigyo-ward, Kyoto 602-8566, Japan.
| |
Collapse
|
24
|
DeCaen PG, Liu X, Abiria S, Clapham DE. Atypical calcium regulation of the PKD2-L1 polycystin ion channel. eLife 2016; 5. [PMID: 27348301 PMCID: PMC4922860 DOI: 10.7554/elife.13413] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 05/12/2016] [Indexed: 11/13/2022] Open
Abstract
Native PKD2-L1 channel subunits are present in primary cilia and other restricted cellular spaces. Here we investigate the mechanism for the channel's unusual regulation by external calcium, and rationalize this behavior to its specialized function. We report that the human PKD2-L1 selectivity filter is partially selective to calcium ions (Ca(2+)) moving into the cell, but blocked by high internal Ca(2+)concentrations, a unique feature of this transient receptor potential (TRP) channel family member. Surprisingly, we find that the C-terminal EF-hands and coiled-coil domains do not contribute to PKD2-L1 Ca(2+)-induced potentiation and inactivation. We propose a model in which prolonged channel activity results in calcium accumulation, triggering outward-moving Ca(2+) ions to block PKD2-L1 in a high-affinity interaction with the innermost acidic residue (D523) of the selectivity filter and subsequent long-term channel inactivation. This response rectifies Ca(2+) flow, enabling Ca(2+) to enter but not leave small compartments such as the cilium.
Collapse
Affiliation(s)
- Paul G DeCaen
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Xiaowen Liu
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Sunday Abiria
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - David E Clapham
- Department of Cardiology, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
25
|
Kimura M, Sase T, Higashikawa A, Sato M, Sato T, Tazaki M, Shibukawa Y. High pH-Sensitive TRPA1 Activation in Odontoblasts Regulates Mineralization. J Dent Res 2016; 95:1057-64. [PMID: 27084672 DOI: 10.1177/0022034516644702] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Calcium hydroxide and mineral trioxide aggregate are widely used for indirect and direct pulp capping and root canal filling. Their dissociation into Ca(2+) and OH(-) in dental pulp creates an alkaline environment, which activates reparative/reactionary dentinogenesis. However, the mechanisms by which odontoblasts detect the pH of the extracellular environment remain unclear. We examined the alkali-sensitive intracellular Ca(2+) signaling pathway in rat odontoblasts. In the presence or absence of extracellular Ca(2+), application of alkaline solution increased intracellular Ca(2+) concentration, or [Ca(2+)]i Alkaline solution-induced [Ca(2+)]i increases depended on extracellular pH (8.5 to 10.5) in both the absence and the presence of extracellular Ca(2+) The amplitude was smaller in the absence than in the presence of extracellular Ca(2+) Each increase in [Ca(2+)]i, activated by pH 7.5, 8.5, or 9.5, depended on extracellular Ca(2+) concentration; the equilibrium binding constant for extracellular Ca(2+) concentration decreased as extracellular pH increased (1.04 mM at pH 7.5 to 0.11 mM at pH 9.5). Repeated applications of alkaline solution did not have a desensitizing effect on alkali-induced [Ca(2+)]i increases and inward currents. In the presence of extracellular Ca(2+), alkaline solution-induced [Ca(2+)]i increases were suppressed by application of an antagonist of transient receptor potential ankyrin subfamily member 1 (TRPA1) channels. Ca(2+) exclusion efficiency during alkaline solution-induced [Ca(2+)]i increases was reduced by a Na(+)-Ca(2+) exchanger antagonist. Alizarin red and von Kossa staining revealed increased mineralization levels under repeated high pH stimulation, whereas the TRPA1 antagonist strongly reduced this effect. These findings indicate that alkaline stimuli-such as the alkaline environment inside dental pulp treated with calcium hydroxide or mineral trioxide aggregate-activate Ca(2+) mobilization via Ca(2+) influx mediated by TRPA1 channels and intracellular Ca(2+) release in odontoblasts. High pH-sensing mechanisms in odontoblasts are important for activating dentinogenesis induced by an alkaline environment.
Collapse
Affiliation(s)
- M Kimura
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - T Sase
- Department of Crown and Bridge Prosthodontics, Tokyo Dental College, Tokyo, Japan
| | - A Higashikawa
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - M Sato
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - T Sato
- Department of Crown and Bridge Prosthodontics, Tokyo Dental College, Tokyo, Japan
| | - M Tazaki
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Y Shibukawa
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| |
Collapse
|
26
|
Function and regulation of TRPP2 ion channel revealed by a gain-of-function mutant. Proc Natl Acad Sci U S A 2016; 113:E2363-72. [PMID: 27071085 DOI: 10.1073/pnas.1517066113] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mutations in polycystin-1 and transient receptor potential polycystin 2 (TRPP2) account for almost all clinically identified cases of autosomal dominant polycystic kidney disease (ADPKD), one of the most common human genetic diseases. TRPP2 functions as a cation channel in its homomeric complex and in the TRPP2/polycystin-1 receptor/ion channel complex. The activation mechanism of TRPP2 is unknown, which significantly limits the study of its function and regulation. Here, we generated a constitutively active gain-of-function (GOF) mutant of TRPP2 by applying a mutagenesis scan on the S4-S5 linker and the S5 transmembrane domain, and studied functional properties of the GOF TRPP2 channel. We found that extracellular divalent ions, including Ca(2+), inhibit the permeation of monovalent ions by directly blocking the TRPP2 channel pore. We also found that D643, a negatively charged amino acid in the pore, is crucial for channel permeability. By introducing single-point ADPKD pathogenic mutations into the GOF TRPP2, we showed that different mutations could have completely different effects on channel activity. The in vivo function of the GOF TRPP2 was investigated in zebrafish embryos. The results indicate that, compared with wild type (WT), GOF TRPP2 more efficiently rescued morphological abnormalities, including curly tail and cyst formation in the pronephric kidney, caused by down-regulation of endogenous TRPP2 expression. Thus, we established a GOF TRPP2 channel that can serve as a powerful tool for studying the function and regulation of TRPP2. The GOF channel may also have potential application for developing new therapeutic strategies for ADPKD.
