1
|
Goldfarb M. Fibroblast growth factor homologous factors: canonical and non-canonical mechanisms of action. J Physiol 2024; 602:4097-4110. [PMID: 39083261 DOI: 10.1113/jp286313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Since their discovery nearly 30 years ago, fibroblast growth factor homologous factors (FHFs) are now known to control the functionality of excitable tissues through a range of mechanisms. Nervous and cardiac system dysfunctions are caused by loss- or gain-of-function mutations in FHF genes. The best understood 'canonical' targets for FHF action are voltage-gated sodium channels, and recent studies have expanded the repertoire of ways that FHFs modulate sodium channel gating. Additional 'non-canonical' functions of FHFs in excitable and non-excitable cells, including cancer cells, have been reported over the past dozen years. This review summarizes and evaluates reported canonical and non-canonical FHF functions.
Collapse
Affiliation(s)
- Mitchell Goldfarb
- Department of Biological Sciences, Hunter College of City University, New York, New York, USA
- Biology Program, The Graduate Center City University, New York, New York, USA
| |
Collapse
|
2
|
Zhang C, Tong F, Zhou B, He M, Liu S, Zhou X, Ma Q, Feng T, Du WJ, Yang H, Xu H, Xiao L, Xu ZZ, Zhu C, Wu R, Wang YQ, Han Q. TMC6 functions as a GPCR-like receptor to sense noxious heat via Gαq signaling. Cell Discov 2024; 10:66. [PMID: 38886367 PMCID: PMC11183229 DOI: 10.1038/s41421-024-00678-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/08/2024] [Indexed: 06/20/2024] Open
Abstract
Thermosensation is vital for the survival, propagation, and adaption of all organisms, but its mechanism is not fully understood yet. Here, we find that TMC6, a membrane protein of unknown function, is highly expressed in dorsal root ganglion (DRG) neurons and functions as a Gαq-coupled G protein-coupled receptor (GPCR)-like receptor to sense noxious heat. TMC6-deficient mice display a substantial impairment in noxious heat sensation while maintaining normal perception of cold, warmth, touch, and mechanical pain. Further studies show that TMC6 interacts with Gαq via its intracellular C-terminal region spanning Ser780 to Pro810. Specifically disrupting such interaction using polypeptide in DRG neurons, genetically ablating Gαq, or pharmacologically blocking Gαq-coupled GPCR signaling can replicate the phenotype of TMC6 deficient mice regarding noxious heat sensation. Noxious heat stimulation triggers intracellular calcium release from the endoplasmic reticulum (ER) of TMC6- but not control vector-transfected HEK293T cell, which can be significantly inhibited by blocking PLC or IP3R. Consistently, noxious heat-induced intracellular Ca2+ release from ER and action potentials of DRG neurons largely reduced when ablating TMC6 or blocking Gαq/PLC/IP3R signaling pathway as well. In summary, our findings indicate that TMC6 can directly function as a Gαq-coupled GPCR-like receptor sensing noxious heat.
Collapse
Affiliation(s)
- Chen Zhang
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Fang Tong
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Bin Zhou
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Mingdong He
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Shuai Liu
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Xiaomeng Zhou
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Qiang Ma
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianyu Feng
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Wan-Jie Du
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Huan Yang
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Hao Xu
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Lei Xiao
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Zhen-Zhong Xu
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Cheng Zhu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, China
| | - Ruiqi Wu
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China.
| | - Yan-Qing Wang
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China.
| | - Qingjian Han
- Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Dong ZS, Zhang XR, Xue DZ, Liu JH, Yi F, Zhang YY, Xian FY, Qiao RY, Liu BY, Zhang HL, Wang C. FGF13 enhances the function of TRPV1 by stabilizing microtubules and regulates acute and chronic itch. FASEB J 2024; 38:e23661. [PMID: 38733310 DOI: 10.1096/fj.202400096r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Itching is an aversive somatosensation that triggers the desire to scratch. Transient receptor potential (TRP) channel proteins are key players in acute and chronic itch. However, whether the modulatory effect of fibroblast growth factor 13 (FGF13) on acute and chronic itch is associated with TRP channel proteins is unclear. Here, we demonstrated that conditional knockout of Fgf13 in dorsal root ganglion neurons induced significant impairment in scratching behaviors in response to acute histamine-dependent and chronic dry skin itch models. Furthermore, FGF13 selectively regulated the function of the TRPV1, but not the TRPA1 channel on Ca2+ imaging and electrophysiological recordings, as demonstrated by a significant reduction in neuronal excitability and current density induced by TRPV1 channel activation, whereas TRPA1 channel activation had no effect. Changes in channel currents were also verified in HEK cell lines. Subsequently, we observed that selective modulation of TRPV1 by FGF13 required its microtubule-stabilizing effect. Furthermore, in FGF13 knockout mice, only the overexpression of FGF13 with a tubulin-binding domain could rescue TRP channel function and the impaired itch behavior. Our findings reveal a novel mechanism by which FGF13 is involved in TRPV1-dependent itch transduction and provide valuable clues for alleviating pathological itch syndrome.
