151
|
Shen Q, Luo Z, Ma S, Tao P, Song C, Wu J, Shang W, Deng T. Bioinspired Infrared Sensing Materials and Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1707632. [PMID: 29750376 DOI: 10.1002/adma.201707632] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 02/08/2018] [Indexed: 05/26/2023]
Abstract
Bioinspired engineering offers a promising alternative approach in accelerating the development of many man-made systems. Next-generation infrared (IR) sensing systems can also benefit from such nature-inspired approach. The inherent compact and uncooled operation of biological IR sensing systems provides ample inspiration for the engineering of portable and high-performance artificial IR sensing systems. This review overviews the current understanding of the biological IR sensing systems, most of which are thermal-based IR sensors that rely on either bolometer-like or photomechanic sensing mechanism. The existing efforts inspired by the biological IR sensing systems and possible future bioinspired approaches in the development of new IR sensing systems are also discussed in the review. Besides these biological IR sensing systems, other biological systems that do not have IR sensing capabilities but can help advance the development of engineered IR sensing systems are also discussed, and the related engineering efforts are overviewed as well. Further efforts in understanding the biological IR sensing systems, the learning from the integration of multifunction in biological systems, and the reduction of barriers to maximize the multidiscipline collaborations are needed to move this research field forward.
Collapse
Affiliation(s)
- Qingchen Shen
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Zhen Luo
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shuai Ma
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Peng Tao
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Chengyi Song
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Jianbo Wu
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Wen Shang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Tao Deng
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
152
|
Seo KH, Jin Y, Jung SY, Lee SH. Comprehensive behavioral analyses of anoctamin1/TMEM16A-conditional knockout mice. Life Sci 2018; 207:323-331. [PMID: 29928889 DOI: 10.1016/j.lfs.2018.06.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/07/2018] [Accepted: 06/15/2018] [Indexed: 10/28/2022]
Abstract
AIMS Anoctamin-1 (TMEM16A) is a calcium-activated chloride channel that is involved in numerous physiological conditions. Its role has been identified in electrophysiological and histological studies of genetic knockout animals. Recent cellular localization studies have shown that anoctamin-1 is co-expressed with presynaptic proteins, therefore its role in presynaptic terminals has been suggested. However, behavioral studies are lacking because conventional knockouts of anoctamin-1 are lethal after birth. In this study, we explored the role of anoctamin-1 in presynaptic terminals by analyzing the behavior of mice with conditional knockouts of anoctamin-1 in synapsin1-expressing cells. MAIN METHODS Using a synapsin1-Cre system, we selectively ablated anoctamin-1 in synapsin1 expressing cells. The mice were used in the behavioral experiments when they were between 6 and 9 months of age. KEY FINDINGS The mice with the conditional knockout of anoctamin-1 in synapsin1-expressing cells displayed impaired social behavior. In addition, the mice showed depressive-like behavior and decreased weight. However, these animals displayed normal locomotor activity, cognitive function, and motor coordination. SIGNIFICANCE These results suggested that anoctamin-1 is involved in psychiatric behavior because of its role in the regulation of synaptic transmission in presynaptic terminals.
Collapse
Affiliation(s)
- Kyoung Hee Seo
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeonsun Jin
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sun-Young Jung
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Sung Hoon Lee
- Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
153
|
Falzone ME, Malvezzi M, Lee BC, Accardi A. Known structures and unknown mechanisms of TMEM16 scramblases and channels. J Gen Physiol 2018; 150:933-947. [PMID: 29915161 PMCID: PMC6028493 DOI: 10.1085/jgp.201711957] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
Falzone et al. interpret the mechanisms underlying the activity of TMEM16 family members from recent structural and functional work. The TMEM16 family of membrane proteins is composed of both Ca2+-gated Cl− channels and Ca2+-dependent phospholipid scramblases. The functional diversity of TMEM16s underlies their involvement in numerous signal transduction pathways that connect changes in cytosolic Ca2+ levels to cellular signaling networks. Indeed, defects in the function of several TMEM16s cause a variety of genetic disorders, highlighting their fundamental pathophysiological importance. Here, we review how our mechanistic understanding of TMEM16 function has been shaped by recent functional and structural work. Remarkably, the recent determination of near-atomic-resolution structures of TMEM16 proteins of both functional persuasions has revealed how relatively minimal rearrangements in the substrate translocation pathway are sufficient to precipitate the dramatic functional differences that characterize the family. These structures, when interpreted in the light of extensive functional analysis, point to an unusual mechanism for Ca2+-dependent activation of TMEM16 proteins in which substrate permeation is regulated by a combination of conformational rearrangements and electrostatics. These breakthroughs pave the way to elucidate the mechanistic bases of ion and lipid transport by the TMEM16 proteins and unravel the molecular links between these transport activities and their function in human pathophysiology.
Collapse
Affiliation(s)
- Maria E Falzone
- Department of Biochemistry, Weill Cornell Medical School, New York, NY
| | - Mattia Malvezzi
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Byoung-Cheol Lee
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Alessio Accardi
- Department of Biochemistry, Weill Cornell Medical School, New York, NY .,Department of Anesthesiology, Weill Cornell Medical School, New York, NY.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medical School, New York, NY
| |
Collapse
|
154
|
Hoffstaetter LJ, Bagriantsev SN, Gracheva EO. TRPs et al.: a molecular toolkit for thermosensory adaptations. Pflugers Arch 2018; 470:745-759. [PMID: 29484488 PMCID: PMC5945325 DOI: 10.1007/s00424-018-2120-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/03/2018] [Accepted: 02/05/2018] [Indexed: 12/19/2022]
Abstract
The ability to sense temperature is crucial for the survival of an organism. Temperature influences all biological operations, from rates of metabolic reactions to protein folding, and broad behavioral functions, from feeding to breeding, and other seasonal activities. The evolution of specialized thermosensory adaptations has enabled animals to inhabit extreme temperature niches and to perform specific temperature-dependent behaviors. The function of sensory neurons depends on the participation of various types of ion channels. Each of the channels involved in neuronal excitability, whether through the generation of receptor potential, action potential, or the maintenance of the resting potential have temperature-dependent properties that can tune the neuron's response to temperature stimuli. Since the function of all proteins is affected by temperature, animals need adaptations not only for detecting different temperatures, but also for maintaining sensory ability at different temperatures. A full understanding of the molecular mechanism of thermosensation requires an investigation of all channel types at each step of thermosensory transduction. A fruitful avenue of investigation into how different molecules can contribute to the fine-tuning of temperature sensitivity is to study the specialized adaptations of various species. Given the diversity of molecular participants at each stage of sensory transduction, animals have a toolkit of channels at their disposal to adapt their thermosensitivity to their particular habitats or behavioral circumstances.
Collapse
Affiliation(s)
- Lydia J Hoffstaetter
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
| |
Collapse
|
155
|
Rahmati N, Hoebeek FE, Peter S, De Zeeuw CI. Chloride Homeostasis in Neurons With Special Emphasis on the Olivocerebellar System: Differential Roles for Transporters and Channels. Front Cell Neurosci 2018; 12:101. [PMID: 29765304 PMCID: PMC5938380 DOI: 10.3389/fncel.2018.00101] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
The intraneuronal ionic composition is an important determinant of brain functioning. There is growing evidence that aberrant homeostasis of the intracellular concentration of Cl- ([Cl-]i) evokes, in addition to that of Na+ and Ca2+, robust impairments of neuronal excitability and neurotransmission and thereby neurological conditions. More specifically, understanding the mechanisms underlying regulation of [Cl-]i is crucial for deciphering the variability in GABAergic and glycinergic signaling of neurons, in both health and disease. The homeostatic level of [Cl-]i is determined by various regulatory mechanisms, including those mediated by plasma membrane Cl- channels and transporters. This review focuses on the latest advances in identification, regulation and characterization of Cl- channels and transporters that modulate neuronal excitability and cell volume. By putting special emphasis on neurons of the olivocerebellar system, we establish that Cl- channels and transporters play an indispensable role in determining their [Cl-]i and thereby their function in sensorimotor coordination.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Freek E. Hoebeek
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- NIDOD Institute, Wilhelmina Children's Hospital, University Medical Center Utrecht and Brain Center Rudolf Magnus, Utrecht, Netherlands
| | - Saša Peter
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam, Netherlands
| |
Collapse
|
156
|
Cordero-Martínez J, Reyes-Miguel T, Rodríguez-Páez L, Garduño-Siciliano L, Maldonado-García D, Roa-Espitia AL, Hernández-González EO. TMEM16A inhibition impedes capacitation and acquisition of hyperactivated motility in guinea pig sperm. J Cell Biochem 2018; 119:5944-5959. [PMID: 29600587 DOI: 10.1002/jcb.26789] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 02/12/2018] [Indexed: 11/12/2022]
Abstract
Ca2+ -activated Cl- channels (CaCCs) are anionic channels that regulate many important physiological functions associated with chloride and calcium flux in some somatic cells. The molecular identity of CaCCs was revealed to be TMEM16A and TMEM16B (also known as Anoctamin or ANO1 and ANO2, respectively) in all eukaryotes. A recent study suggests the presence of TMEM16A in human sperm and a relationship with the rhZP-induced acrosome reaction. However, to the best of our knowledge, little is known about the role of TMEM16A in other spermatic processes such as capacitation or motility. In this study, we evaluated the effects of two TMEM16A antagonists on capacitation, acrosome reaction, and motility in guinea pig sperm; these antagonists were T16Ainh-A01, belonging to a second generation of potent antagonists of TMEM16A, and niflumic acid (NFA), a well-known antagonist of TMEM16A (CaCCs). First of all, we confirmed that the absence of Cl- in the capacitation medium changes motility parameters, capacitation, and the progesterone-induced acrosome reaction. Using a specific antibody, TMEM16A was found as a protein band of ∼120 kDa, which localization was in the apical crest of the acrosome and the middle piece of the flagellum. Inhibition of TMEM16A by T16Ainh-A01 affected sperm physiology by reducing capacitation, blocking the progesterone-induced acrosome reaction under optimal capacitation conditions, inhibiting progressive motility, and the acquisition of hyperactivated motility, diminishing [Ca2+ ]i, and increasing [Cl- ]i. These changes in sperm kinematic parameters provide new evidence of the important role played by TMEM16A in the production of sperm capable of fertilizing oocytes.