Collapse
|
27
|
Fernandes ES, Cerqueira ARA, Soares AG, Costa SKP. Capsaicin and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 929:91-125. [PMID: 27771922 DOI: 10.1007/978-3-319-41342-6_5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A significant number of experimental and clinical studies published in peer-reviewed journals have demonstrated promising pharmacological properties of capsaicin in relieving signs and symptoms of non-communicable diseases (chronic diseases). This chapter provides an overview made from basic and clinical research studies of the potential therapeutic effects of capsaicin, loaded in different application forms, such as solution and cream, on chronic diseases (e.g. arthritis, chronic pain, functional gastrointestinal disorders and cancer). In addition to the anti-inflammatory and analgesic properties of capsaicin largely recognized via, mainly, interaction with the TRPV1, the effects of capsaicin on different cell signalling pathways will be further discussed here. The analgesic, anti-inflammatory or apoptotic effects of capsaicin show promising results in arthritis, neuropathic pain, gastrointestinal disorders or cancer, since evidence demonstrates that the oral or local application of capsaicin reduce inflammation and pain in rheumatoid arthritis, promotes gastric protection against ulcer and induces apoptosis of the tumour cells. Sadly, these results have been paralleled by conflicting studies, which indicate that high concentrations of capsaicin are likely to evoke deleterious effects, thus suggesting that capsaicin activates different pathways at different concentrations in both human and rodent tissues. Thus, to establish effective capsaicin doses for chronic conditions, which can be benefited from capsaicin therapeutic effects, is a real challenge that must be pursued.
Collapse
Affiliation(s)
- E S Fernandes
- Programa de Pós-Graduação, Universidade Ceuma, São Luís-MA, Brazil.,Vascular Biology Section, Cardiovascular Division, King's College London, London, UK
| | - A R A Cerqueira
- Department of Pharmacology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), Av. Prof. Lineu Prestes, 1524 - Room 326, Butantan, São Paulo, 05508-900, Sao Paulo, Brazil
| | - A G Soares
- Department of Pharmacology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), Av. Prof. Lineu Prestes, 1524 - Room 326, Butantan, São Paulo, 05508-900, Sao Paulo, Brazil
| | - Soraia K P Costa
- Department of Pharmacology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), Av. Prof. Lineu Prestes, 1524 - Room 326, Butantan, São Paulo, 05508-900, Sao Paulo, Brazil.
| |
Collapse
|
28
|
Hussein S, Zheng W, Dyte C, Wang Q, Yang J, Zhang F, Tang J, Cao Y, Chen XZ. Acid-induced off-response of PKD2L1 channel in Xenopus oocytes and its regulation by Ca(2.). Sci Rep 2015; 5:15752. [PMID: 26502994 PMCID: PMC4621500 DOI: 10.1038/srep15752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 09/14/2015] [Indexed: 01/18/2023] Open
Abstract
Polycystic kidney disease (PKD) protein 2 Like 1 (PKD2L1), also called transient receptor potential polycystin-3 (TRPP3), regulates Ca(2+)-dependent hedgehog signalling in primary cilia, intestinal development and sour tasting but with an unclear mechanism. PKD2L1 is a Ca(2+)-permeable cation channel that is activated by extracellular Ca(2+) (on-response) in Xenopus oocytes. PKD2L1 co-expressed with PKD protein 1 Like 3 (PKD1L3) exhibits extracellular acid-induced activation (off-response, i.e., activation following acid removal) but whether PKD1L3 participates in acid sensing remains unclear. Here we used the two-microelectrode voltage-clamp, site directed mutagenesis, Western blotting, reverse transcriptase-polymerase chain reaction (RT-PCR) and immunofluorescence, and showed that PKD2L1 expressed in oocytes exhibits sustained off-response currents in the absence of PKD1L3. PKD1L3 co-expression augmented the PKD2L1 plasma membrane localization but did not alter the observed properties of the off-response. PKD2L1 off-response was inhibited by an increase in intracellular Ca(2+). We also identified two intra-membrane residues aspartic acid 349 (D349) and glutamic acid 356 (E356) in the third transmembrane domain that are critical for PKD2L1 channel function. Our study suggests that PKD2L1 may itself sense acids and defines off-response properties in the absence of PKD1L3.