Collapse
Affiliation(s)
- Zi-Shan Dong
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
| | - Xue-Rou Zhang
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Da-Zhong Xue
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, China
| | - Jia-Hui Liu
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
| | - Fan Yi
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
| | - Yi-Yi Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
| | - Fu-Yu Xian
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
| | - Ruo-Yang Qiao
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Bo-Yi Liu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Hai-Lin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
| | - Chuan Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
4
|
Ransdell JL, Carrasquillo Y, Bosch MK, Mellor RL, Ornitz DM, Nerbonne JM. Loss of Intracellular Fibroblast Growth Factor 14 (iFGF14) Increases the Excitability of Mature Hippocampal and Cortical Pyramidal Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592532. [PMID: 38746081 PMCID: PMC11092765 DOI: 10.1101/2024.05.04.592532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Mutations in FGF14 , which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia type 27 (SCA27), a multisystem disorder associated with progressive deficits in motor coordination and cognitive function. Mice ( Fgf14 -/- ) lacking iFGF14 display similar phenotypes, and we have previously shown that the deficits in motor coordination reflect reduced excitability of cerebellar Purkinje neurons, owing to the loss of iFGF14-mediated regulation of the voltage-dependence of inactivation of the fast transient component of the voltage-gated Na + (Nav) current, I NaT . Here, we present the results of experiments designed to test the hypothesis that loss of iFGF14 also attenuates the intrinsic excitability of mature hippocampal and cortical pyramidal neurons. Current-clamp recordings from adult mouse hippocampal CA1 pyramidal neurons in acute in vitro slices, however, revealed that repetitive firing rates were higher in Fgf14 -/- , than in wild type (WT), cells. In addition, the waveforms of individual action potentials were altered in Fgf14 -/- hippocampal CA1 pyramidal neurons, and the loss of iFGF14 reduced the time delay between the initiation of axonal and somal action potentials. Voltage-clamp recordings revealed that the loss of iFGF14 altered the voltage-dependence of activation, but not inactivation, of I NaT in CA1 pyramidal neurons. Similar effects of the loss of iFGF14 on firing properties were evident in current-clamp recordings from layer 5 visual cortical pyramidal neurons. Additional experiments demonstrated that the loss of iFGF14 does not alter the distribution of anti-Nav1.6 or anti-ankyrin G immunofluorescence labeling intensity along the axon initial segments (AIS) of mature hippocampal CA1 or layer 5 visual cortical pyramidal neurons in situ . Taken together, the results demonstrate that, in contrast with results reported for neonatal (rat) hippocampal pyramidal neurons in dissociated cell culture, the loss of iFGF14 does not disrupt AIS architecture or Nav1.6 localization/distribution along the AIS of mature hippocampal (or cortical) pyramidal neurons in situ .
Collapse
|
5
|
Xu Y, Qiu Z, Gu C, Yu S, Wang S, Li C, Yao X, Li W. Propionate alleviates itch in murine models of atopic dermatitis by modulating sensory TRP channels of dorsal root ganglion. Allergy 2024; 79:1271-1290. [PMID: 38164798 DOI: 10.1111/all.15998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Itch is the most common symptom of atopic dermatitis (AD) and significantly decreases the quality of life. Skin microbiome is involved in AD pathogenesis, whereas its role in the regulation of itch remains elusive. In this study, we aimed to investigate the effects of skin microbial metabolite propionate on acute and chronic pruritus and to explore the mechanism. METHODS Using various mouse models of itch, the roles of propionate were explored by behavioral tests and histopathology/immunofluorescent analysis. Primary-cultured dorsal root ganglion neurons and HEK293 cells expressing recombinant human TRP channels were utilized for in vitro calcium imaging/in vivo miniature two-photon imaging in combination with electrophysiology and molecular docking approaches for investigation of the mechanism. RESULTS Propionate significantly alleviated itch and alloknesis in various mouse models of pruritus and AD and decreased the density of intraepidermal nerve fibers. Propionate reduced the responsiveness of dorsal root ganglion neurons to pruritogens in vitro, attenuated the hyper-excitability in sensory neurons in MC903-induced AD model, and inhibited capsaicin-evoked hTRPV1 currents (IC50 = 20.08 ± 1.11 μM) via interacting with the vanilloid binding site. Propionate also decreased the secretion of calcitonin gene-related peptide by nerves in MC903-induced AD mouse model, which further attenuated itch and skin inflammation. CONCLUSION Our study revealed a protective effect of propionate against persistent itch through direct modulation of sensory TRP channels and neuropeptide production in neurons. Regulation of itch via the skin microbiome might be a novel strategy for the treatment of AD.
Collapse
Affiliation(s)
- Yao Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Zhuoqiong Qiu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Chaoying Gu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Su Yu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Shangshang Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Changlin Li
- Guangdong Institute of Intelligence Science and Technology, Zhuhai, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for skin diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wei Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| |
Collapse
|
6
|
Zhang X, Shao J, Wang C, Liu C, Hao H, Li X, An Y, He J, Zhao W, Zhao Y, Kong Y, Jia Z, Wan S, Yuan Y, Zhang H, Zhang H, Du X. TMC7 functions as a suppressor of Piezo2 in primary sensory neurons blunting peripheral mechanotransduction. Cell Rep 2024; 43:114014. [PMID: 38568807 DOI: 10.1016/j.celrep.2024.114014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024] Open
Abstract
The transmembrane channel-like (TMC) protein family comprises eight members, with TMC1 and TMC2 being extensively studied. This study demonstrates substantial co-expression of TMC7 with the mechanosensitive channel Piezo2 in somatosensory neurons. Genetic deletion of TMC7 in primary sensory ganglia neurons in vivo enhances sensitivity in both physiological and pathological mechanosensory transduction. This deletion leads to an increase in proportion of rapidly adapting (RA) currents conducted by Piezo2 in dorsal root ganglion (DRG) neurons and accelerates RA deactivation kinetics. In HEK293 cells expressing both proteins, TMC7 significantly suppresses the current amplitudes of co-expressed Piezo2. Our findings reveal that TMC7 and Piezo2 exhibit physical interactions, and both proteins also physically interact with cytoskeletal β-actin. We hypothesize that TMC7 functions as an inhibitory modulator of Piezo2 in DRG neurons, either through direct inhibition or by disrupting the transmission of mechanical forces from the cytoskeleton to the channel.