Collapse
Affiliation(s)
- Joaquín Cordero-Martínez
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, San Pedro Zacatenco, Del. Gustavo A. Madero, México City, Mexico.,Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Prolongación Manuel Carpio y Plan de Ayala s/n Col, Santo Tomás, Del. Miguel Hidalgo, México City, Mexico
| | - Tania Reyes-Miguel
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, San Pedro Zacatenco, Del. Gustavo A. Madero, México City, Mexico
| | - Lorena Rodríguez-Páez
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Prolongación Manuel Carpio y Plan de Ayala s/n Col, Santo Tomás, Del. Miguel Hidalgo, México City, Mexico
| | - Leticia Garduño-Siciliano
- Laboratorio de Toxicología de Productos Naturales. Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Prol. Av. Wilfrido Massieu s/n, esq. Manuel L. Stampa, Col. Unidad Profesional Adolfo López Mateos, Del. Gustavo A. Madero, México City, Mexico
| | - Deneb Maldonado-García
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, San Pedro Zacatenco, Del. Gustavo A. Madero, México City, Mexico
| | - Ana L Roa-Espitia
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, San Pedro Zacatenco, Del. Gustavo A. Madero, México City, Mexico
| | - Enrique O Hernández-González
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, San Pedro Zacatenco, Del. Gustavo A. Madero, México City, Mexico
| |
Collapse
|
157
|
Ferrante M, Tahvildari B, Duque A, Hadzipasic M, Salkoff D, Zagha EW, Hasselmo ME, McCormick DA. Distinct Functional Groups Emerge from the Intrinsic Properties of Molecularly Identified Entorhinal Interneurons and Principal Cells. Cereb Cortex 2018; 27:3186-3207. [PMID: 27269961 DOI: 10.1093/cercor/bhw143] [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: 11/14/2022] Open
Abstract
Inhibitory interneurons are an important source of synaptic inputs that may contribute to network mechanisms for coding of spatial location by entorhinal cortex (EC). The intrinsic properties of inhibitory interneurons in the EC of the mouse are mostly undescribed. Intrinsic properties were recorded from known cell types, such as, stellate and pyramidal cells and 6 classes of molecularly identified interneurons (regulator of calcineurin 2, somatostatin, serotonin receptor 3a, neuropeptide Y neurogliaform (NGF), neuropeptide Y non-NGF, and vasoactive intestinal protein) in acute brain slices. We report a broad physiological diversity between and within cell classes. We also found differences in the ability to produce postinhibitory rebound spikes and in the frequency and amplitude of incoming EPSPs. To understand the source of this intrinsic variability we applied hierarchical cluster analysis to functionally classify neurons. These analyses revealed physiologically derived cell types in EC that mostly corresponded to the lines identified by biomarkers with a few unexpected and important differences. Finally, we reduced the complex multidimensional space of intrinsic properties to the most salient five that predicted the cellular biomolecular identity with 81.4% accuracy. These results provide a framework for the classification of functional subtypes of cortical neurons by their intrinsic membrane properties.
Collapse
Affiliation(s)
- Michele Ferrante
- Center for Memory and Brain.,Center for Systems Neuroscience.,Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - Babak Tahvildari
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Alvaro Duque
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Muhamed Hadzipasic
- Interdepartmental Program in Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - David Salkoff
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Edward William Zagha
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| | - Michael E Hasselmo
- Center for Memory and Brain.,Center for Systems Neuroscience.,Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - David A McCormick
- Department of Neurobiology.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520-8001, USA
| |
Collapse
|
158
|
|
159
|
Danielsson J, Vink J, Hyuga S, Fu XW, Funayama H, Wapner R, Blanks AM, Gallos G. Anoctamin Channels in Human Myometrium: A Novel Target for Tocolysis. Reprod Sci 2018; 25:1589-1600. [PMID: 29471754 DOI: 10.1177/1933719118757683] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Spontaneous preterm labor leading to preterm birth is a significant obstetric problem leading to neonatal morbidity and mortality. Current tocolytics are not completely effective and novel targets may afford a therapeutic benefit. OBJECTIVE To determine whether the anoctamin (ANO) family, including the calcium-activated chloride channel ANO1, is present in pregnant human uterine smooth muscle (USM) and whether pharmacological and genetic modulation of ANO1 modulates USM contraction. METHODS Reverse transcription-polymerase chain reaction (RT-PCR), quantitative RT-PCR, and immunohistochemical staining were done to determine which members of the ANO family are expressed in human USM. Uterine smooth muscle strips were studied in an organ bath to determine whether ANO1 antagonists inhibit oxytocin-induced USM contractions. Anoctamin 1 small interfering RNA (siRNA) knockdown was performed to determine its effect on filamentous-/globular (F/G)-actin ratio, a measurement of actin polymerization's role in promoting smooth muscle contraction. RESULTS Messenger RNA (mRNA) encoding all members of the ANO family (except ANO7) are expressed in pregnant USM tissue. Anoctamin 1 mRNA expression was decreased 15.2-fold in pregnant USM compared to nonpregnant. Anoctamin 1 protein is expressed in pregnant human USM tissue. Functional organ bath studies with pregnant human USM tissue demonstrated that the ANO1 antagonist benzbromarone attenuates the force and frequency of oxytocin-induced contractions. In human USM cells, siRNA knockdown of ANO1 decreases F-/G-actin ratios. CONCLUSION Multiple members of the ANO family, including the calcium-activated chloride channel ANO1, are expressed in human USM. Antagonism of ANO1 by pharmacological inhibition and genetic knockdown leads to an attenuation of contraction in pregnant human USM. Anoctamin 1 is a potentially novel target for tocolysis.
Collapse
Affiliation(s)
- Jennifer Danielsson
- 1 Department of Anesthesiology, Columbia University Medical Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Joy Vink
- 2 Department of Obstetrics and Gynecology, Columbia University Medical Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Shunsuke Hyuga
- 1 Department of Anesthesiology, Columbia University Medical Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Xiao Wen Fu
- 1 Department of Anesthesiology, Columbia University Medical Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Hiromi Funayama
- 3 Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ronald Wapner
- 2 Department of Obstetrics and Gynecology, Columbia University Medical Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Andrew M Blanks
- 4 Cell and Developmental Biology, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - George Gallos
- 1 Department of Anesthesiology, Columbia University Medical Center, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
160
|
Peters CJ, Gilchrist JM, Tien J, Bethel NP, Qi L, Chen T, Wang L, Jan YN, Grabe M, Jan LY. The Sixth Transmembrane Segment Is a Major Gating Component of the TMEM16A Calcium-Activated Chloride Channel. Neuron 2018; 97:1063-1077.e4. [PMID: 29478917 DOI: 10.1016/j.neuron.2018.01.048] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 12/03/2017] [Accepted: 01/26/2018] [Indexed: 11/29/2022]
Abstract
Calcium-activated chloride channels (CaCCs) formed by TMEM16A or TMEM16B are broadly expressed in the nervous system, smooth muscles, exocrine glands, and other tissues. With two calcium-binding sites and a pore within each monomer, the dimeric CaCC exhibits voltage-dependent calcium sensitivity. Channel activity also depends on the identity of permeant anions. To understand how CaCC regulates neuronal signaling and how CaCC is, in turn, modulated by neuronal activity, we examined the molecular basis of CaCC gating. Here, we report that voltage modulation of TMEM16A-CaCC involves voltage-dependent occupancy of calcium- and anion-binding site(s) within the membrane electric field as well as a voltage-dependent conformational change intrinsic to the channel protein. These gating modalities all critically depend on the sixth transmembrane segment.
Collapse
Affiliation(s)
- Christian J Peters
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - John M Gilchrist
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jason Tien
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Neville P Bethel
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lijun Qi
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tingxu Chen
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lynn Wang
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh Nung Jan
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Michael Grabe
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lily Y Jan
- Departments of Physiology, Biophysics and Biochemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
161
|
Zou Y, Xu F, Tang Z, Zhong T, Cao J, Guo Q, Huang C. Distinct calcitonin gene-related peptide expression pattern in primary afferents contribute to different neuropathic symptoms following chronic constriction or crush injuries to the rat sciatic nerve. Mol Pain 2018; 12:1744806916681566. [PMID: 28256957 PMCID: PMC5521344 DOI: 10.1177/1744806916681566] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Although calcitonin gene-related peptide is a recognized pain transducer, the expression of calcitonin gene-related peptide in primary afferents may be differentially affected following different types of nerve injury. Here, we examined whether different calcitonin gene-related peptide expression patterns in primary afferents contributes to distinct sensory disturbances in three animal models of sciatic nerve injury: chronic constriction injury, mild (100g force) or strong (1000g force) transient crush in rats. Assessments of withdrawal reflexes and spontaneous behavior indicated that chronic constriction injury and mild crush resulted in positive neuropathic symptoms (static/dynamic mechanical allodynia, heat hyperalgesia, cold allodynia, spontaneous pain). However, strong crush led to both positive (dynamic mechanical allodynia, cold allodynia, spontaneous pain) and negative symptoms (static mechanical hypoesthesia, heat hypoalgesia). Calcitonin gene-related peptide immunoreactivity in dorsal root ganglia and corresponding spinal cord segments, and calcitonin gene-related peptide mRNA levels in dorsal root ganglia, indicated that the primary afferent calcitonin gene-related peptide supply was markedly reduced only after strong crush. This reduction paralleled the development of negative symptoms (static mechanical hypoesthesia and heat hypoalgesia). Administration of exogenous calcitonin gene-related peptide intrathecally after strong crush did not alter heat hypoalgesia but ameliorated static mechanical hypoesthesia, an effect blocked by a calcitonin gene-related peptide receptor antagonist. Thus, reducing the primary afferent calcitonin gene-related peptide supply contributed to subsequent negative neuropathic symptoms, especially to static mechanical stimuli. Moreover, nerve injury caused a subcellular redistribution of calcitonin gene-related peptide from small- and medium-size dorsal root ganglia neurons to large-size dorsal root ganglia neurons, which paralleled the development of positive neuropathic symptoms. Intrathecal administration of the calcitonin gene-related peptide receptor antagonist ameliorated these positive symptoms, indicating that the expression of calcitonin gene-related peptide in large-size dorsal root ganglia neurons is important for the positive neuropathic symptoms in all three models. Taken together, these results suggest that distinct calcitonin gene-related peptide expression pattern in primary afferents contribute to different neuropathic symptoms following chronic constriction or crush injuries to the rat sciatic nerve.
Collapse
Affiliation(s)
- Yu Zou
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Fangting Xu
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Zhaohui Tang
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Tao Zhong
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Jiawei Cao
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Qulian Guo
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Changsheng Huang
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
162
|
Castillo K, Diaz-Franulic I, Canan J, Gonzalez-Nilo F, Latorre R. Thermally activated TRP channels: molecular sensors for temperature detection. Phys Biol 2018; 15:021001. [PMID: 29135465 DOI: 10.1088/1478-3975/aa9a6f] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Temperature sensing is one of the oldest capabilities of living organisms, and is essential for sustaining life, because failure to avoid extreme noxious temperatures can result in tissue damage or death. A subset of members of the transient receptor potential (TRP) ion channel family is finely tuned to detect temperatures ranging from extreme cold to noxious heat, giving rise to thermoTRP channels. Structural and functional experiments have shown that thermoTRP channels are allosteric proteins, containing different domains that sense changes in temperature, among other stimuli, triggering pore opening. Although temperature-dependence is well characterized in thermoTRP channels, the molecular nature of temperature-sensing elements remains unknown. Importantly, thermoTRP channels are involved in pain sensation, related to pathological conditions. Here, we provide an overview of thermoTRP channel activation. We also discuss the structural and functional evidence supporting the existence of an intrinsic temperature sensor in this class of channels, and we explore the basic thermodynamic principles for channel activation. Finally, we give a view of their role in painful pathophysiological conditions.