Collapse
Affiliation(s)
- Shaimaa Hussein
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Wang Zheng
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Chris Dyte
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Qian Wang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - JungWoo Yang
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Fan Zhang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jingfeng Tang
- Membrane Protein Disease and Cancer Research Center, Hubei University of Technology, Wuhan, China
| | - Ying Cao
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Membrane Protein Disease and Cancer Research Center, Hubei University of Technology, Wuhan, China
| |
Collapse
|
29
|
Hu M, Liu Y, Wu J, Liu X. Influx-Operated Ca(2+) Entry via PKD2-L1 and PKD1-L3 Channels Facilitates Sensory Responses to Polymodal Transient Stimuli. Cell Rep 2015; 13:798-811. [PMID: 26489466 DOI: 10.1016/j.celrep.2015.09.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/18/2015] [Accepted: 09/13/2015] [Indexed: 12/21/2022] Open
Abstract
The polycystic TRP subfamily member PKD2-L1, in complex with PKD1-L3, is involved in physiological responses to diverse stimuli. A major challenge to understanding whether and how PKD2-L1/PKD1-L3 acts as a bona fide molecular transducer is that recombinant channels usually respond with small or undetectable currents. Here, we discover a type of Ca(2+) influx-operated Ca(2+) entry (ICE) that generates pronounced Ca(2+) spikes. Triggered by rapid onset/offset of Ca(2+), voltage, or acid stimuli, Ca(2+)-dependent activation amplifies a small Ca(2+) influx via the channel. Ca(2+) concurrently drives a self-limiting negative feedback (Ca(2+)-dependent inactivation) that is regulated by the Ca(2+)-binding EF hands of PKD2-L1. Our results suggest a biphasic ICE with opposite Ca(2+) feedback regulation that facilitates sensory responses to multimodal transient stimuli. We suggest that such a mechanism may also occur for other sensory modalities and other Ca(2+) channels.
Collapse
Affiliation(s)
- Mingfeng Hu
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yuxia Liu
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jinzhi Wu
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaodong Liu
- X-Lab for Transmembrane Signaling Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China; School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
30
|
Phua SC, Lin YC, Inoue T. An intelligent nano-antenna: Primary cilium harnesses TRP channels to decode polymodal stimuli. Cell Calcium 2015; 58:415-22. [PMID: 25828566 PMCID: PMC4564334 DOI: 10.1016/j.ceca.2015.03.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 11/30/2022]
Abstract
The primary cilium is a solitary hair-like organelle on the cell surface that serves as an antenna sensing ever-changing environmental conditions. In this review, we will first recapitulate the molecular basis of the polymodal sensory function of the primary cilia, specifically focusing on transient receptor potential (TRP) channels that accumulate inside the organelle and conduct calcium ions (Ca(2+)). Each subfamily member, namely TRPP2 TRPP3, TRPC1 and TRPV4, is gated by multiple environmental factors, including chemical (receptor ligands, intracellular second messengers such as Ca(2+)), mechanical (fluid shear stress, hypo-osmotic swelling), or physical (temperature, voltage) stimuli. Both activity and heterodimer compositions of the TRP channels may be dynamically regulated for precise tuning to the varying dynamic ranges of the individual input stimuli. We will thus discuss the potential regulation of TRP channels by local second messengers. Despite its reported importance in embryonic patterning and tissue morphogenesis, the precise functional significance of the downstream Ca(2+) signals of the TRP channels remains unknown. We will close our review by featuring recent technological advances in visualizing and analyzing signal transduction inside the primary cilia, together with current perspectives illuminating the functional significance of intraciliary Ca(2+) signals.
Collapse
Affiliation(s)
- Siew Cheng Phua
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA.
| | - Yu-Chun Lin
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Takanari Inoue
- Department of Cell Biology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Center for Cell Dynamics, Institute for Basic Biomedical Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Precursory Research for Embryonic Science and Technology (PRESTO) Investigator, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan.
| |
Collapse
|
31
|
Orts-Del'Immagine A, Seddik R, Tell F, Airault C, Er-Raoui G, Najimi M, Trouslard J, Wanaverbecq N. A single polycystic kidney disease 2-like 1 channel opening acts as a spike generator in cerebrospinal fluid-contacting neurons of adult mouse brainstem. Neuropharmacology 2015. [PMID: 26220314 DOI: 10.1016/j.neuropharm.2015.07.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cerebrospinal fluid contacting neurons (CSF-cNs) are found around the central canal of all vertebrates. They present a typical morphology, with a single dendrite that projects into the cavity and ends in the CSF with a protuberance. These anatomical features have led to the suggestion that CSF-cNs might have sensory functions, either by sensing CSF movement or composition, but the physiological mechanisms for any such role are unknown. This hypothesis was recently supported by the demonstration that in several vertebrate species medullo-spinal CSF-cNs selectively express Polycystic Kidney Disease 2-Like 1 proteins (PKD2L1). PKD2L1 are members of the 'transient receptor potential (TRP)' superfamily, form non-selective cationic channels of high conductance, are regulated by various stimuli including protons and are therefore suggested to act as sensory receptors. Using patch-clamp whole-cell recordings of CSF-cNs in brainstem slices obtained from wild type and mutant PKD2L1 mice, we demonstrate that spontaneously active unitary currents in CSF-cNs are due to PKD2L1 channels that are capable, with a single opening, of triggering action potentials. Thus PKD2L1 might contribute to the setting of CSF-cN spiking activity. We also reveal that CSF-cNs have the capacity of discriminating between alkalinization and acidification following activation of specific conductances (PKD2L1 vs. ASIC) generating specific responses. Altogether, this study reinforces the idea that CSF-cNs represent sensory neurons intrinsic to the central nervous system and suggests a role for PKD2L1 channels as spike generators.