Collapse
Affiliation(s)
- Xiaoxue Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jichen Shao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Caixue Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China; The Forth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chao Liu
- Department of Animal Care, The Key Laboratory of Experimental Animal, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Han Hao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinmeng Li
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yating An
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinsha He
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weixin Zhao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiwen Zhao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Youzhen Kong
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhanfeng Jia
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shaopo Wan
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, Hebei, China
| | - Yi Yuan
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, Hebei, China
| | - Huiran Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Zhu H, Tao Y, Wang S, Zhu X, Lin K, Zheng N, Chen LM, Xu F, Wu R. fMRI, LFP, and anatomical evidence for hierarchical nociceptive routing pathway between somatosensory and insular cortices. Neuroimage 2024; 289:120549. [PMID: 38382864 DOI: 10.1016/j.neuroimage.2024.120549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
The directional organization of multiple nociceptive regions, particularly within obscure operculoinsular areas, underlying multidimensional pain processing remains elusive. This study aims to establish the fundamental organization between somatosensory and insular cortices in routing nociceptive information. By employing an integrated multimodal approach of high-field fMRI, intracranial electrophysiology, and transsynaptic viral tracing in rats, we observed a hierarchically organized connection of S1/S2 → posterior insula → anterior insula in routing nociceptive information. The directional nociceptive pathway determined by early fMRI responses was consistent with that examined by early evoked LFP, intrinsic effective connectivity, and anatomical projection, suggesting fMRI could provide a valuable facility to discern directional neural circuits in animals and humans non-invasively. Moreover, our knowledge of the nociceptive hierarchical organization of somatosensory and insular cortices and the interface role of the posterior insula may have implications for the development of targeted pain therapies.
Collapse
Affiliation(s)
- Hongyan Zhu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yan Tao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Siqi Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xutao Zhu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kunzhang Lin
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ning Zheng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Li Min Chen
- Vanderbilt University Institute of Imaging Science and Department of Psychology, Vanderbilt University, Nashville, TN 37232, USA.
| | - Fuqiang Xu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Ruiqi Wu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 200031, China.
| |
Collapse
|
8
|
Xie MX, Lai RC, Xiao YB, Zhang X, Cao XY, Tian XY, Chen AN, Chen ZY, Cao Y, Li X, Zhang XL. Endophilin A2 controls touch and mechanical allodynia via kinesin-mediated Piezo2 trafficking. Mil Med Res 2024; 11:17. [PMID: 38475827 DOI: 10.1186/s40779-024-00520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 02/02/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Tactile and mechanical pain are crucial to our interaction with the environment, yet the underpinning molecular mechanism is still elusive. Endophilin A2 (EndoA2) is an evolutionarily conserved protein that is documented in the endocytosis pathway. However, the role of EndoA2 in the regulation of mechanical sensitivity and its underlying mechanisms are currently unclear. METHODS Male and female C57BL/6 mice (8-12 weeks) and male cynomolgus monkeys (7-10 years old) were used in our experiments. Nerve injury-, inflammatory-, and chemotherapy-induced pathological pain models were established for this study. Behavioral tests of touch, mechanical pain, heat pain, and cold pain were performed in mice and nonhuman primates. Western blotting, immunostaining, co-immunoprecipitation, proximity ligation and patch-clamp recordings were performed to gain insight into the mechanisms. RESULTS The results showed that EndoA2 was primarily distributed in neurofilament-200-positive (NF200+) medium-to-large diameter dorsal root ganglion (DRG) neurons of mice and humans. Loss of EndoA2 in mouse NF200+ DRG neurons selectively impaired the tactile and mechanical allodynia. Furthermore, EndoA2 interacted with the mechanically sensitive ion channel Piezo2 and promoted the membrane trafficking of Piezo2 in DRG neurons. Moreover, as an adaptor protein, EndoA2 also bound to kinesin family member 5B (KIF5B), which was involved in the EndoA2-mediated membrane trafficking process of Piezo2. Loss of EndoA2 in mouse DRG neurons damaged Piezo2-mediated rapidly adapting mechanically activated currents, and re-expression of EndoA2 rescued the MA currents. In addition, interference with EndoA2 also suppressed touch sensitivity and mechanical hypersensitivity in nonhuman primates. CONCLUSIONS Our data reveal that the KIF5B/EndoA2/Piezo2 complex is essential for Piezo2 trafficking and for sustaining transmission of touch and mechanical hypersensitivity signals. EndoA2 regulates touch and mechanical allodynia via kinesin-mediated Piezo2 trafficking in sensory neurons. Our findings identify a potential new target for the treatment of mechanical pain.
Collapse
Affiliation(s)
- Man-Xiu Xie
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Ren-Chun Lai
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Yi-Bin Xiao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xi Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xian-Ying Cao
- Engineering Technology Research Center for Elderly Health Management in Hainan Province, Haikou, 571137, China
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - Xiao-Yu Tian
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - An-Nan Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zi-Yi Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yan Cao
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - Xiao Li
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - Xiao-Long Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
9
|
Wu D, Zhong S, Du H, Han S, Wei X, Gong Q. MiR-184-5p represses neuropathic pain by regulating CCL1/CCR8 signaling interplay in the spinal cord in diabetic mice. Neurol Res 2024; 46:54-64. [PMID: 37842802 DOI: 10.1080/01616412.2023.2257454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/18/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Diabetic neuropathic pain (DNP) is a serious complication for diabetic patients involving nervous system. MicroRNAs (miRNAs) are small-noncoding RNAs which are dysregulated in neuropathic pain, and might be critical molecules for pain treatment. Our previous study has shown miR-184-5p was significantly downregulated in DNP. Therefore, the mechanism of miR-184-5p in DNP was investigated in this study. METHODS A DNP model was established through streptozotocin (STZ). The pharmacological tools were injected intrathecally, and pain behavior was evaluated by paw withdrawal mechanical thresholds (PWMTs). Bioinformatics analysis, Dual-luciferase reporter assay and fluorescence-in-situ-hybridization (FISH) were used to seek and confirm the potential target genes of miR-184-5p. The expression of relative genes and proteins was analyzed by quantitative reverse transcriptase real-time PCR (qPCR) and western blotting. RESULTS MiR-184-5p expression was down-regulated in spinal dorsal on days 7 and 14 after STZ, while intrathecal administration of miR-184-5p agomir attenuates neuropathic pain induced by DNP and intrathecal miR-184-5p antagomir induces pain behaviors in naïve mice. Chemokine CC motif ligand 1 (CCL1) was found to be a potential target of miR-184-5p and the protein expression of CCL1 and the mRNA expression of CCR8 were up-regulated in spinal dorsal on days 7 and 14 after STZ. The luciferase reporter assay and FISH demonstrated that CCL1 is a direct target of miR-184-5p. MiR-184-5p overexpression attenuated the expression of CCL1/CCR8 in DNP; intrathecal miR-184-5p antagomir increased the expression of CCL1/CCR8 in spinal dorsal of naïve mice. CONCLUSION This research illustrates that miR-184-5p alleviates DNP through the inhibition of CCL1/CCR8 signaling expression.