Collapse
Affiliation(s)
- Karen Castillo
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso 2366103, Chile. www.cinv.cl
| | | | | | | | | |
Collapse
|
163
|
Schreiber R, Ousingsawat J, Wanitchakool P, Sirianant L, Benedetto R, Reiss K, Kunzelmann K. Regulation of TMEM16A/ANO1 and TMEM16F/ANO6 ion currents and phospholipid scrambling by Ca 2+ and plasma membrane lipid. J Physiol 2018; 596:217-229. [PMID: 29134661 PMCID: PMC5767690 DOI: 10.1113/jp275175] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS TMEM16 proteins can operate as Ca2+ -activated Cl- channels or scramble membrane phospholipids, which are both highly relevant mechanisms during disease. Overexpression of TMEM16A and TMEM16F were found to be partially active at 37°C and at resting intracellular Ca2+ concentrations. We show that TMEM16 Cl- currents and phospholipid scrambling can be activated by modification of plasma membrane phospholipids, through reactive oxygen species and phospholipase A2. While phospholipids and Cl- ions are likely to use the same pore within TMEM16F, TMEM16A only conducts Cl- ions. Lipid regulation of TMEM16 proteins is highly relevant during inflammation and regulated cell death such as apoptosis and ferroptosis. ABSTRACT TMEM16/anoctamin (ANO) proteins form Ca2+ -activated ion channels or phospholipid scramblases. We found that both TMEM16A/ANO1 and TMEM16F/ANO6 produced Cl- currents when activated by intracellular Ca2+ , but only TMEM16F was able to expose phosphatidylserine to the outer leaflet of the plasma membrane. Mutations within TMEM16F or TMEM16A/F chimeras similarly changed Cl- currents and phospholipid scrambling, suggesting the same intramolecular pathway for Cl- and phospholipids. When overexpressed, TMEM16A and TMEM16F produced spontaneous Cl- currents at 37°C even at resting intracellular Ca2+ levels, which was abolished by inhibition of phospholipase A2 (PLA2 ). Connversely, activation of PLA2 or application of active PLA2 , as well as lipid peroxidation induced by reactive oxygen species (ROS) using staurosporine or tert-butyl hydroperoxide, enhanced ion currents by TMEM16A/F and in addition activated phospholipid scrambling by TMEM16F. Thus, TMEM16 proteins are activated by an increase in intracellular Ca2+ , or independent of intracellular Ca2+ , by modifications occurring in plasma and intracellular membrane phospholipids. These results may help to explain why regions distant to the TMEM16 pore and the Ca2+ binding sites control Cl- currents and phospholipid scrambling. Regulation of TMEM16 proteins through modification of membrane phospholipids occurs during regulated cell death such as apoptosis and ferroptosis. It contributes to inflammatory and nerve injury-induced hypersensitivity and generation of pain and therefore provides a regulatory mechanism that is particularly relevant during disease.
Collapse
Affiliation(s)
- Rainer Schreiber
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Jiraporn Ousingsawat
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | | | - Lalida Sirianant
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Roberta Benedetto
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| | - Karina Reiss
- Department of DermatologyUniversity of KielSchittenhelmstrasse 7Kiel24105Germany
| | - Karl Kunzelmann
- Institut für PhysiologieUniversität RegensburgUniversitätsstraße 31D‐93053RegensburgGermany
| |
Collapse
|
164
|
Abstract
Heat sensation, the ability to detect warm and noxious temperatures, is an ancient and indispensable sensory process. Noxious temperatures can have detrimental effects on the physiology and integrity of cells, and therefore, the detection of environmental hot temperatures is absolutely crucial for survival. Temperature-sensitive ion channels, which conduct ions in a highly temperature-dependent manner, have been put forward as molecular thermometers expressed at the endings of sensory neurons. In particular, several temperature-sensitive members of the transient receptor potential (TRP) superfamily of ion channels have been identified, and a multitude of in vivo studies have shown that the capsaicin-sensitive TRPV1 channel plays a key role as a noxious heat sensor. However, Trpv1-deficient mice display a residual heat sensitivity suggesting the existence of additional heat sensor(s). In this chapter, we provide evidence for the role of the non-selective calcium-permeable TRPM3 ion channel as an additional heat sensor that acts independently of TRPV1, and give an update of the modulation of this channel by various molecular mechanisms. Finally, we compare antagonists of TRPM3 to specific blockers of TRPV1 as potential analgesic drugs to treat pathological pain.
Collapse
|
165
|
Jeon S, Caterina MJ. Molecular basis of peripheral innocuous warmth sensitivity. HANDBOOK OF CLINICAL NEUROLOGY 2018; 156:69-82. [PMID: 30454610 DOI: 10.1016/b978-0-444-63912-7.00004-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The perception of innocuous warmth is a sensory capability that facilitates thermoregulatory, social, hedonic, and even predatory functions. It has long been recognized that innocuous warmth perception is triggered by activation of a subpopulation of specially tuned peripheral thermosensory neurons. In addition, there is growing evidence that thermotransduction by nonneuronal cells, such as skin keratinocytes, might contribute to or modulate our thermosensory experience. Yet, the precise molecular mechanisms underlying warmth transduction are only now being uncovered. Recent molecular genetics approaches have led to the identification of multiple candidate warmth-transducing molecules that appear to confer thermosensitivity upon innocuous warmth afferents and/or neighboring cell types. Most, but not all, of these candidate transducers are members of the transient receptor potential (TRP) ion channel family. Among the latter, evidence supporting a function in innocuous warmth sensation is strongest for TRPV1 and TRPM2 in mammals and for TRPA1 in nonmammalian species.
Collapse
Affiliation(s)
- Sangmin Jeon
- Departments of Neurosurgery, Biological Chemistry, and Neuroscience, and Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Michael J Caterina
- Departments of Neurosurgery, Biological Chemistry, and Neuroscience, and Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
166
|
Takayama Y, Tominaga M. Involvement of TRPV1-ANO1 Interactions in Pain-Enhancing Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1099:29-36. [PMID: 30306512 DOI: 10.1007/978-981-13-1756-9_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Primary sensory neurons detect potentially dangerous environmental situations via many "sensor" proteins located on the plasma membrane. Although receptor-type cation channels are thought to be the major sensors in sensory neurons, anion channels are also important players in the peripheral nervous system. Recently, we showed that transient receptor potential vanilloid 1 (TRPV1) interacts with anoctamin 1 (ANO1, also called TMEM16A) in primary sensory neurons and that this interaction enhanced TRPV1-mediated pain sensation. In that study, we induced ANO1 currents by application of capsaicin to small DRG neurons and showed that ANO1-dependent depolarization following TRPV1 activation could evoke more action potentials. Furthermore, capsaicin-evoked pain-related behaviors in mice were strongly inhibited by a selective ANO1 blocker. Together these findings indicate that selective ANO1 inhibition can reduce pain sensation. We also investigated non-specific inhibitory effects on ion channel activities to control ion dynamics via the TRPV1-ANO1 complex. We found that 4-isopropylcyclohexanol (4-iPr-CyH-OH) had an analgesic effect on burning pain sensations through its inhibition of TRPV1 and ANO1 together. Additionally, 4-iPr-CyH-OH did not have clear agonistic effects on TRPV1, TRPA1, and ANO1 activity individually. These results indicate that 4-iPr-CyH-OH could function globally to mediate TRP-ANO1 complex functions to reduce skin hypersensitivity and could form the basis for novel analgesic agents.
Collapse
Affiliation(s)
- Y Takayama
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), Okazaki, Aichi, Japan.
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), Okazaki, Aichi, Japan
| |
Collapse
|
167
|
Abstract
The sensation of pain plays a vital protecting role, alerting organisms about potentially damaging stimuli. Tissue injury is detected by nerve endings of specialized peripheral sensory neurons called nociceptors that are equipped with different ion channels activated by thermal, mechanic, and chemical stimuli. Several transient receptor potential channels have been identified as molecular transducers of thermal stimuli in pain-sensing neurons. Skin injury or inflammation leads to increased sensitivity to thermal and mechanic stimuli, clinically defined as allodynia or hyperalgesia. This hypersensitivity is also characteristic of systemic inflammatory disorders and neuropathic pain conditions. Mechanisms of thermal hyperalgesia include peripheral sensitization of nociceptor afferents and maladaptive changes in pain-encoding neurons within the central nervous system. An important aspect of pain management involves attempts to minimize the development of nociceptor hypersensitivity. However, knowledge about the cellular and molecular mechanisms causing thermal hyperalgesia and allodynia in human subjects is still limited, and such knowledge would be an essential step for the development of more effective therapies.
Collapse
Affiliation(s)
- Félix Viana
- Alicante Institute of Neurosciences, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Spain.
| |
Collapse
|
168
|
Anoctamin 9/TMEM16J is a cation channel activated by cAMP/PKA signal. Cell Calcium 2017; 71:75-85. [PMID: 29604966 DOI: 10.1016/j.ceca.2017.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/16/2017] [Accepted: 12/26/2017] [Indexed: 12/21/2022]
Abstract
Anoctamins (ANOs) are multifunctional membrane proteins that consist of 10 homologs. ANO1 (TMEM16A) and ANO2 (TMEM16B) are anion channels activated by intracellular calcium that meditate numerous physiological functions. ANO6 is a scramblase that redistributes phospholipids across the cell membrane. The other homologs are not well characterized. We found ANO9/TMEM16J is a cation channel activated by a cAMP-dependent protein kinase A (PKA). Intracellular cAMP-activated robust currents in whole cells expressing ANO9, which were inhibited by a PKA blocker. A cholera toxin that persistently stimulated adenylate cyclase activated ANO9 as did the application of PKA. The cAMP-induced ANO9 currents were permeable to cations. The cAMP-dependent ANO9 currents were augmented by intracellular Ca2+. Ano9 transcripts were predominant in the intestines. Human intestinal SW480 cells expressed high levels of Ano9 transcripts and showed PKA inhibitor-reversible cAMP-dependent currents. We conclude that ANO9 is a cation channel activated by a cAMP/PKA pathway and could play a role in intestine function.