Collapse
Affiliation(s)
| | - Riad Seddik
- Aix Marseille Université, PPSN EA 4674, 13397, Marseille, France
| | - Fabien Tell
- Aix Marseille Université, CNRS, CRN2M UMR 7286, 13344, Marseille, France
| | - Coraline Airault
- Aix Marseille Université, PPSN EA 4674, 13397, Marseille, France
| | - Ghizlane Er-Raoui
- Aix Marseille Université, PPSN EA 4674, 13397, Marseille, France; Université Sultan Moulay Slimane, 23000, Béni Mellal, Morocco
| | - Mohamed Najimi
- Université Sultan Moulay Slimane, 23000, Béni Mellal, Morocco
| | - Jérôme Trouslard
- Aix Marseille Université, PPSN EA 4674, 13397, Marseille, France.
| | | |
Collapse
|
32
|
Murayama T, Maruyama IN. Alkaline pH sensor molecules. J Neurosci Res 2015; 93:1623-30. [PMID: 26154399 DOI: 10.1002/jnr.23621] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/12/2015] [Accepted: 06/26/2015] [Indexed: 12/22/2022]
Abstract
Animals can survive only within a narrow pH range. This requires continual monitoring of environmental and body-fluid pH. Although a variety of acidic pH sensor molecules have been reported, alkaline pH sensor function is not well understood. This Review describes neuronal alkaline pH sensors, grouped according to whether they monitor extracellular or intracellular alkaline pH. Extracellular sensors include the receptor-type guanylyl cyclase, the insulin receptor-related receptor, ligand-gated Cl- channels, connexin hemichannels, two-pore-domain K+ channels, and transient receptor potential (TRP) channels. Intracellular sensors include TRP channels and gap junction channels. Identification of molecular mechanisms underlying alkaline pH sensing is crucial for understanding how animals respond to environmental alkaline pH and how body-fluid pH is maintained within a narrow range.
Collapse
Affiliation(s)
- Takashi Murayama
- Information Processing Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Ichiro N Maruyama
- Information Processing Biology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
33
|
PKD2L1/PKD1L3 channel complex with an alkali-activated mechanism and calcium-dependent inactivation. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2015; 44:483-92. [PMID: 26066678 DOI: 10.1007/s00249-015-1040-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 04/20/2015] [Accepted: 05/07/2015] [Indexed: 10/23/2022]
Abstract
Polycystic kidney disease-like (PKDL) genes that are expressed in sour taste cells have been proposed to be involved in the transduction of sourness by producing off-responses, which shows a large inward current after withdrawing the acid stimuli. However, the underlying mechanisms of off-responses are still unclear. Here, we demonstrate that an alkali-activated mechanism is responsible for eliciting off-responses, as evidenced by both experimental and theoretical analyses. In addition, we showed that the decaying phase of offset responses in PKD2L1/PKD1L3 channels was substantially accelerated by extracellular Ca(2+).
Collapse
|
34
|
Bushman JD, Ye W, Liman ER. A proton current associated with sour taste: distribution and functional properties. FASEB J 2015; 29:3014-26. [PMID: 25857556 DOI: 10.1096/fj.14-265694] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 03/11/2015] [Indexed: 02/06/2023]
Abstract
Sour taste is detected by taste receptor cells that respond to acids through yet poorly understood mechanisms. The cells that detect sour express the protein PKD2L1, which is not the sour receptor but nonetheless serves as a useful marker for sour cells. By use of mice in which the PKD2L1 promoter drives expression of yellow fluorescent protein, we previously reported that sour taste cells from circumvallate papillae in the posterior tongue express a proton current. To establish a correlation between this current and sour transduction, we examined its distribution by patch-clamp recording. We find that the current is present in PKD2L1-expressing taste cells from mouse circumvallate, foliate, and fungiform papillae but not in a variety of other cells, including spinal cord neurons that express PKD2L1. We describe biophysical properties of the current, including pH-dependent Zn(2+) inhibition, lack of voltage-dependent gating, and activation at modest pH values (6.5) that elicit action potentials in isolated cells. Consistent with a channel that is constitutively open, the cytosol of sour taste cells is acidified. These data define a functional signature for the taste cell proton current and indicate that its expression is mostly restricted to the subset of taste cells that detect sour.