Collapse
Affiliation(s)
- Danlei Wu
- Department of Pain Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuotao Zhong
- Department of Pain Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huiying Du
- Department of Anesthesiology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Shuang Han
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, China
| | - Xuhong Wei
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, China
| | - Qingjuan Gong
- Department of Pain Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Dvorak NM, Di Re J, Vasquez TES, Marosi M, Shah P, Contreras YMM, Bernabucci M, Singh AK, Stallone J, Green TA, Laezza F. Fibroblast growth factor 13-mediated regulation of medium spiny neuron excitability and cocaine self-administration. Front Neurosci 2023; 17:1294567. [PMID: 38099204 PMCID: PMC10720079 DOI: 10.3389/fnins.2023.1294567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/31/2023] [Indexed: 12/17/2023] Open
Abstract
Cocaine use disorder (CUD) is a prevalent neuropsychiatric disorder with few existing treatments. Thus, there is an unmet need for the identification of new pharmacological targets for CUD. Previous studies using environmental enrichment versus isolation paradigms have found that the latter induces increased cocaine self-administration with correlative increases in the excitability of medium spiny neurons (MSN) of the nucleus accumbens shell (NAcSh). Expanding upon these findings, we sought in the present investigation to elucidate molecular determinants of these phenomena. To that end, we first employed a secondary transcriptomic analysis and found that cocaine self-administration differentially regulates mRNA for fibroblast growth factor 13 (FGF13), which codes for a prominent auxiliary protein of the voltage-gated Na+ (Nav) channel, in the NAcSh of environmentally enriched rats (i.e., resilient behavioral phenotype) compared to environmentally isolated rats (susceptible phenotype). Based upon this finding, we used in vivo genetic silencing to study the causal functional and behavioral consequences of knocking down FGF13 in the NAcSh. Functional studies revealed that knockdown of FGF13 in the NAcSh augmented excitability of MSNs by increasing the activity of Nav channels. These electrophysiological changes were concomitant with a decrease in cocaine demand elasticity (i.e., susceptible phenotype). Taken together, these data support FGF13 as being protective against cocaine self-administration, which positions it well as a pharmacological target for CUD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Thomas A. Green
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
11
|
Tuttle AM, Miller LN, Royer LJ, Wen H, Kelly JJ, Calistri NL, Heiser LM, Nechiporuk AV. Single-cell analysis of Rohon-Beard neurons implicates Fgf signaling in axon maintenance and cell survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554953. [PMID: 37693470 PMCID: PMC10491107 DOI: 10.1101/2023.08.26.554953] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Peripheral sensory neurons are a critical part of the nervous system that transmit a multitude of sensory stimuli to the central nervous system. During larval and juvenile stages in zebrafish, this function is mediated by Rohon-Beard somatosensory neurons (RBs). RBs are optically accessible and amenable to experimental manipulation, making them a powerful system for mechanistic investigation of sensory neurons. Previous studies provided evidence that RBs fall into multiple subclasses; however, the number and molecular make up of these potential RB subtypes have not been well defined. Using a single-cell RNA sequencing (scRNA-seq) approach, we demonstrate that larval RBs in zebrafish fall into three, largely non-overlapping classes of neurons. We also show that RBs are molecularly distinct from trigeminal neurons in zebrafish. Cross-species transcriptional analysis indicates that one RB subclass is similar to a mammalian group of A-fiber sensory neurons. Another RB subclass is predicted to sense multiple modalities, including mechanical stimulation and chemical irritants. We leveraged our scRNA-seq data to determine that the fibroblast growth factor (Fgf) pathway is active in RBs. Pharmacological and genetic inhibition of this pathway led to defects in axon maintenance and RB cell death. Moreover, this can be phenocopied by treatment with dovitinib, an FDA-approved Fgf inhibitor with a common side effect of peripheral neuropathy. Importantly, dovitinib-mediated axon loss can be suppressed by loss of Sarm1, a positive regulator of neuronal cell death and axonal injury. This offers a molecular target for future clinical intervention to fight neurotoxic effects of this drug.
Collapse
|
12
|
Cheng H, Miao P, Wang Y, Guo Y, Gao L, Lou Y, Yang F, Liang M, Feng J. A Heterozygous Variant of FGF13 Caused X-Linked Developmental and Epileptic Encephalopathy 90 in a Chinese Family. Cytogenet Genome Res 2023; 163:36-41. [PMID: 37536293 DOI: 10.1159/000531932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/25/2023] [Indexed: 08/05/2023] Open
Abstract
Developmental and epileptic encephalopathy (DEE) refers to a group of severe epilepsy encephalopathy and development disorders, and its typical clinical features include seizures, drug resistance, and developmental delay or regression. To date, limited studies have reported DEEs driven by FGF13. Here, we reported a girl with developmental and epileptic encephalopathy 90 caused by variant of FGF13. Her electroencephalogram (EEG) showed discontinuous hypsarrhythmia, and a heterozygous nonsynonymous variant in FGF13 [NM_004114.4: c.5C>G, p.(Ala2Gly)] was identified from the proband. The variant was not reported in public databases such as gnomAD and Exome Aggregation Consortium (ExAC), and was predicted to be damaging to proteins and classified as likely pathogenic according to the ACMG guidelines. The seizure was finally controlled by a combination of ACTH + zonisamide (10 mg/kg.d) + levetiracetam (52 mg/kg.d) + clonazepam (0.7 mg/kg.d).