Collapse
|
169
|
He M, Ye W, Wang WJ, Sison ES, Jan YN, Jan LY. Cytoplasmic Cl - couples membrane remodeling to epithelial morphogenesis. Proc Natl Acad Sci U S A 2017; 114:E11161-E11169. [PMID: 29229864 PMCID: PMC5748203 DOI: 10.1073/pnas.1714448115] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Chloride is the major free anion in the extracellular space (>100 mM) and within the cytoplasm in eukaryotes (10 ∼ 20 mM). Cytoplasmic Cl- level is dynamically regulated by Cl- channels and transporters. It is well established that movement of Cl- across the cell membrane is coupled with cell excitability through changes in membrane potential and with water secretion. However, whether cytoplasmic Cl- plays additional roles in animal development and tissue homeostasis is unknown. Here we use genetics, cell biological and pharmacological tools to demonstrate that TMEM16A, an evolutionarily conserved calcium-activated chloride channel (CaCC), regulates cytoplasmic Cl- homeostasis and promotes plasma membrane remodeling required for mammalian epithelial morphogenesis. We demonstrate that TMEM16A-mediated control of cytoplasmic Cl- regulates the organization of the major phosphoinositide species PtdIns(4,5)P2 into microdomains on the plasma membrane, analogous to processes that cluster soluble and membrane proteins into phase-separated droplets. We further show that an adequate cytoplasmic Cl- level is required for proper endocytic trafficking and membrane supply during early stages of ciliogenesis and adherens junction remodeling. Our study thus uncovers a critical function of CaCC-mediated cytoplasmic Cl- homeostasis in controlling the organization of PtdIns(4,5)P2 microdomains and membrane remodeling. This newly defined role of cytoplasmic Cl- may shed light on the mechanisms of intracellular Cl- signaling events crucial for regulating tissue architecture and organelle biogenesis during animal development.
Collapse
Affiliation(s)
- Mu He
- Department of Physiology, University of California, San Francisco, CA 94158
| | - Wenlei Ye
- Department of Physiology, University of California, San Francisco, CA 94158
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Eirish S Sison
- Department of Physiology, University of California, San Francisco, CA 94158
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94158
| | - Yuh Nung Jan
- Department of Physiology, University of California, San Francisco, CA 94158
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94158
| | - Lily Yeh Jan
- Department of Physiology, University of California, San Francisco, CA 94158;
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158
- Howard Hughes Medical Institute, University of California, San Francisco, CA 94158
| |
Collapse
|
170
|
Zhang M, Gao CX, Wang YP, Ma KT, Li L, Yin JW, Dai ZG, Wang S, Si JQ. The association between the expression of PAR2 and TMEM16A and neuropathic pain. Mol Med Rep 2017; 17:3744-3750. [PMID: 29257338 PMCID: PMC5802179 DOI: 10.3892/mmr.2017.8295] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022] Open
Abstract
Chronic constriction injury (CCI) of the sciatic nerve may induce dorsal root ganglion (DRG) neuronal hyperexcitability and behaviorally expressed hyperalgesia. CCI is a model of neuropathic pain. To investigate the association between the expression of protease activated receptor 2 (PAR2), TMEM16A and neuropathic pain, the expression of PAR2 and TMEM16A proteins in the DRG neurons of rats following CCI of the sciatic nerve was investigated. Following the creation of the CCI model, the thermal withdrawal latency (TWL) was examined by a hot plate test. An immunofluorescence assay and western blot assay were performed to determine the expression of PAR2 and TMEM16A proteins in the ipsilateral L4–6 DRG neurons. The concentration of inositol 1,4,5-triphosphate (IP3) in the L4–6 DRG was determined by ELISA. In the CCI-D7 (7 days after CCI) and CCI-D14 (14 days after CCI) treatment groups, the TWL of rats was significantly shorter than that in the sham operated group (P<0.01; n=12). The expression of PAR2 and TMEM16A proteins in the CCI-D7 and CCI-D14 groups were significantly upregulated compared with the sham operated group (P<0.05; n=12). Additionally, it was revealed that PAR2 and TMEM16A were co-expressed in DRG neurons. It was also observed that IP3 significantly increased in the CCI-D7 and CCI-D14 groups compared with the sham operation group (P<0.05; n=6) as PAR2 and TMEM16A also increased. These findings suggest that the upregulation of PAR2 and TMEM16A in DRG neurons, the co-expression of the two proteins and increasing IP3 are critical to the development of neuropathic pain.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Cun-Xiang Gao
- Department of Urology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Yan-Ping Wang
- Houbo College, Xinjiang Medical University, Karamay, Xinjiang 834000, P.R. China
| | - Ke-Tao Ma
- Electrophysiological Laboratory, Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Electrophysiological Laboratory, Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jiang-Wen Yin
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Zhi-Gang Dai
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jun-Qiang Si
- Electrophysiological Laboratory, Laboratory of Xinjiang Endemic and Ethnic Diseases, Medical College, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
171
|
Yamamura H, Nishimura K, Hagihara Y, Suzuki Y, Imaizumi Y. TMEM16A and TMEM16B channel proteins generate Ca 2+-activated Cl - current and regulate melatonin secretion in rat pineal glands. J Biol Chem 2017; 293:995-1006. [PMID: 29187602 DOI: 10.1074/jbc.ra117.000326] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 11/28/2017] [Indexed: 11/06/2022] Open
Abstract
Pinealocytes regulate circadian rhythm by synthesizing and secreting melatonin. These cells generate action potentials; however, the contribution of specific ion channels to melatonin secretion from pinealocytes remains unclear. In this study, the involvement and molecular identity of Ca2+-activated Cl- (ClCa) channels in the regulation of melatonin secretion were examined in rat pineal glands. Treatment with the ClCa channel blockers, niflumic acid or T16Ainh-A01, significantly reduced melatonin secretion in pineal glands. After pineal K+ currents were totally blocked under whole-cell patch clamp conditions, depolarization and subsequent repolarization induced a slowly activating outward current and a substantial inward tail current, respectively. Both of these current changes were dependent on intracellular Ca2+ concentration and inhibited by niflumic acid and T16Ainh-A01. Quantitative real-time PCR, Western blotting, and immunocytochemical analyses revealed that TMEM16A and TMEM16B were highly expressed in pineal glands. siRNA knockdown of TMEM16A and/or TMEM16B showed that both channels contribute to ClCa currents in pinealocytes. Conversely, co-expression of TMEM16A and TMEM16B channels or the expression of this tandem channel in HEK293 cells mimicked the electrophysiological characteristics of ClCa currents in pinealocytes. Moreover, bimolecular fluorescence complementation, FRET, and co-immunoprecipitation experiments suggested that TMEM16A and TMEM16B can form heteromeric channels, as well as homomeric channels. In conclusion, pineal ClCa channels are composed of TMEM16A and TMEM16B subunits, and these fluxes regulate melatonin secretion in pineal glands.
Collapse
Affiliation(s)
- Hisao Yamamura
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Kaori Nishimura
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yumiko Hagihara
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yoshiaki Suzuki
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yuji Imaizumi
- From the Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| |
Collapse
|
172
|
Weir GA, Middleton SJ, Clark AJ, Daniel T, Khovanov N, McMahon SB, Bennett DL. Using an engineered glutamate-gated chloride channel to silence sensory neurons and treat neuropathic pain at the source. Brain 2017; 140:2570-2585. [PMID: 28969375 PMCID: PMC5841150 DOI: 10.1093/brain/awx201] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022] Open
Abstract
See Basbaum (doi:10.1093/brain/awx227) for a scientific commentary on this article. Peripheral neuropathic pain arises as a consequence of injury to sensory neurons; the development of ectopic activity in these neurons is thought to be critical for the induction and maintenance of such pain. Local anaesthetics and anti-epileptic drugs can suppress hyperexcitability; however, these drugs are complicated by unwanted effects on motor, central nervous system and cardiac function, and alternative more selective treatments to suppress hyperexcitability are therefore required. Here we show that a glutamate-gated chloride channel modified to be activated by low doses of ivermectin (but not glutamate) is highly effective in silencing sensory neurons and reversing neuropathic pain-related hypersensitivity. Activation of the glutamate-gated chloride channel expressed in either rodent or human induced pluripotent stem cell-derived sensory neurons in vitro potently inhibited their response to both electrical and algogenic stimuli. We have shown that silencing is achieved both at nerve terminals and the soma and is independent of membrane hyperpolarization and instead likely mediated by lowering of the membrane resistance. Using intrathecal adeno-associated virus serotype 9-based delivery, the glutamate-gated chloride channel was successfully targeted to mouse sensory neurons in vivo, resulting in high level and long-lasting expression of the channel selectively in sensory neurons. This enabled reproducible and reversible modulation of thermal and mechanical pain thresholds in vivo; analgesia was observed for 3 days after a single systemic dose of ivermectin. We did not observe any motor or proprioceptive deficits and noted no reduction in cutaneous afferent innervation or upregulation of the injury marker ATF3 following prolonged glutamate-gated chloride channel expression. Established mechanical and cold pain-related hypersensitivity generated by the spared nerve injury model of neuropathic pain was reversed by ivermectin treatment. The efficacy of ivermectin in ameliorating behavioural hypersensitivity was mirrored at the cellular level by a cessation of ectopic activity in sensory neurons. These findings demonstrate the importance of aberrant afferent input in the maintenance of neuropathic pain and the potential for targeted chemogenetic silencing as a new treatment modality in neuropathic pain.