Collapse
Affiliation(s)
- Jeremy D Bushman
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Wenlei Ye
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Emily R Liman
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
35
|
A novel PKD2L1 C-terminal domain critical for trimerization and channel function. Sci Rep 2015; 5:9460. [PMID: 25820328 PMCID: PMC4377555 DOI: 10.1038/srep09460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/06/2015] [Indexed: 11/08/2022] Open
Abstract
As a transient receptor potential (TRP) superfamily member, polycystic kidney disease 2-like-1 (PKD2L1) is also called TRPP3 and has similar membrane topology as voltage-gated cation channels. PKD2L1 is involved in hedgehog signaling, intestinal development, and sour tasting. PKD2L1 and PKD1L3 form heterotetramers with 3:1 stoichiometry. C-terminal coiled-coil-2 (CC2) domain (G699-W743) of PKD2L1 was reported to be important for its trimerization but independent studies showed that CC2 does not affect PKD2L1 channel function. It thus remains unclear how PKD2L1 proteins oligomerize into a functional channel. By SDS-PAGE, blue native PAGE and mutagenesis we here identified a novel C-terminal domain called C1 (K575-T622) involved in stronger homotrimerization than the non-overlapping CC2, and found that the PKD2L1 N-terminus is critical for dimerization. By electrophysiology and Xenopus oocyte expression, we found that C1, but not CC2, is critical for PKD2L1 channel function. Our co-immunoprecipitation and dynamic light scattering experiments further supported involvement of C1 in trimerization. Further, C1 acted as a blocking peptide that inhibits PKD2L1 trimerization as well as PKD2L1 and PKD2L1/PKD1L3 channel function. Thus, our study identified C1 as the first PKD2L1 domain essential for both PKD2L1 trimerization and channel function, and suggest that PKD2L1 and PKD2L1/PKD1L3 channels share the PKD2L1 trimerization process.
Collapse
|
36
|
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. [DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
37
|
Djenoune L, Khabou H, Joubert F, Quan FB, Nunes Figueiredo S, Bodineau L, Del Bene F, Burcklé C, Tostivint H, Wyart C. Investigation of spinal cerebrospinal fluid-contacting neurons expressing PKD2L1: evidence for a conserved system from fish to primates. Front Neuroanat 2014; 8:26. [PMID: 24834029 PMCID: PMC4018565 DOI: 10.3389/fnana.2014.00026] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 04/10/2014] [Indexed: 12/11/2022] Open
Abstract
Over 90 years ago, Kolmer and Agduhr identified spinal cerebrospinal fluid-contacting neurons (CSF-cNs) based on their morphology and location within the spinal cord. In more than 200 vertebrate species, they observed ciliated neurons around the central canal that extended a brush of microvilli into the cerebrospinal fluid (CSF). Although their morphology is suggestive of a primitive sensory cell, their function within the vertebrate spinal cord remains unknown. The identification of specific molecular markers for these neurons in vertebrates would benefit the investigation of their physiological roles. PKD2L1, a transient receptor potential channel that could play a role as a sensory receptor, has been found in cells contacting the central canal in mouse. In this study, we demonstrate that PKD2L1 is a specific marker for CSF-cNs in the spinal cord of mouse (Mus musculus), macaque (Macaca fascicularis) and zebrafish (Danio rerio). In these species, the somata of spinal PKD2L1+ CSF-cNs were located below or within the ependymal layer and extended an apical bulbous extension into the central canal. We found GABAergic PKD2L1-expressing CSF-cNs in all three species. We took advantage of the zebrafish embryo for its transparency and rapid development to identify the progenitor domains from which pkd2l1+ CSF-cNs originate. pkd2l1+ CSF-cNs were all GABAergic and organized in two rows—one ventral and one dorsal to the central canal. Their location and marker expression is consistent with previously described Kolmer–Agduhr cells. Accordingly, pkd2l1+ CSF-cNs were derived from the progenitor domains p3 and pMN defined by the expression of nkx2.2a and olig2 transcription factors, respectively. Altogether our results suggest that a system of CSF-cNs expressing the PKD2L1 channel is conserved in the spinal cord across bony vertebrate species.
Collapse
Affiliation(s)
- Lydia Djenoune
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France ; Muséum National d'Histoire Naturelle Paris, France ; Centre National de la Recherche Scientifique UMR 7221 Paris, France
| | - Hanen Khabou
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| | - Fanny Joubert
- UPMC Univ. Paris 06 Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR S 1158 Paris, France
| | - Feng B Quan
- Muséum National d'Histoire Naturelle Paris, France ; Centre National de la Recherche Scientifique UMR 7221 Paris, France
| | - Sophie Nunes Figueiredo
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| | - Laurence Bodineau
- UPMC Univ. Paris 06 Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR S 1158 Paris, France
| | - Filippo Del Bene
- Institut Curie Paris, France ; Centre National de la Recherche Scientifique UMR 3215 Paris, France ; Institut National de la Santé et de la Recherche Médicale U 934 Paris, France
| | - Céline Burcklé
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| | - Hervé Tostivint
- Muséum National d'Histoire Naturelle Paris, France ; Centre National de la Recherche Scientifique UMR 7221 Paris, France
| | - Claire Wyart
- Institut du Cerveau et de la Moelle Épinière, Hôpital de la Pitié-Salpêtrière Paris, France ; Institut National de la Santé et de la Recherche Médicale UMR 1127 Paris, France ; Centre National de la Recherche Scientifique UMR 7225 Paris, France ; UPMC Univ. Paris 06 Paris, France
| |
Collapse
|
38
|
The primary cilium calcium channels and their role in flow sensing. Pflugers Arch 2014; 467:157-65. [PMID: 24764075 DOI: 10.1007/s00424-014-1516-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/06/2014] [Indexed: 12/20/2022]
Abstract
The primary cilium has been the focus of intense research since it was discovered that mutations in ciliary/basal body localized proteins give rise to a multitude of disorders. While these studies have revealed the contribution of this sensory organelle to multiple signalling pathways, little is known about how it actually mediates downstream events and why its loss causes disease states. Ciliopathies are linked to defects in either structure or function of cilia and are often associated with kidney cysts. The ciliopathy, autosomal dominant polycystic kidney disease (ADPKD), is caused by mutations to the PKD1 or PKD2 gene. The PKD gene products localize to the primary cilium, where they have been proposed to form a mechanosensory complex, sensitive to flow. Since mouse knockout models of Pkd1 or Pkd2 develop structurally normal cilia, it has been hypothesized that the loss of polycystins may lead to an impairment of flow sensing. Today, technically challenging patch clamp recordings of the primary cilium have become available, and the genetic relationship between polycystins (TRPPs) and the primary cilium has recently been dissected in detail.