Collapse
Affiliation(s)
- Haiying Cheng
- Department of Paediatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pu Miao
- Department of Paediatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ye Wang
- Department of Paediatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yufan Guo
- Department of Paediatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liuyan Gao
- Department of Paediatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuting Lou
- Department of Paediatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | - Jianhua Feng
- Department of Paediatrics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Marra C, Hartke TV, Ringkamp M, Goldfarb M. Enhanced sodium channel inactivation by temperature and FHF2 deficiency blocks heat nociception. Pain 2023; 164:1321-1331. [PMID: 36607284 PMCID: PMC10166761 DOI: 10.1097/j.pain.0000000000002822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 10/13/2022] [Accepted: 10/31/2022] [Indexed: 01/07/2023]
Abstract
ABSTRACT Transient voltage-gated sodium currents are essential for the initiation and conduction of action potentials in neurons and cardiomyocytes. The amplitude and duration of sodium currents are tuned by intracellular fibroblast growth factor homologous factors (FHFs/iFGFs) that associate with the cytoplasmic tails of voltage-gated sodium channels (Na v s), and genetic ablation of Fhf genes disturbs neurological and cardiac functions. Among reported phenotypes, Fhf2null mice undergo lethal hyperthermia-induced cardiac conduction block attributable to the combined effects of FHF2 deficiency and elevated temperature on the cardiac sodium channel (Na v 1.5) inactivation rate. Fhf2null mice also display a lack of heat nociception, while retaining other somatosensory capabilities. Here, we use electrophysiological and computational methods to show that the heat nociception deficit can be explained by the combined effects of elevated temperature and FHF2 deficiency on the fast inactivation gating of Na v 1.7 and tetrodotoxin-resistant sodium channels expressed in dorsal root ganglion C fibers. Hence, neurological and cardiac heat-associated deficits in Fhf2null mice derive from shared impacts of FHF deficiency and temperature towards Na v inactivation gating kinetics in distinct tissues.
Collapse
Affiliation(s)
- Christopher Marra
- Department of Biological Sciences, Hunter College of City University, New York, NY, United States
- Program in Biology, Graduate Center of City University, New York, NY, United States
| | - Timothy V. Hartke
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Matthias Ringkamp
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Mitchell Goldfarb
- Department of Biological Sciences, Hunter College of City University, New York, NY, United States
- Program in Biology, Graduate Center of City University, New York, NY, United States
| |
Collapse
|
14
|
Wang B, Jiang B, Li G, Dong F, Luo Z, Cai B, Wei M, Huang J, Wang K, Feng X, Tong F, Wang S, Wang Q, Han Q, Li C, Zhang X, Yang L, Bao L. Somatosensory neurons express specific sets of lincRNAs, and lincRNA CLAP promotes itch sensation in mice. EMBO Rep 2023; 24:e54313. [PMID: 36524339 PMCID: PMC9900349 DOI: 10.15252/embr.202154313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
Somatosensory neurons are highly heterogeneous with distinct types of neural cells responding to specific stimuli. However, the distribution and roles of cell-type-specific long intergenic noncoding RNAs (lincRNAs) in somatosensory neurons remain largely unexplored. Here, by utilizing droplet-based single-cell RNA-seq (scRNA-seq) and full-length Smart-seq2, we show that lincRNAs, but not coding mRNAs, are enriched in specific types of mouse somatosensory neurons. Profiling of lincRNAs from single neurons located in dorsal root ganglia (DRG) identifies 200 lincRNAs localized in specific types or subtypes of somatosensory neurons. Among them, the conserved cell-type-specific lincRNA CLAP associates with pruritus and is abundantly expressed in somatostatin (SST)-positive neurons. CLAP knockdown reduces histamine-induced Ca2+ influx in cultured SST-positive neurons and in vivo reduces histamine-induced scratching in mice. In vivo knockdown of CLAP also decreases the expression of neuron-type-specific and itch-related genes in somatosensory neurons, and this partially depends on the RNA binding protein MSI2. Our data reveal a cell-type-specific landscape of lincRNAs and a function for CLAP in somatosensory neurons in sensory transmission.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
| | - Bowen Jiang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Guo‐Wei Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Fei Dong
- Institute of Neuroscience and State Key Laboratory of NeuroscienceCAS Center for Excellence in Brain Science and Intelligence TechnologyShanghaiChina
| | - Zheng Luo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Bing Cai
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
| | - Manyi Wei
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Jiansong Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Kaikai Wang
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
| | - Xin Feng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Fang Tong
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Sashuang Wang
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain MedicineHuazhong University of Science and Technology Union Shenzhen HospitalShenzhenChina
| | - Qiong Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Qingjian Han
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Changlin Li
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
- Research Unit of Pain, Chinese Academy of Medical Sciences, Shanghai Advanced Research InstituteChinese Academy of SciencesShanghaiChina
| | - Xu Zhang
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
- Institute of Neuroscience and State Key Laboratory of NeuroscienceCAS Center for Excellence in Brain Science and Intelligence TechnologyShanghaiChina
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
- Research Unit of Pain, Chinese Academy of Medical Sciences, Shanghai Advanced Research InstituteChinese Academy of SciencesShanghaiChina
| | - Li Yang
- Center for Molecular Medicine, Children's Hospital, Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Lan Bao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
| |
Collapse
|
15
|
Zhang FM, Wang B, Hu H, Zhang YY, Chen HH, Jiang ZJ, Zeng MX, Liu XJ. Transcriptional profiles of TGF-β superfamily members in the lumbar DRGs and the effects of activins A and C on inflammatory pain in rats. J Physiol Biochem 2023:10.1007/s13105-022-00943-z. [PMID: 36696051 DOI: 10.1007/s13105-022-00943-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/28/2022] [Indexed: 01/26/2023]
Abstract
Signaling by the transforming growth factor (TGF)-β superfamily is necessary for proper neural development and is involved in pain processing under both physiological and pathological conditions. Sensory neurons that reside in the dorsal root ganglia (DRGs) initially begin to perceive noxious signaling from their innervating peripheral target tissues and further convey pain signaling to the central nervous system. However, the transcriptional profile of the TGF-β superfamily members in DRGs during chronic inflammatory pain remains elusive. We developed a custom microarray to screen for transcriptional changes in members of the TGF-β superfamily in lumbar DRGs of rats with chronic inflammatory pain and found that the transcription of the TGF-β superfamily members tends to be downregulated. Among them, signaling of the activin/inhibin and bone morphogenetic protein/growth and differentiation factor (BMP/GDF) families dramatically decreased. In addition, peripherally pre-local administration of activins A and C worsened formalin-induced acute inflammatory pain, whereas activin C, but not activin A, improved formalin-induced persistent inflammatory pain by inhibiting the activation of astrocytes. This is the first report of the TGF-β superfamily transcriptional profiles in lumbar DRGs under chronic inflammatory pain conditions, in which transcriptional changes in cytokines or pathway components were found to contribute to, or be involved in, inflammatory pain processing. Our data will provide more targets for pain research.