Collapse
Affiliation(s)
- Greg A Weir
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alex J Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Tarun Daniel
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | | | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
173
|
Zhang Y, Zhang Z, Xiao S, Tien J, Le S, Le T, Jan LY, Yang H. Inferior Olivary TMEM16B Mediates Cerebellar Motor Learning. Neuron 2017; 95:1103-1111.e4. [PMID: 28858616 DOI: 10.1016/j.neuron.2017.08.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/17/2017] [Accepted: 08/04/2017] [Indexed: 01/31/2023]
Abstract
Ca2+-activated ion channels shape membrane excitability and Ca2+ dynamics in response to cytoplasmic Ca2+ elevation. Compared to the Ca2+-activated K+ channels, known as BK and SK channels, the physiological importance of Ca2+-activated Cl- channels (CaCCs) in neurons has been largely overlooked. Here we report that CaCCs coexist with BK and SK channels in inferior olivary (IO) neurons that send climbing fibers to innervate cerebellar Purkinje cells for the control of motor learning and timing. Ca2+ influx through the dendritic high-threshold voltage-gated Ca2+ channels activates CaCCs, which contribute to membrane repolarization of IO neurons. Loss of TMEM16B expression resulted in the absence of CaCCs in IO neurons, leading to markedly diminished action potential firing of IO neurons in TMEM16B knockout mice. Moreover, these mutant mice exhibited severe cerebellar motor learning deficits. Our findings thus advance the understanding of the neurophysiology of CaCCs and the ionic basis of IO neuron excitability.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Zhushan Zhang
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Shaohua Xiao
- Departments of Physiology, Biochemistry and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jason Tien
- Departments of Physiology, Biochemistry and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Son Le
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Trieu Le
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Lily Y Jan
- Departments of Physiology, Biochemistry and Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
174
|
Wang H, Zou L, Ma K, Yu J, Wu H, Wei M, Xiao Q. Cell-specific mechanisms of TMEM16A Ca 2+-activated chloride channel in cancer. Mol Cancer 2017; 16:152. [PMID: 28893247 PMCID: PMC5594453 DOI: 10.1186/s12943-017-0720-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/01/2017] [Indexed: 02/08/2023] Open
Abstract
TMEM16A (known as anoctamin 1) Ca2+-activated chloride channel is overexpressed in many tumors. TMEM16A overexpression can be caused by gene amplification in many tumors harboring 11q13 amplification. TMEM16A expression is also controlled in many cancer cells via transcriptional regulation, epigenetic regulation and microRNAs. In addition, TMEM16A activates different signaling pathways in different cancers, e.g. the EGFR and CAMKII signaling in breast cancer, the p38 and ERK1/2 signaling in hepatoma, the Ras-Raf-MEK-ERK1/2 signaling in head and neck squamous cell carcinoma and bladder cancer, and the NFκB signaling in glioma. Furthermore, TMEM16A overexpression has been reported to promote, inhibit, or produce no effects on cell proliferation and migration in different cancer cells. Since TMEM16A exerts different roles in different cancer cells via activation of distinct signaling pathways, we try to develop the idea that TMEM16A regulates cancer cell proliferation and migration in a cell-dependent mechanism. The cell-specific role of TMEM16A may depend on the cellular environment that is predetermined by TMEM16A overexpression mechanisms specific for a particular cancer type. TMEM16A may exert its cell-specific role via its associated protein networks, phosphorylation by different kinases, and involvement of different signaling pathways. In addition, we discuss the role of TMEM16A channel activity in cancer, and its clinical use as a prognostic and predictive marker in different cancers. This review highlights the cell-type specific mechanisms of TMEM16A in cancer, and envisions the promising use of TMEM16A inhibitors as a potential treatment for TMEM16A-overexpressing cancers.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Liang Zou
- Department of Anesthesiology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 China
| | - Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Jiankun Yu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122 China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122 China
| |
Collapse
|
175
|
Chai R, Chen Y, Yuan H, Wang X, Guo S, Qi J, Zhang H, Zhan Y, An H. Identification of Resveratrol, an Herbal Compound, as an Activator of the Calcium-Activated Chloride Channel, TMEM16A. J Membr Biol 2017; 250:483-492. [PMID: 28852814 DOI: 10.1007/s00232-017-9975-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 07/30/2017] [Indexed: 12/21/2022]
Abstract
Calcium-activated chloride channels (CaCCs) play vital roles in a variety of physiological processes. Dysfunction of the CaCCs is implicated in many diseases. Drug discovery targeting at CaCCs has recently become possible with the determination that TMEM16A is the molecular identity of CaCCs. In this study, we demonstrated that resveratrol (RES), a Chinese traditional medicine compound, is a novel activator of TMEM16A. The yellow fluorescence protein quenching assay and measurement of intracellular calcium fluorescence intensity show that RES activates TMEM16A channels in an intracellular Ca2+-independent way. The data of inside-out patch clamp revealed that RES dose-dependently activates TMEM16A (EC50 = 47.92 ± 9.35 μM). Furthermore, RES enhanced the contractions of the ileum of guinea pigs by activating the TMEM16A channel, which indicated that RES might be a promising drug for the treatment of gastrointestinal hypomotility. As RES was able to induce TMEM16A channel activation, TMEM16A can be added to the list of RES drug targets.
Collapse
Affiliation(s)
- Ran Chai
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300401, China.,Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Hongbo Yuan
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300401, China.,Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Xuzhao Wang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Shuai Guo
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Jinlong Qi
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Pharmacology and Toxicology for New Drug, Hebei Province, Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hailin Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Pharmacology and Toxicology for New Drug, Hebei Province, Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yong Zhan
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300401, China. .,Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China.
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China.
| |
Collapse
|
176
|
Strege PR, Gibbons SJ, Mazzone A, Bernard CE, Beyder A, Farrugia G. EAVK segment "c" sequence confers Ca 2+-dependent changes to the kinetics of full-length human Ano1. Am J Physiol Gastrointest Liver Physiol 2017; 312:G572-G579. [PMID: 28336549 PMCID: PMC5495914 DOI: 10.1152/ajpgi.00429.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 01/31/2023]
Abstract
Anoctamin1 (Ano1 and TMEM16A) is a calcium-activated chloride channel specifically expressed in the interstitial cells of Cajal (ICC) of the gastrointestinal tract muscularis propria. Ano1 is necessary for normal electrical slow waves and ICC proliferation. The full-length human Ano1 sequence includes an additional exon, exon "0," at the NH2 terminus. Ano1 with exon 0 [Ano1(0)] had a lower EC50 for intracellular calcium ([Ca2+]i) and faster chloride current (ICl) kinetics. The Ano1 alternative splice variant with segment "c" encoding exon 13 expresses on the first intracellular loop four additional amino acid residues, EAVK, which alter ICl at low [Ca2+]i Exon 13 is expressed in 75-100% of Ano1 transcripts in most human tissues but only 25% in the human stomach. Our aim was to determine the effect of EAVK deletion on Ano1(0)ICl parameters. By voltage-clamp electrophysiology, we examined ICl in HEK293 cells transiently expressing Ano1(0) with or without the EAVK sequence [Ano1(0)ΔEAVK]. The EC50 values of activating and deactivating ICl for [Ca2+]i were 438 ± 7 and 493 ± 9 nM for Ano1(0) but higher for Ano1(0)ΔEAVK at 746 ± 47 and 761 ± 26 nM, respectively. Meanwhile, the EC50 values for the ratio of instantaneous to steady-state ICl were not different between variants. Congruently, the time constant of activation was slower for Ano1(0)ΔEAVK than Ano1(0) currents at intermediate [Ca2+]i These results suggest that EAVK decreases the calcium sensitivity of Ano1(0) current activation and deactivation by slowing activation kinetics. Differential expression of EAVK in the human stomach may function as a switch to increase sensitivity to [Ca2+]i via faster gating of Ano1.NEW & NOTEWORTHY Calcium-activated chloride channel anoctamin1 (Ano1) is necessary for normal slow waves in the gastrointestinal interstitial cells of Cajal. Exon 0 encodes the NH2 terminus of full-length human Ano1 [Ano1(0)], while exon 13 encodes residues EAVK on its first intracellular loop. Splice variants lack EAVK more often in the stomach than other tissues. Ano1(0) without EAVK [Ano1(0)ΔEAVK] has reduced sensitivity for intracellular calcium, attributable to slower kinetics. Differential expression of EAVK may function as a calcium-sensitive switch in the human stomach.
Collapse
|
177
|
Kamaleddin MA. Molecular, biophysical, and pharmacological properties of calcium-activated chloride channels. J Cell Physiol 2017; 233:787-798. [PMID: 28121009 DOI: 10.1002/jcp.25823] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/15/2022]
Abstract
Calcium-activated chloride channels (CaCCs) are a family of anionic transmembrane ion channels. They are mainly responsible for the movement of Cl- and other anions across the biological membranes, and they are widely expressed in different tissues. Since the Cl- flow into or out of the cell plays a crucial role in hyperpolarizing or depolarizing the cells, respectively, the impact of intracellular Ca2+ concentration on these channels is attracting a lot of attentions. After summarizing the molecular, biophysical, and pharmacological properties of CaCCs, the role of CaCCs in normal cellular functions will be discussed, and I will emphasize how dysregulation of CaCCs in pathological conditions can account for different diseases. A better understanding of CaCCs and a pivotal regulatory role of Ca2+ can shed more light on the therapeutic strategies for different neurological disorders that arise from chloride dysregulation, such as asthma, cystic fibrosis, and neuropathic pain.
Collapse
Affiliation(s)
- Mohammad Amin Kamaleddin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.,Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
178
|
Affiliation(s)
- Nikita Gamper
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.,Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
179
|
Qu L, Fu K, Shimada SG, LaMotte RH. Cl - channel is required for CXCL10-induced neuronal activation and itch response in a murine model of allergic contact dermatitis. J Neurophysiol 2017; 118:619-624. [PMID: 28446581 DOI: 10.1152/jn.00187.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/14/2017] [Accepted: 04/26/2017] [Indexed: 12/21/2022] Open
Abstract
Persistent itch often accompanies allergic contact dermatitis (ACD), but the underlying mechanisms remain largely unexplored. We previously demonstrated that CXCL10/CXCR3 signaling activated a subpopulation of cutaneous primary sensory neurons and mediated itch response after contact hypersensitivity (CHS), a murine model of ACD, induced by squaric acid dibutylester. The purpose of this study was to determine the ionic mechanisms underlying CXCL10-induced neuronal activation and allergic itch. In whole cell recordings, CXCL10 triggered a current in dorsal root ganglion (DRG) neurons innervating the area of CHS. This current was modulated by intracellular Cl- and blocked by the general Cl- channel inhibitors. Moreover, increasing Ca2+ buffering capacity reduced this current. In addition, blockade of Cl- channels significantly suppressed CXCL10-induced Ca2+ response. In behavioral tests, injection of CXCL10 into CHS site exacerbated itch-related scratching behaviors. Moreover, the potentiating behavioral effects of CXCL10 were attenuated by either of two Cl- channel blockers. Thus we suggest that the Cl- channel acts as a downstream target mediating the excitatory and pruritic behavioral effects of CXCL10. Cl- channels may provide a promising therapeutic target for the treatment of allergic itch in which CXCL10/CXCR3 signaling may participate.NEW & NOTEWORTHY The ionic mechanisms underlying CXCL10-induced neuronal activation and allergic itch are largely unexplored. This study revealed that CXCL10 evoked an ionic current mainly carried by Cl- channels. We suggest that Cl- channels are likely key molecular candidates responsible for the CXCL10-evoked neuronal activation and itch-like behaviors in a murine model of allergic contact dermatitis induced by the antigen squaric acid dibutylester. Cl- channels may emerge as a promising drug target for the treatment of allergic itch in which CXCL10/CXCR3 signaling may participate.
Collapse
Affiliation(s)
- Lintao Qu
- Departments of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, Maryland; and .,Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut
| | - Kai Fu
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut
| | - Steven G Shimada
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut
| | - Robert H LaMotte
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
180
|
Seo Y, Ryu K, Park J, Jeon DK, Jo S, Lee HK, Namkung W. Inhibition of ANO1 by luteolin and its cytotoxicity in human prostate cancer PC-3 cells. PLoS One 2017; 12:e0174935. [PMID: 28362855 PMCID: PMC5376326 DOI: 10.1371/journal.pone.0174935] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/17/2017] [Indexed: 11/18/2022] Open
Abstract
Anoctamin 1 (ANO1), a calcium-activated chloride channel, is highly amplified in prostate cancer, the most common form of cancer and leading causes of cancer death in men, and downregulation of ANO1 expression or its functional activity is known to inhibit cell proliferation, migration and invasion in prostate cancer cells. Here, we performed a cell-based screening for the identification of ANO1 inhibitors as potential anticancer therapeutic agents for prostate cancer. Screening of ~300 selected bioactive natural products revealed that luteolin is a novel potent inhibitor of ANO1. Electrophysiological studies indicated that luteolin potently inhibited ANO1 chloride channel activity in a dose-dependent manner with an IC50 value of 9.8 μM and luteolin did not alter intracellular calcium signaling in PC-3 prostate cancer cells. Luteolin inhibited cell proliferation and migration of PC-3 cells expressing high levels of ANO1 more potently than that of ANO1-deficient PC-3 cells. Notably, luteolin not only inhibited ANO1 channel activity, but also strongly decreased protein expression levels of ANO1. Our results suggest that downregulation of ANO1 by luteolin is a potential mechanism for the anticancer effect of luteolin.