Collapse
|
39
|
Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94:81-140. [PMID: 24382884 DOI: 10.1152/physrev.00023.2013] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The detection and processing of painful stimuli in afferent sensory neurons is critically dependent on a wide range of different types of voltage- and ligand-gated ion channels, including sodium, calcium, and TRP channels, to name a few. The functions of these channels include the detection of mechanical and chemical insults, the generation of action potentials and regulation of neuronal firing patterns, the initiation of neurotransmitter release at dorsal horn synapses, and the ensuing activation of spinal cord neurons that project to pain centers in the brain. Long-term changes in ion channel expression and function are thought to contribute to chronic pain states. Many of the channels involved in the afferent pain pathway are permeable to calcium ions, suggesting a role in cell signaling beyond the mere generation of electrical activity. In this article, we provide a broad overview of different calcium-permeable ion channels in the afferent pain pathway and their role in pain pathophysiology.
Collapse
|
40
|
Gating modulation by heat of the polycystin transient receptor potential channel PKD2L1 (TRPP3). Pflugers Arch 2014; 466:1933-40. [PMID: 24429999 DOI: 10.1007/s00424-013-1439-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 12/30/2013] [Indexed: 10/25/2022]
Abstract
Polycystic kidney disease 2-like 1 (PKD2L1), previously called transient receptor potential polycystin 3 (TRPP3), forms a voltage-dependent nonselective cation channel that exhibits large tail currents triggered by repolarization after depolarization. Since it has previously been proposed that temperature sensitivity of some TRP channels is linked to the voltage-dependent gating, we here investigated heating effects on PKD2L1 currents in human embryonic kidney HEK293T cells overexpressing mouse PKD2L1. Tail PKD2L1 currents were increased by heating to 32 °C, but decreased at more than 36 °C. Voltage dependency of the PKD2L1 channel was shifted by heating in a bimodal fashion: an increase in temperature to 32 °C and to 36 °C shifted the activation curves toward the left and the right, respectively. In addition, heating accelerated deactivation of tail PKD2L1 currents. To analyze the channel gating kinetics, single-channel events of the PKD2L1 channel were recorded at hyperpolarized potentials under whole-cell configurations. A rise in temperature decreased the open probability of the channel. Dwell-time analysis showed that both open and closed dwell times during heating were shorter than those at room temperature. Interestingly, a rapid temperature drop after heating markedly enhanced the PKD2L1 currents at both single-channel and whole-cell levels. The rebound activation of the PKD2L1 channel was due to an increase in the open probability but not in the single-channel conductance. These results suggest that heating opens but subsequently inactivates PKD2L1 channels, which is essential for the rebound activation of the channel after heating.
Collapse
|
41
|
Abstract
It has been exciting times since the identification of polycystic kidney disease 1 (PKD1) and PKD2 as the genes mutated in autosomal dominant polycystic kidney disease (ADPKD). Biological roles of the encoded proteins polycystin-1 and TRPP2 have been deduced from phenotypes in ADPKD patients, but recent insights from vertebrate and invertebrate model organisms have significantly expanded our understanding of the physiological functions of these proteins. The identification of additional TRPP (TRPP3 and TRPP5) and polycystin-1-like proteins (PKD1L1, PKD1L2, PKD1L3, and PKDREJ) has added yet another layer of complexity to these fascinating cellular signalling units. TRPP proteins assemble with polycystin-1 family members to form receptor-channel complexes. These protein modules have important biological roles ranging from tubular morphogenesis to determination of left-right asymmetry. The founding members of the polycystin family, TRPP2 and polycystin-1, are a prime example of how studying human disease genes can provide insights into fundamental biological mechanisms using a so-called "reverse translational" approach (from bedside to bench). Here, we discuss the current literature on TRPP ion channels and polycystin-1 family proteins including expression, structure, physical interactions, physiology, and lessons from animal model systems and human disease.