Collapse
Affiliation(s)
- Feng-Ming Zhang
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Bing Wang
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China
| | - Han Hu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, No. 1 Beigou Xiangshan, Beijing, 100093, China
| | - Ying-Ying Zhang
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China
| | - Hao-Hao Chen
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Zuo-Jie Jiang
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Mei-Xing Zeng
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China
| | - Xing-Jun Liu
- School of Pharmacy, Nantong University, Jiangsu Province, 226001, Nantong, China.
- Pain and Related Disease Research Lab, Shantou University Medical College, Shantou, 515041, Guangdong Province, China.
| |
Collapse
|
16
|
Shen KF, Yue J, Wu ZF, Wu KF, Zhu G, Yang XL, Wang ZK, Wang J, Liu SY, Yang H, Zhang CQ. Fibroblast growth factor 13 is involved in the pathogenesis of temporal lobe epilepsy. Cereb Cortex 2022; 32:5259-5272. [PMID: 35195262 DOI: 10.1093/cercor/bhac012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/08/2022] [Accepted: 01/09/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Temporal lobe epilepsy (TLE) is the most common drug-resistant epilepsy in adults, with pathological mechanisms remaining to be fully elucidated. Fibroblast Growth Factor 13 (FGF13) encodes an intracellular protein involved in microtubule stabilization and regulation of voltage-gated sodium channels (VGSCs) function. FGF13 mutation has been identified in patients with inherent seizure, suggesting a potential association between FGF13 and the etiology of TLE. Here, we set to explore the pathological role of FGF13 in the etiology of TLE. RESULTS We found that the expression of FGF13 was increased in the cortical lesions and CA1 region of sclerotic hippocampus and correlated with the seizure frequency in TLE patients. Also, Fgf13 expression was increased in the hippocampus of chronic TLE mice generated by kainic acid (KA) injection. Furthermore, Fgf13 knockdown or overexpression was respectively found to attenuate or potentiate the effects of KA on axonal length, somatic area and the VGSCs-mediated current in the hippocampal neurons. CONCLUSIONS Taken together, these findings suggest that FGF13 is involved in the pathogenesis of TLE by modulating microtubule activity and neuronal excitability.
Collapse
Affiliation(s)
- Kai-Feng Shen
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Jiong Yue
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Zhi-Feng Wu
- Department of Pedatrics, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Ke-Fu Wu
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Gang Zhu
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Xiao-Lin Yang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Zhong-Ke Wang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Jing Wang
- Department of Pain Management, Henan Provincial People's hospital, 7 Weiwu Road, Jinshui District, Zhengzhou 450008, China.,Zhongshan Medical School, Sun Yat-sen University, 74 Zhongshan Second Road, Yuexiu District, Guangzhou 510080, China
| | - Shi-Yong Liu
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Chun-Qing Zhang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| |
Collapse
|
17
|
Abstract
Sleep is a fundamental, evolutionarily conserved, plastic behavior that is regulated by circadian and homeostatic mechanisms as well as genetic factors and environmental factors, such as light, humidity, and temperature. Among environmental cues, temperature plays an important role in the regulation of sleep. This review presents an overview of thermoreception in animals and the neural circuits that link this process to sleep. Understanding the influence of temperature on sleep can provide insight into basic physiologic processes that are required for survival and guide strategies to manage sleep disorders.