Collapse
Affiliation(s)
- Yohan Seo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, Korea
| | - Kunhi Ryu
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Jinhong Park
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Dong-kyu Jeon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Sungwoo Jo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Ho K. Lee
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, Korea
| | - Wan Namkung
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
181
|
Malysz J, Gibbons SJ, Saravanaperumal SA, Du P, Eisenman ST, Cao C, Oh U, Saur D, Klein S, Ordog T, Farrugia G. Conditional genetic deletion of Ano1 in interstitial cells of Cajal impairs Ca 2+ transients and slow waves in adult mouse small intestine. Am J Physiol Gastrointest Liver Physiol 2017; 312:G228-G245. [PMID: 27979828 PMCID: PMC5401988 DOI: 10.1152/ajpgi.00363.2016] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 01/31/2023]
Abstract
Myenteric plexus interstitial cells of Cajal (ICC-MY) in the small intestine are Kit+ electrical pacemakers that express the Ano1/TMEM16A Ca2+-activated Cl- channel, whose functions in the gastrointestinal tract remain incompletely understood. In this study, an inducible Cre-LoxP-based approach was used to advance the understanding of Ano1 in ICC-MY of adult mouse small intestine. KitCreERT2/+;Ano1Fl/Fl mice were treated with tamoxifen or vehicle, and small intestines (mucosa free) were examined. Quantitative RT-PCR demonstrated ~50% reduction in Ano1 mRNA in intestines of conditional knockouts (cKOs) compared with vehicle-treated controls. Whole mount immunohistochemistry showed a mosaic/patchy pattern loss of Ano1 protein in ICC networks. Ca2+ transients in ICC-MY network of cKOs displayed reduced duration compared with highly synchronized controls and showed synchronized and desynchronized profiles. When matched, the rank order for Ano1 expression in Ca2+ signal imaged fields of view was as follows: vehicle controls>>>cKO(synchronized)>cKO(desynchronized). Maintenance of Ca2+ transients' synchronicity despite high loss of Ano1 indicates a large functional reserve of Ano1 in the ICC-MY network. Slow waves in cKOs displayed reduced duration and increased inter-slow-wave interval and occurred in regular- and irregular-amplitude oscillating patterns. The latter activity suggested ongoing interaction by independent interacting oscillators. Lack of slow waves and depolarization, previously reported for neonatal constitutive knockouts, were also seen. In summary, Ano1 in adults regulates gastrointestinal function by determining Ca2+ transients and electrical activity depending on the level of Ano1 expression. Partial Ano1 loss results in Ca2+ transients and slow waves displaying reduced duration, while complete and widespread absence of Ano1 in ICC-MY causes lack of slow wave and desynchronized Ca2+ transients.NEW & NOTEWORTHY The Ca2+-activated Cl- channel, Ano1, in interstitial cells of Cajal (ICC) is necessary for normal gastrointestinal motility. We knocked out Ano1 to varying degrees in ICC of adult mice. Partial knockout of Ano1 shortened the widths of electrical slow waves and Ca2+ transients in myenteric ICC but Ca2+ transient synchronicity was preserved. Near-complete knockout was necessary for transient desynchronization and loss of slow waves, indicating a large functional reserve of Ano1 in ICC.
Collapse
Affiliation(s)
- John Malysz
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | - Simon J Gibbons
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | | | - Peng Du
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Seth T Eisenman
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | - Chike Cao
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | - Uhtaek Oh
- Sensory Research Center, CRI, College of Pharmacy, Seoul National University, Seoul, Republic of Korea; and
| | - Dieter Saur
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Sabine Klein
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Tamas Ordog
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
182
|
4-isopropylcyclohexanol has potential analgesic effects through the inhibition of anoctamin 1, TRPV1 and TRPA1 channel activities. Sci Rep 2017; 7:43132. [PMID: 28225032 PMCID: PMC5320485 DOI: 10.1038/srep43132] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/19/2017] [Indexed: 12/31/2022] Open
Abstract
Interactions between calcium-activated chloride channel anoctamin 1 (ANO1) and transient receptor potential vanilloid 1 (TRPV1) enhance pain sensations in mice, suggesting that ANO1 inhibition could have analgesic effects. Here we show that menthol and the menthol analogue isopropylcyclohexane (iPr-CyH) inhibited ANO1 channels in mice. The iPr-CyH derivative 4-isopropylcyclohexanol (4-iPr-CyH-OH) inhibited mouse ANO1 currents more potently than iPr-CyH. Moreover, 4-iPr-CyH-OH inhibited the activities of TRPV1, TRP ankyrin 1 (TRPA1), TRP melastatin 8 (TRPM8) and TRPV4. Single-channel analysis revealed that 4-iPr-CyH-OH reduced TRPV1 and TRPA1 current open-times without affecting unitary amplitude or closed-time, suggesting that it affected gating rather than blocking the channel pore. The ability of 4-iPr-CyH-OH to inhibit action potential generation and reduce pain-related behaviors induced by capsaicin in mice suggests that 4-iPr-CyH-OH could have analgesic applications. Thus, 4-iPr-CyH-OH is a promising base chemical to develop novel analgesics that target ANO1 and TRP channels.
Collapse
|
183
|
FGF13 Selectively Regulates Heat Nociception by Interacting with Nav1.7. Neuron 2017; 93:806-821.e9. [DOI: 10.1016/j.neuron.2017.01.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/15/2016] [Accepted: 01/04/2017] [Indexed: 12/19/2022]
|
184
|
Cruz-Rangel S, De Jesús-Pérez JJ, Aréchiga-Figueroa IA, Rodríguez-Menchaca AA, Pérez-Cornejo P, Hartzell HC, Arreola J. Extracellular protons enable activation of the calcium-dependent chloride channel TMEM16A. J Physiol 2017; 595:1515-1531. [PMID: 27859335 DOI: 10.1113/jp273111] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/27/2016] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS The calcium-activated chloride channel TMEM16A provides a pathway for chloride ion movements that are key in preventing polyspermy, allowing fluid secretion, controlling blood pressure, and enabling gastrointestinal activity. TMEM16A is opened by voltage-dependent calcium binding and regulated by permeant anions and intracellular protons. Here we show that a low proton concentration reduces TMEM16A activity while maximum activation is obtained when the external proton concentration is high. In addition, protonation conditions determine the open probability of TMEM16A without changing its calcium sensitivity. External glutamic acid 623 (E623) is key for TMEM16A's ability to respond to external protons. At physiological pH, E623 is un-protonated and TMEM16A is activated when intracellular calcium increases; however, under acidic conditions E623 is partially protonated and works synergistically with intracellular calcium to activate the channel. These findings are critical for understanding physiological and pathological processes that involve changes in pH and chloride flux via TMEM16A. ABSTRACT Transmembrane protein 16A (TMEM16A), also known as ANO1, the pore-forming subunit of a Ca2+ -dependent Cl- channel (CaCC), is activated by direct, voltage-dependent, binding of intracellular Ca2+ . Endogenous CaCCs are regulated by extracellular protons; however, the molecular basis of such regulation remains unidentified. Here, we evaluated the effects of different extracellular proton concentrations ([H+ ]o ) on mouse TMEM16A expressed in HEK-293 cells using whole-cell and inside-out patch-clamp recordings. We found that increasing the [H+ ]o from 10-10 to 10-5.5 m caused a progressive increase in the chloride current (ICl ) that is described by titration of a protonatable site with pK = 7.3. Protons regulate TMEM16A in a voltage-independent manner, regardless of channel state (open or closed), and without altering its apparent Ca2+ sensitivity. Noise analysis showed that protons regulate TMEM16A by tuning its open probability without modifying the single channel current. We found a robust reduction of the proton effect at high [Ca2+ ]i . To identify protonation targets we mutated all extracellular glutamate and histidine residues and 4 of 11 aspartates. Most mutants were sensitive to protons. However, mutation that substituted glutamic acid (E) for glutamine (Q) at amino acid position 623 (E623Q) displayed a titration curve shifted to the left relative to wild type channels and the ICl was nearly insensitive to proton concentrations between 10-5.5 and 10-9.0 m. Additionally, ICl of the mutant containing an aspartic acid (D) to asparagine (N) substitution at position 405 (D405N) mutant was partially inhibited by a proton concentration of 10-5.5 m, but 10-9.0 m produced the same effect as in wild type. Based on our findings we propose that external protons titrate glutamic acid 623, which enables voltage activation of TMEM16A at non-saturating [Ca2+ ]i .
Collapse
Affiliation(s)
- Silvia Cruz-Rangel
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, San Luis Potosí, SLP, 78290, México
| | - José J De Jesús-Pérez
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, San Luis Potosí, SLP, 78290, México
| | - Iván A Aréchiga-Figueroa
- CONACYT-Universidad Autónoma de San Luis Potosí School of Medicine, Ave. V. Carranza 2405, San Luis Potosí, SLP, 78290, México
| | - Aldo A Rodríguez-Menchaca
- Department of Physiology and Biophysics, Universidad Autónoma de San Luis Potosí School of Medicine, Ave. V. Carranza 2405, San Luis Potosí, SLP, 78290, México
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, Universidad Autónoma de San Luis Potosí School of Medicine, Ave. V. Carranza 2405, San Luis Potosí, SLP, 78290, México
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, Ave. Dr. Manuel Nava #6, San Luis Potosí, SLP, 78290, México
| |
Collapse
|
185
|
Han Q, Kim YH, Wang X, Liu D, Zhang ZJ, Bey AL, Lay M, Chang W, Berta T, Zhang Y, Jiang YH, Ji RR. SHANK3 Deficiency Impairs Heat Hyperalgesia and TRPV1 Signaling in Primary Sensory Neurons. Neuron 2016; 92:1279-1293. [PMID: 27916453 DOI: 10.1016/j.neuron.2016.11.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 09/04/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022]
Abstract
Abnormal pain sensitivity is commonly associated with autism spectrum disorders (ASDs) and affects the life quality of ASD individuals. SHANK3 deficiency was implicated in ASD and pain dysregulation. Here, we report functional expression of SHANK3 in mouse dorsal root ganglion (DRG) sensory neurons and spinal cord presynaptic terminals. Homozygous and heterozygous Shank3 complete knockout (Δe4-22) results in impaired heat hyperalgesia in inflammatory and neuropathic pain. Specific deletion of Shank3 in Nav1.8-expressing sensory neurons also impairs heat hyperalgesia in homozygous and heterozygous mice. SHANK3 interacts with transient receptor potential subtype V1 (TRPV1) via Proline-rich region and regulates TRPV1 surface expression. Furthermore, capsaicin-induced spontaneous pain, inward currents in DRG neurons, and synaptic currents in spinal cord neurons are all reduced after Shank3 haploinsufficiency. Finally, partial knockdown of SHANK3 expression in human DRG neurons abrogates TRPV1 function. Our findings reveal a peripheral mechanism of SHANK3, which may underlie pain deficits in SHANK3-related ASDs.