Collapse
Affiliation(s)
- Mariam Semmo
- Renal Division, Department of Medicine, University Medical Centre Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany,
| | | | | |
Collapse
|
42
|
Abstract
Mechanosensitive channels allow cells to respond to changes in membrane stretch that occur due to external stimuli like pressure or flow or that occur because of osmotically induced cell swelling or shrinkage. Ion fluxes through the channels change the membrane potential and ion concentrations and link the stretch to cellular signalling. Changes in cellular activity evoked by mechanical stimuli can be used to elicit local tissue responses or can be transmitted further to generate more widespread responses. Channels can respond directly to membrane stress, can be conferred mechanosensitive by interaction with structural proteins, or can be activated by mechanosensitive signalling pathways. Because mechanosensitive channels are often nonselective cation channels, and invertebrate TRP isoforms are involved in mechanosensation, many of the mammalian TRP isoforms have been investigated with regard to their mechanosensitivity. There is evidence that members of the TRPC, TRPV, TRPM, TRPA and TRPP subfamilies could be in some way mechanosensitive, and each of the activation mechanisms described above is used by a TRP channel. TRP channels may be involved in mechanosensitive processes ranging from flow and pressure sensing in the vasculature and other organs to mechanosensation in sensory neurones and sensory organs. There is also evidence for a role of mechano- or osmosensitive TRP isoforms in osmosensing and the regulation of cell volume. Often, a number of different TRP isoforms have been implicated in a single type of mechanosensitive response. In many cases, the involvement of the isoforms needs to be confirmed, and their exact role in the signalling process determined.
Collapse
Affiliation(s)
- Tim D Plant
- Pharmakologisches Institut, BPC-Marburg, FB-Medizin, Philipps-Universität Marburg, Karl-von-Frisch-Straße 1, 35032, Marburg, Germany,
| |
Collapse
|
43
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Catterall WA, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: ion channels. Br J Pharmacol 2013; 170:1607-51. [PMID: 24528239 PMCID: PMC3892289 DOI: 10.1111/bph.12447] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Ion channels are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
- *
Author for correspondence;
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - William A Catterall
- University of Washington, School of Medicine, Department of PharmacologyBox 357280, Seattle, WA 98195-7280, USA
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
44
|
Abstract
Transient receptor potential (TRP) channels are cellular sensors for a wide spectrum of physical and chemical stimuli. They are involved in the formation of sight, hearing, touch, smell, taste, temperature, and pain sensation. TRP channels also play fundamental roles in cell signaling and allow the host cell to respond to benign or harmful environmental changes. As TRP channel activation is controlled by very diverse processes and, in many cases, exhibits complex polymodal properties, understanding how each TRP channel responds to its unique forms of activation energy is both crucial and challenging. The past two decades witnessed significant advances in understanding the molecular mechanisms that underlie TRP channels activation. This review focuses on our current understanding of the molecular determinants for TRP channel activation.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Physiology and Membrane Biology, University of California School of Medicine, Davis, California, USA.
| |
Collapse
|
45
|
Abstract
TRP channels constitute a large superfamily of cation channel forming proteins, all related to the gene product of the transient receptor potential (trp) locus in Drosophila. In mammals, 28 different TRP channel genes have been identified, which exhibit a large variety of functional properties and play diverse cellular and physiological roles. In this article, we provide a brief and systematic summary of expression, function, and (patho)physiological role of the mammalian TRP channels.
Collapse
Affiliation(s)
- Maarten Gees
- Laboratory Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Campus Gasthuisberg, Leuven, Belgium
| | | | | | | |
Collapse
|
46
|
Eijkelkamp N, Quick K, Wood JN. Transient Receptor Potential Channels and Mechanosensation. Annu Rev Neurosci 2013; 36:519-46. [DOI: 10.1146/annurev-neuro-062012-170412] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Niels Eijkelkamp
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands;
| | - Kathryn Quick
- Wolfson Institute for Biomedical Research, University College London, London WC1 6BT, United Kingdom; ,
| | - John N. Wood
- Wolfson Institute for Biomedical Research, University College London, London WC1 6BT, United Kingdom; ,
| |
Collapse
|
47
|
Yu Y, Ulbrich MH, Li MH, Dobbins S, Zhang WK, Tong L, Isacoff EY, Yang J. Molecular mechanism of the assembly of an acid-sensing receptor ion channel complex. Nat Commun 2013; 3:1252. [PMID: 23212381 PMCID: PMC3575195 DOI: 10.1038/ncomms2257] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 11/05/2012] [Indexed: 12/11/2022] Open
Abstract
Polycystic kidney disease (PKD) family proteins associate with transient receptor potential (TRP) channel family proteins to form functionally important complexes. PKD proteins differ from known ion channel-forming proteins and are generally thought to act as membrane receptors. Here we find that PKD1L3, a PKD protein, functions as a channel-forming subunit in an acid-sensing heteromeric complex formed by PKD1L3 and TRPP3, a TRP channel protein. Single amino-acid mutations in the putative pore region of both proteins alter the channel's ion selectivity. The PKD1L3/TRPP3 complex in the plasma membrane of live cells contains one PKD1L3 and three TRPP3. A TRPP3 C-terminal coiled-coil domain forms a trimer in solution and in crystal, and has a crucial role in the assembly and surface expression of the PKD1L3/TRPP3 complex. These results demonstrate that PKD subunits constitute a new class of channel-forming proteins, enriching our understanding of the function of PKD proteins and PKD/TRPP complexes.