Collapse
|
18
|
Rodocker HI, Bordbar A, Larson MJE, Biltz RG, Wangler L, Fadda P, Godbout JP, Tedeschi A. Breaking Mental Barriers Promotes Recovery After Spinal Cord Injury. Front Mol Neurosci 2022; 15:868563. [PMID: 35875670 PMCID: PMC9301320 DOI: 10.3389/fnmol.2022.868563] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/14/2022] [Indexed: 11/23/2022] Open
Abstract
Functional recovery after spinal cord injury (SCI) often proves difficult as physical and mental barriers bar survivors from enacting their designated rehabilitation programs. We recently demonstrated that adult mice administered gabapentinoids, clinically approved drugs prescribed to mitigate chronic neuropathic pain, recovered upper extremity function following cervical SCI. Given that rehabilitative training enhances neuronal plasticity and promotes motor recovery, we hypothesized that the combination of an aerobic-based rehabilitation regimen like treadmill training with gabapentin (GBP) administration will maximize recovery in SCI mice by strengthening synaptic connections along the sensorimotor axis. Whereas mice administered GBP recovered forelimb functions over the course of weeks and months following SCI, no additive forelimb recovery as the result of voluntary treadmill training was noted in these mice. To our surprise, we also failed to find an additive effect in mice administered vehicle. As motivation is crucial in rehabilitation interventions, we scored active engagement toward the rehabilitation protocol and found that mice administered GBP were consistently participating in the rehabilitation program. In contrast, mice administered vehicle exhibited a steep decline in participation, especially at chronic time points. Whereas neuroinflammatory gene expression profiles were comparable between experimental conditions, we discovered that mice administered GBP had increased hippocampal neurogenesis and exhibited less anxiety-like behavior after SCI. We also found that an external, social motivator effectively rescues participation in mice administered vehicle and promotes forelimb recovery after chronic SCI. Thus, not only does a clinically relevant treatment strategy preclude the deterioration of mental health after chronic SCI, but group intervention strategies may prove to be physically and emotionally beneficial for SCI individuals.
Collapse
Affiliation(s)
- Haven I. Rodocker
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Arman Bordbar
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Molly J. E. Larson
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Rebecca G. Biltz
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Lynde Wangler
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Paolo Fadda
- Department of Cancer Biology, The Ohio State University, Columbus, OH, United States
| | - Jonathan P. Godbout
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
19
|
Zhang Y, Miao Q, Shi S, Hao H, Li X, Pu Z, Yang Y, An H, Zhang W, Kong Y, Pang X, Gu C, Gamper N, Wu Y, Zhang H, Du X. Protein disulfide isomerase modulation of TRPV1 controls heat hyperalgesia in chronic pain. Cell Rep 2022; 39:110625. [PMID: 35385753 DOI: 10.1016/j.celrep.2022.110625] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
Protein disulfide isomerase (PDI) plays a key role in maintaining cellular homeostasis by mediating protein folding via catalyzing disulfide bond formation, breakage, and rearrangement in the endoplasmic reticulum. Increasing evidence suggests that PDI can be a potential treatment target for several diseases. However, the function of PDI in the peripheral sensory nervous system is unclear. Here we report the expression and secretion of PDI from primary sensory neurons is upregulated in inflammatory and neuropathic pain models. Deletion of PDI in nociceptive DRG neurons results in a reduction in inflammatory and neuropathic heat hyperalgesia. We demonstrate that secreted PDI activates TRPV1 channels through oxidative modification of extracellular cysteines of the channel, indicating that PDI acts as an unconventional positive modulator of TRPV1. These findings suggest that PDI in primary sensory neurons plays an important role in development of heat hyperalgesia and can be a potential therapeutic target for chronic pain.
Collapse
Affiliation(s)
- Yongxue Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Pharmacy, The First Hospital of Handan, Handan, Hebei, China
| | - Qi Miao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sai Shi
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, Hebei, China
| | - Han Hao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinmeng Li
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zeyao Pu
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yakun Yang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, Hebei, China
| | - Wei Zhang
- Department of Spinal Surgery of the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Youzhen Kong
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xu Pang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cunyang Gu
- Department of Pathology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China; Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, UK
| | - Yi Wu
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China.
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
20
|
Dvorak NM, Tapia CM, Baumgartner TJ, Singh J, Laezza F, Singh AK. Pharmacological Inhibition of Wee1 Kinase Selectively Modulates the Voltage-Gated Na + Channel 1.2 Macromolecular Complex. Cells 2021; 10:3103. [PMID: 34831326 PMCID: PMC8619224 DOI: 10.3390/cells10113103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Voltage-gated Na+ (Nav) channels are a primary molecular determinant of the action potential (AP). Despite the canonical role of the pore-forming α subunit in conferring this function, protein-protein interactions (PPI) between the Nav channel α subunit and its auxiliary proteins are necessary to reconstitute the full physiological activity of the channel and to fine-tune neuronal excitability. In the brain, the Nav channel isoforms 1.2 (Nav1.2) and 1.6 (Nav1.6) are enriched, and their activities are differentially regulated by the Nav channel auxiliary protein fibroblast growth factor 14 (FGF14). Despite the known regulation of neuronal Nav channel activity by FGF14, less is known about cellular signaling molecules that might modulate these regulatory effects of FGF14. To that end, and building upon our previous investigations suggesting that neuronal Nav channel activity is regulated by a kinase network involving GSK3, AKT, and Wee1, we interrogate in our current investigation how pharmacological inhibition of Wee1 kinase, a serine/threonine and tyrosine kinase that is a crucial component of the G2-M cell cycle checkpoint, affects the Nav1.2 and Nav1.6 channel macromolecular complexes. Our results show that the highly selective inhibitor of Wee1 kinase, called Wee1 inhibitor II, modulates FGF14:Nav1.2 complex assembly, but does not significantly affect FGF14:Nav1.6 complex assembly. These results are functionally recapitulated, as Wee1 inhibitor II entirely alters FGF14-mediated regulation of the Nav1.2 channel, but displays no effects on the Nav1.6 channel. At the molecular level, these effects of Wee1 inhibitor II on FGF14:Nav1.2 complex assembly and FGF14-mediated regulation of Nav1.2-mediated Na+ currents are shown to be dependent upon the presence of Y158 of FGF14, a residue known to be a prominent site for phosphorylation-mediated regulation of the protein. Overall, our data suggest that pharmacological inhibition of Wee1 confers selective modulatory effects on Nav1.2 channel activity, which has important implications for unraveling cellular signaling pathways that fine-tune neuronal excitability.