Collapse
Affiliation(s)
- Qingjian Han
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yong Ho Kim
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiaoming Wang
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Di Liu
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zhi-Jun Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alexandra L Bey
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mark Lay
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Wonseok Chang
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Temugin Berta
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yan Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yong-Hui Jiang
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
186
|
Choi G, Hwang SW. Modulation of the Activities of Neuronal Ion Channels by Fatty Acid-Derived Pro-Resolvents. Front Physiol 2016; 7:523. [PMID: 27877134 PMCID: PMC5099253 DOI: 10.3389/fphys.2016.00523] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022] Open
Abstract
Progress of inflammation depends on the balance between two biological mechanisms: pro-inflammatory and pro-resolving processes. Many extracellular and intracellular molecular components including cytokines, growth factors, steroids, neurotransmitters, and lipidergic mediators and their receptors contribute to the two processes, generated from cellular participants during inflammation. Fatty acid-derived mediators are crucial in directing the inflammatory phase and orchestrating heterogeneous reactions of participants such as inflamed cells, innate immune cells, vascular components, innervating neurons, etc. As well as activating specific types of receptor molecules, lipidergic mediators can actively control the functions of various ion channels via direct binding and/or signal transduction, thereby altering cellular functions. Lipid mediators can be divided into two classes based on which of the two processes they promote: pro-inflammatory, which includes prostaglandins and leukotrienes, and pro-resolving, which includes lipoxins, resolvins, and maresins. The research on the modulations of neuronal ion channels regarding the actions of the pro-inflammatory class has begun relatively earlier while the focus is currently expanding to cover the ion channel interaction with pro-resolvents. As a result, knowledge of inhibitory mechanisms by the pro-resolvents, historically seldom found for other known endogenous modulators or pro-inflammatory mediators, is accumulating particularly upon sensory neuronal cation channels. Diverse mechanistic explanations at molecular levels are being proposed and refined. Here we overviewed the interactions of lipidergic pro-resolvents with neuronal ion channels and outcomes from the interactions, focusing on transient receptor potential (TRP) ion channels. We also discuss unanswered hypotheses and perspectives regarding their interactions.
Collapse
Affiliation(s)
- Geunyeol Choi
- Department of Biomedical Sciences, Korea University Seoul, South Korea
| | - Sun Wook Hwang
- Department of Biomedical Sciences, Korea UniversitySeoul, South Korea; Department of Physiology, Korea University College of MedicineSeoul, South Korea
| |
Collapse
|
187
|
Abstract
The mammalian nervous system encodes many different forms of pain, from those that arise as a result of short-term low-grade interactions with noxious thermal, chemical, or mechanical sources to more serious forms of pain induced by trauma and disease. In this Review, we highlight recent advances in our understanding of the neural circuits that encode these types of pain. Promising therapeutic strategies based on recent advances are also highlighted.
Collapse
Affiliation(s)
- Cedric Peirs
- Departments of Neurobiology and Otolaryngology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, BST3, Pittsburgh, PA 15213, USA
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Rebecca P Seal
- Departments of Neurobiology and Otolaryngology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, BST3, Pittsburgh, PA 15213, USA.
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
188
|
Takkala P, Zhu Y, Prescott SA. Combined Changes in Chloride Regulation and Neuronal Excitability Enable Primary Afferent Depolarization to Elicit Spiking without Compromising its Inhibitory Effects. PLoS Comput Biol 2016; 12:e1005215. [PMID: 27835641 PMCID: PMC5105942 DOI: 10.1371/journal.pcbi.1005215] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/20/2016] [Indexed: 11/19/2022] Open
Abstract
The central terminals of primary afferent fibers experience depolarization upon activation of GABAA receptors (GABAAR) because their intracellular chloride concentration is maintained above electrochemical equilibrium. Primary afferent depolarization (PAD) normally mediates inhibition via sodium channel inactivation and shunting but can evoke spikes under certain conditions. Antidromic (centrifugal) conduction of these spikes may contribute to neurogenic inflammation while orthodromic (centripetal) conduction could contribute to pain in the case of nociceptive fibers. PAD-induced spiking is assumed to override presynaptic inhibition. Using computer simulations and dynamic clamp experiments, we sought to identify which biophysical changes are required to enable PAD-induced spiking and whether those changes necessarily compromise PAD-mediated inhibition. According to computational modeling, a depolarizing shift in GABA reversal potential (EGABA) and increased intrinsic excitability (manifest as altered spike initiation properties) were necessary for PAD-induced spiking, whereas increased GABAAR conductance density (ḡGABA) had mixed effects. We tested our predictions experimentally by using dynamic clamp to insert virtual GABAAR conductances with different EGABA and kinetics into acutely dissociated dorsal root ganglion (DRG) neuron somata. Comparable experiments in central axon terminals are prohibitively difficult but the biophysical requirements for PAD-induced spiking are arguably similar in soma and axon. Neurons from naïve (i.e. uninjured) rats were compared before and after pharmacological manipulation of intrinsic excitability, and against neurons from nerve-injured rats. Experimental data confirmed that, in most neurons, both predicted changes were necessary to yield PAD-induced spiking. Importantly, such changes did not prevent PAD from inhibiting other spiking or from blocking spike propagation. In fact, since the high value of ḡGABA required for PAD-induced spiking still mediates strong inhibition, we conclude that PAD-induced spiking does not represent failure of presynaptic inhibition. Instead, diminished PAD caused by reduction of ḡGABA poses a greater risk to presynaptic inhibition and the sensory processing that relies upon it.
Collapse
Affiliation(s)
- Petri Takkala
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Yi Zhu
- Center for Pain Research, Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Steven A. Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Center for Pain Research, Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Physiology and the Institute of Biomaterials and Biomedical Engineering, University of Toronto, Ontario, Canada
| |
Collapse
|
189
|
Pang RP, Xie MX, Yang J, Shen KF, Chen X, Su YX, Yang C, Tao J, Liang SJ, Zhou JG, Zhu HQ, Wei XH, Li YY, Qin ZH, Liu XG. Downregulation of ClC-3 in dorsal root ganglia neurons contributes to mechanical hypersensitivity following peripheral nerve injury. Neuropharmacology 2016; 110:181-189. [DOI: 10.1016/j.neuropharm.2016.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 01/18/2023]
|
190
|
Buntinx L, Voets T, Morlion B, Vangeel L, Janssen M, Cornelissen E, Vriens J, de Hoon J, Levtchenko E. TRPV1 dysfunction in cystinosis patients harboring the homozygous 57 kb deletion. Sci Rep 2016; 6:35395. [PMID: 27734949 PMCID: PMC5062165 DOI: 10.1038/srep35395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/27/2016] [Indexed: 11/12/2022] Open
Abstract
Cystinosis is a rare autosomal recessive disorder characterized by lysosomal cystine accumulation due to loss of function of the lysosomal cystine transporter (CTNS). The most common mutation in cystinosis patients of Northern Europe consists of a 57-kb deletion. This deletion not only inactivates the CTNS gene but also extends into the non-coding region upstream of the start codon of the TRPV1 gene, encoding the capsaicin- and heat-sensitive ion channel TRPV1. To evaluate the consequences of the 57-kb deletion on functional TRPV1 expression, we compared thermal, mechanical and chemical sensitivity of cystinosis patients with matched healthy controls. Whereas patients heterozygous for the 57-kb deletion showed normal sensory responses, homozygous subjects exhibited a 60% reduction in vasodilation and pain evoked by capsaicin, as well as an increase in heat detection threshold. Responses to cold, mechanical stimuli or cinnamaldehyde, an agonist of the related nociceptor channel TRPA1, were unaltered. We conclude that cystinosis patients homozygous for the 57-kb deletion exhibit a strong reduction of TRPV1 function, leading to sensory deficiencies akin to the phenotype of TRPV1-deficient mice. These deficits may account for the reported sensory alterations and thermoregulatory deficits in these patients, and provide a paradigm for life-long TRPV1 deficiency in humans.
Collapse
Affiliation(s)
- L Buntinx
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - T Voets
- Department of Cellular and Molecular Medicine, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - B Morlion
- Center for algology and pain management, Department of Cardiovascular Sciences, KULeuven, Weligerveld 1, 3212 Pellenberg, Belgium
| | - L Vangeel
- Department of Cellular and Molecular Medicine, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - M Janssen
- Department of internal medicine, Radboud UMC Nijmegen, Geert Grooteplein-Zuid 22, 6525 GA Nijmegen, The Netherlands
| | - E Cornelissen
- Department of Pediatric Nephrology, Radboud UMC Nijmegen, Geert Grooteplein-Zuid 22, 6525 GA Nijmegen, The Netherlands
| | - J Vriens
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - J de Hoon
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - E Levtchenko
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
191
|
Ma K, Wang H, Yu J, Wei M, Xiao Q. New Insights on the Regulation of Ca 2+ -Activated Chloride Channel TMEM16A. J Cell Physiol 2016; 232:707-716. [PMID: 27682822 DOI: 10.1002/jcp.25621] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022]
Abstract
TMEM16A, also known as anoctamin 1, is a recently identified Ca2+ -activated chloride channel and the first member of a 10-member TMEM16 family. TMEM16A dysfunction is implicated in many diseases such as cancer, hypertension, and cystic fibrosis. TMEM16A channels are well known to be dually regulated by voltage and Ca2+ . In addition, recent studies have revealed that TMEM16A channels are regulated by many molecules such as calmodulin, protons, cholesterol, and phosphoinositides, and a diverse range of stimuli such as thermal and mechanical stimuli. A better understanding of the regulatory mechanisms of TMEM16A is important to understand its physiological and pathological role. Recently, the crystal structure of a TMEM16 family member from the fungus Nectria haematococcaten (nhTMEM16) is discovered, and provides valuable information for studying the structure and function of TMEM16A. In this review, we discuss the structure and function of TMEM16A channels based on the crystal structure of nhTMEM16A and focus on the regulatory mechanisms of TMEM16A channels. J. Cell. Physiol. 232: 707-716, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ke Ma
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Hui Wang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Jiankun Yu
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, P. R. China
| |
Collapse
|
192
|
Cold sensitivity of TRPA1 is unveiled by the prolyl hydroxylation blockade-induced sensitization to ROS. Nat Commun 2016; 7:12840. [PMID: 27628562 PMCID: PMC5027619 DOI: 10.1038/ncomms12840] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 08/05/2016] [Indexed: 11/08/2022] Open
Abstract
Mammalian transient receptor potential ankyrin 1 (TRPA1) is a polymodal nociceptor that plays an important role in pain generation, but its role as a cold nociceptor is still controversial. Here, we propose that TRPA1 can sense noxious cold via transduction of reactive oxygen species (ROS) signalling. We show that inhibiting hydroxylation of a proline residue within the N-terminal ankyrin repeat of human TRPA1 by mutation or using a prolyl hydroxylase (PHD) inhibitor potentiates the cold sensitivity of TRPA1 in the presence of hydrogen peroxide. Inhibiting PHD in mice triggers mouse TRPA1 sensitization sufficiently to sense cold-evoked ROS, which causes cold hypersensitivity. Furthermore, this phenomenon underlies the acute cold hypersensitivity induced by the chemotherapeutic agent oxaliplatin or its metabolite oxalate. Thus, our findings provide evidence that blocking prolyl hydroxylation reveals TRPA1 sensitization to ROS, which enables TRPA1 to convert ROS signalling into cold sensitivity. The transient receptor potential ankyrin 1 (TRPA1) is a cation channel that is involved in nociceptive pain sensing. Here, the authors show that hydroxylation of a proline in the N terminus of TRPA1 renders it sensitive to reactive oxygen species resulting from noxious cold.