Collapse
Affiliation(s)
- Yong Yu
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Alberto Travagli R. Neurones in the dorsal vagal complex may be more tasteful than expected. J Physiol 2012; 590:3637-8. [PMID: 22904361 DOI: 10.1113/jphysiol.2012.237750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- R Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State University – College of Medicine, Hershey, PA, USA.
| |
Collapse
|
49
|
Orts-Del'immagine A, Wanaverbecq N, Tardivel C, Tillement V, Dallaporta M, Trouslard J. Properties of subependymal cerebrospinal fluid contacting neurones in the dorsal vagal complex of the mouse brainstem. J Physiol 2012; 590:3719-41. [PMID: 22570378 DOI: 10.1113/jphysiol.2012.227959] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cerebrospinal fluid (CSF) contacting neurones have been observed in various brain regions such as the hypothalamus, the dorsal nucleus of the raphe and around the central canal (cc) of the spinal cord but their functional role remains unclear. At the level of the spinal cord, subependymal cerebrospinal fluid contacting neurones (S-CSF-cNs) present a peculiar morphology with a soma close to the ependymal layer, a process projecting towards the cc and ending with a bud and a cilium. These neurones were recently shown to express polycystin kidney disease 2-like 1 (PKD2L1 or TRPP3) channels that are members of the polycystin subtype of the transient receptor potential (TRP) channel superfamily and that have been proposed as either chemo- or mechanoreceptors in several tissues. Using immunohistological techniques and whole-cell electrophysiological recordings in brain slices obtained from PKD2L1:EGFP transgenic adult mice, we looked for and determined the functional properties of S-CSF-cNs in the dorsal vagal complex (DVC), a hindbrain structure controlling autonomic functions such as blood pressure, energy balance and food intake. Here, we demonstrate that S-CSF-cNs received GABAergic and/or glycinergic synaptic entries and were also characterised by the presence of non-selective cationic channels of large conductance that could be detected even under whole-cell configuration. The channel activity was not affected by Psalmopoeus cambridgei toxin 1, a blocker of acid sensing ion channels (ASICs), but was blocked by amiloride and by a strong extracellular acidification. In contrast, extracellular alkalinisation and hypo-osmotic shocks increased channel activity. Based on these properties, we suggest that the single-channel activity recorded in medullar S-CSF-cNs is carried by PKD2L1 channels. Our study therefore reinforces the idea that PKD2L1 is a marker of S-CSF-cNs and points toward a role for S-CSF-cNs in the detection of circulating signals and of modifications in the extracellular environment.
Collapse
Affiliation(s)
- Adeline Orts-Del'immagine
- Laboratoire de Physiologie et Physiopathologie du Système Nerveux Somato-moteur et Neurovégétatif (PPSN) EA 4674 Aix-Marseille Université (AMU), Faculté des Sciences et Techniques St. Jérôme, BP 352, Avenue Escadrille Normandie Niemen, F-13397 Marseille cedex 20, France
| | | | | | | | | | | |
Collapse
|
50
|
Yang J, Wang Q, Zheng W, Tuli J, Li Q, Wu Y, Hussein S, Dai XQ, Shafiei S, Li XG, Shen PY, Tu JC, Chen XZ. Receptor for activated C kinase 1 (RACK1) inhibits function of transient receptor potential (TRP)-type channel Pkd2L1 through physical interaction. J Biol Chem 2011; 287:6551-61. [PMID: 22174419 DOI: 10.1074/jbc.m111.305854] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Pkd2L1 (also called TRPP3) is a non-selective cation channel permeable to Ca(2+), Na(+), and K(+) and is activated by Ca(2+). It is also part of an acid-triggered off-response cation channel complex. We previously reported roles of the Pkd2L1 C-terminal fragments in its channel function, but the role of the N terminus remains unclear. Using a yeast two-hybrid screening, we found that the Pkd2L1 N terminus interacts with the receptor for activated C kinase 1 (RACK1), a scaffolding/anchoring protein implicated in various cellular functions. This interaction requires the last two Trp-Asp (WD) motifs of RACK1 and fragment Ala(19)-Pro(45) of Pkd2L1. The interaction was confirmed by GST pulldown, blot overlay, and co-immunoprecipitation assays. By (45)Ca tracer uptake and two-microelectrode voltage clamp electrophysiology, we found that in Xenopus oocytes with RACK1 overexpression Pkd2L1 channel activity is abolished or substantially reduced. Combining with oocyte surface biotinylation experiments, we demonstrated that RACK1 inhibits the function of Pkd2L1 channel on the plasma membrane in addition to reducing its total and plasma membrane expression. Overexpressing Pkd2L1 N- or C-terminal fragments as potential blocking peptides for the Pkd2L1-RACK1 interaction, we found that Pkd2L1 N-terminal fragment Met(1)-Pro(45), but not Ile(40)-Ile(97) or C-terminal fragments, abolishes the inhibition of Pkd2L1 channel by overexpressed and oocyte-native RACK1 likely through disrupting the Pkd2L1-RACK1 association. Taken together, our study demonstrated that RACK1 inhibits Pkd2L1 channel function through binding to domain Met(1)-Pro(45) of Pkd2L1. Thus, Pkd2L1 is a novel target channel whose function is regulated by the versatile scaffolding protein RACK1.
Collapse
Affiliation(s)
- Jungwoo Yang
- Department of Physiology, University of Alberta, 7-29 Medical Sciences Building, Edmonton, Alberta T6G 2H7, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|