Collapse
|
193
|
Affiliation(s)
- Ine Vandewauw
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| |
Collapse
|
194
|
The TRPM2 ion channel is required for sensitivity to warmth. Nature 2016; 536:460-3. [PMID: 27533035 PMCID: PMC5720344 DOI: 10.1038/nature19074] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/01/2016] [Indexed: 12/26/2022]
Abstract
How do we detect warmth? Thermally-activated ion channels expressed in somatosensory neurons detect the entire thermal range from extreme heat (TRPV2), painful heat (TRPV1, TRPM3, ANO1), non-painful warmth (TRPV3 and TRPV4) and non-painful coolness (TRPM8) through to painful cold (TRPA1)1–7. Genetic deletion of each of these ion channels, however, has only modest effects on thermal behaviour in mice6–12, with the exception of TRPM8, whose deletion has marked effects on the perception of moderate coolness in the range 10°C - 25°C13. The molecular mechanism responsible for detecting non-painful warmth, in particular, is unresolved. Here we used calcium imaging to identify a population of novel thermally-sensitive somatosensory neurons which do not express any of the known thermally-activated TRP channels. We then used a combination of calcium imaging, electrophysiology and RNA sequencing to show that the ion channel generating heat sensitivity in these neurons is TRPM2. Autonomic neurons, usually thought of as exclusively motor, also express TRPM2 and respond directly to heat. Mice in which TRPM2 had been genetically deleted showed a striking deficit in their sensation of non-noxious warm temperatures, consistent with the idea that TRPM2 initiates a “warm” signal which drives cool-seeking behaviour.
Collapse
|
195
|
Identification and characterization of ANO9 in stage II and III colorectal carcinoma. Oncotarget 2016; 6:29324-34. [PMID: 26317553 PMCID: PMC4745729 DOI: 10.18632/oncotarget.4979] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 07/11/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The precise role and potential underlying mechanisms of anoctamin 9 (ANO9) remain largely unknown. This study aims to characterize the role and oncogenic mechanisms of ANO9 in stage II and III colorectal cancer (CRC). METHODS We examined the expression of ANO9 in colorectal cancerous tissues and cells using real-time quantitative PCR and immunohistochemistry, respectively. Multiple cellular and molecular approaches such as gene transfection, CCK-8 assay, flow cytometry, and invasion assay were also performed to explore its oncogenic mechanisms. Furthermore, the clinical significance of ANO9 in clinical CRC specimens was assessed by clinical correlation and survival analyses. RESULTS Lower expression of ANO9 messenger RNA (mRNA) was frequently detected both in CRC tissues with recurrence and metastasis-derived cell lines. Compared with matched nontumorous tissues, lower expression of ANO9 protein was observed in tumors, which was significantly correlated with tumorigenesis (p < 0.05). In vitro functional studies showed that ANO9 contributed to tumor cell proliferation, apoptosis, and invasion. Moreover, investigation of clinical CRC specimens showed that ANO9 were markedly overexpressed in metastatic tissue compared with primary tissue. Decreased expression of ANO9 was correlated with poor prognostic outcomes. CONCLUSIONS This study highlighted the role of ANO9 in progression and metastasis of stage II and III CRC. These findings suggested that up-regulation of ANO9, as a metastasis-related gene, could be a novel approach for inhibiting CRC progression.
Collapse
|
196
|
Tentonin 3/TMEM150c Confers Distinct Mechanosensitive Currents in Dorsal-Root Ganglion Neurons with Proprioceptive Function. Neuron 2016; 91:107-18. [DOI: 10.1016/j.neuron.2016.05.029] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 04/02/2016] [Accepted: 05/16/2016] [Indexed: 11/18/2022]
|
197
|
Tékus V, Horváth Á, Hajna Z, Borbély É, Bölcskei K, Boros M, Pintér E, Helyes Z, Pethő G, Szolcsányi J. Noxious heat threshold temperature and pronociceptive effects of allyl isothiocyanate (mustard oil) in TRPV1 or TRPA1 gene-deleted mice. Life Sci 2016; 154:66-74. [DOI: 10.1016/j.lfs.2016.04.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/05/2016] [Accepted: 04/23/2016] [Indexed: 01/18/2023]
|
198
|
Seo Y, Lee HK, Park J, Jeon DK, Jo S, Jo M, Namkung W. Ani9, A Novel Potent Small-Molecule ANO1 Inhibitor with Negligible Effect on ANO2. PLoS One 2016; 11:e0155771. [PMID: 27219012 PMCID: PMC4878759 DOI: 10.1371/journal.pone.0155771] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/03/2016] [Indexed: 12/13/2022] Open
Abstract
Anoctamin1 (ANO1)/transmembrane protein 16A (TMEM16A), a calcium-activated chloride channel (CaCC), is involved in many physiological functions such as fluid secretion, smooth muscle contraction, nociception and cancer progression. To date, only a few ANO1 inhibitors have been described, and these have low potency and selectivity for ANO1. Here, we performed a high-throughput screening to identify highly potent and selective small molecule inhibitors of ANO1. Three novel ANO1 inhibitors were discovered from screening of 54,400 synthetic small molecules, and they were found to fully block ANO1 channel activity with an IC50 < 3 μM. Electrophysiological analysis revealed that the most potent inhibitor, 2-(4-chloro-2-methylphenoxy)-N-[(2-methoxyphenyl)methylideneamino]-acetamide (Ani9), completely inhibited ANO1 chloride current with submicromolar potency. Notably, unlike previous small-molecule ANO1 inhibitors identified to date, Ani9 displayed high selectivity for ANO1 as compared to ANO2, which shares a high amino acid homology to ANO1. In addition, Ani9 did not affect the intracellular calcium signaling and CFTR chloride channel activity. Our results suggest that Ani9 may be a useful pharmacological tool for studying ANO1 and a potential development candidate for drug therapy of cancer, hypertension, pain, diarrhea and asthma.
Collapse
Affiliation(s)
- Yohan Seo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
| | - Ho K. Lee
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
| | - Jinhong Park
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
| | - Dong-kyu Jeon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
| | - Sungwoo Jo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
| | - Minjae Jo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
| | - Wan Namkung
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 406–840, Korea
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120–749, Korea
- * E-mail:
| |
Collapse
|
199
|
Lee YS, Lee JK, Bae Y, Lee BS, Kim E, Cho CH, Ryoo K, Yoo J, Kim CH, Yi GS, Lee SG, Lee CJ, Kang SS, Hwang EM, Park JY. Suppression of 14-3-3γ-mediated surface expression of ANO1 inhibits cancer progression of glioblastoma cells. Sci Rep 2016; 6:26413. [PMID: 27212225 PMCID: PMC4876403 DOI: 10.1038/srep26413] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/03/2016] [Indexed: 01/03/2023] Open
Abstract
Anoctamin-1 (ANO1) acts as a Ca2+-activated Cl− channel in various normal tissues, and its expression is increased in several different types of cancer. Therefore, understanding the regulation of ANO1 surface expression is important for determining its physiological and pathophysiological functions. However, the trafficking mechanism of ANO1 remains elusive. Here, we report that segment a (N-terminal 116 amino acids) of ANO1 is crucial for its surface expression, and we identified 14-3-3γ as a binding partner for anterograde trafficking using yeast two-hybrid screening. The surface expression of ANO1 was enhanced by 14-3-3γ, and the Thr9 residue of ANO1 was critical for its interaction with 14-3-3γ. Gene silencing of 14-3-3γ and/or ANO1 demonstrated that suppression of ANO1 surface expression inhibited migration and invasion of glioblastoma cells. These findings provide novel therapeutic implications for glioblastomas, which are associated with poor prognosis.
Collapse
Affiliation(s)
- Young-Sun Lee
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea.,Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea.,Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jae Kwang Lee
- Neuroscience Program, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Yeonju Bae
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Bok-Soon Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Eunju Kim
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea.,Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Chang-Hoon Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Kanghyun Ryoo
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Jiyun Yoo
- Division of Applied Life Science (BK21 plus), Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Gwan-Su Yi
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Seok-Geun Lee
- Department of Science in Korean Medicine, College of Korean Medicine, KHU-KIST department of Convergging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| | - C Justin Lee
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
200
|
Abstract
Persistent itch is a common symptom of allergic contact dermatitis (ACD) and represents a significant health burden. The chemokine CXCL10 is predominantly produced by epithelial cells during ACD. Although the chemokine CXCL10 and its receptor CXCR3 are implicated in the pathophysiology of ACD, it is largely unexplored for itch and pain accompanying this disorder. Here, we showed that CXCL10 and CXCR3 mRNA, protein, and signaling activity were upregulated in the dorsal root ganglion after contact hypersensitivity (CHS), a murine model of ACD, induced by squaric acid dibutylester. CXCL10 directly activated a subset of cutaneous dorsal root ganglion neurons innervating the area of CHS through neuronal CXCR3. In behavioral tests, a CXCR3 antagonist attenuated spontaneous itch- but not pain-like behaviors directed to the site of CHS. Injection of CXCL10 into the site of CHS elicited site-directed itch- but not pain-like behaviors, but neither type of CXCL10-evoked behaviors was observed in control mice. These results suggest that CXCL10/CXCR3 signaling mediates allergic itch but not inflammatory pain in the context of skin inflammation. Thus, upregulation of CXCL10/CXCR3 signaling in sensory neurons may contribute to itch associated with ACD. Targeting the CXCL10/CXCR3 signaling might be beneficial for the treatment of allergic itch.
Collapse
|