Collapse
Affiliation(s)
| | | | | | | | | | - Aditya K. Singh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 75901, USA; (N.M.D.); (C.M.T.); (T.J.B.); (J.S.); (F.L.)
| |
Collapse
|
21
|
Yu H, Wang H, Qie A, Wang J, Liu Y, Gu G, Yang J, Zhang H, Pan W, Tian Z, Wang C. FGF13 enhances resistance to platinum drugs by regulating hCTR1 and ATP7A via a microtubule-stabilizing effect. Cancer Sci 2021; 112:4655-4668. [PMID: 34533854 PMCID: PMC8586689 DOI: 10.1111/cas.15137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/25/2022] Open
Abstract
Platinum‐based regimens are the most widely used chemotherapy regimens, but cancer cells often develop resistance, which impedes therapy outcome for patients. Previous studies have shown that fibroblast growth factor 13 (FGF13) is associated with resistance to platinum drugs in HeLa cells. However, the mechanism and universality of this effect have not been clarified. Here, we found that FGF13 was associated with poor platinum‐based chemotherapy outcomes in a variety of cancers, such as lung, endometrial, and cervical cancers, through bioinformatics analysis. We then found that FGF13 simultaneously regulates the expression and distribution of hCTR1 and ATP7A in cancer cells, causes reduced platinum influx, and promotes platinum sequestration and efflux upon cisplatin exposure. We subsequently observed that FGF13‐mediated platinum resistance requires the microtubule‐stabilizing effect of FGF13. Only overexpression of FGF13 with the ‐SMIYRQQQ‐ tubulin‐binding domain could induce the platinum resistance effect. This phenomenon was also observed in SK‐MES‐1 cells, KLE cells, and 5637 cells. Our research reveals the mechanism of FGF13‐induced platinum drug resistance and suggests that FGF13 can be a sensibilization target and prognostic biomarker for chemotherapy.
Collapse
Affiliation(s)
- Hang Yu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China.,College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Handong Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Anran Qie
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaqi Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guoqiang Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Yang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Hanqiu Zhang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Wensen Pan
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
22
|
Pan X, Zhao J, Zhou Z, Chen J, Yang Z, Wu Y, Bai M, Jiao Y, Yang Y, Hu X, Cheng T, Lu Q, Wang B, Li CL, Lu YJ, Diao L, Zhong YQ, Pan J, Zhu J, Xiao HS, Qiu ZL, Li J, Wang Z, Hui J, Bao L, Zhang X. 5'-UTR SNP of FGF13 causes translational defect and intellectual disability. eLife 2021; 10:63021. [PMID: 34184986 PMCID: PMC8241442 DOI: 10.7554/elife.63021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
The congenital intellectual disability (ID)-causing gene mutations remain largely unclear, although many genetic variations might relate to ID. We screened gene mutations in Chinese Han children suffering from severe ID and found a single-nucleotide polymorphism (SNP) in the 5′-untranslated region (5′-UTR) of fibroblast growth factor 13 (FGF13) mRNA (NM_001139500.1:c.-32c>G) shared by three male children. In both HEK293 cells and patient-derived induced pluripotent stem cells, this SNP reduced the translation of FGF13, which stabilizes microtubules in developing neurons. Mice carrying the homologous point mutation in 5′-UTR of Fgf13 showed delayed neuronal migration during cortical development, and weakened learning and memory. Furthermore, this SNP reduced the interaction between FGF13 5′-UTR and polypyrimidine-tract-binding protein 2 (PTBP2), which was required for FGF13 translation in cortical neurons. Thus, this 5′-UTR SNP of FGF13 interferes with the translational process of FGF13 and causes deficits in brain development and cognitive functions.
Collapse
Affiliation(s)
- Xingyu Pan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Shanghai Brain-Intelligence Project Center, Shanghai, China
| | - Jingrong Zhao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhiying Zhou
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Jijun Chen
- Shanghai Brain-Intelligence Project Center, Shanghai, China
| | - Zhenxing Yang
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Yuxuan Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Meizhu Bai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yang Jiao
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Yun Yang
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xuye Hu
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Tianling Cheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qianyun Lu
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Bin Wang
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Chang-Lin Li
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Ying-Jin Lu
- Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Lei Diao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yan-Qing Zhong
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Pan
- Shanghai Brain-Intelligence Project Center, Shanghai, China
| | - Jianmin Zhu
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Hua-Sheng Xiao
- Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China
| | - Zi-Long Qiu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Zefeng Wang
- CAS-MPG Partner Institute for Computational Biology, Chinese Academy of Sciences, Shanghai, China
| | - Jingyi Hui
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Lan Bao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Xu Zhang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Shanghai Brain-Intelligence Project Center, Shanghai, China.,Shanghai Clinical Center, Chinese Academy of Sciences/Xu-Hui Central Hospital, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China.,Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
23
|
Dvorak NM, Wadsworth PA, Wang P, Zhou J, Laezza F. Development of Allosteric Modulators of Voltage-Gated Na + Channels: A Novel Approach for an Old Target. Curr Top Med Chem 2021; 21:841-848. [PMID: 34036922 DOI: 10.2174/1568026621666210525105359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022]
Abstract
Given their primacy in governing the action potential (AP) of excitable cells, voltage-gated Na+ (Nav) channels are important pharmacological targets of therapeutics for a diverse array of clinical indications. Despite historically being a traditional drug target, therapeutics targeting Nav channels lack isoform selectivity, giving rise to off-target side effects. To develop isoform-selective modulators of Nav channels with improved target-specificity, the identification and pharmacological targeting of allosteric sites that display structural divergence among Nav channel isoforms represents an attractive approach. Despite the high homology among Nav channel α subunit isoforms (Nav1.1-Nav1.9), there is considerable amino acid sequence divergence among their constituent C-terminal domains (CTD), which enables structurally and functionally specific protein: protein interactions (PPI) with auxiliary proteins. Although pharmacological targeting of such PPI interfaces between the CTDs of Nav channels and auxiliary proteins represents an innovate approach for developing isoform-selective modulators of Nav channels, appreciable modulation of PPIs using small molecules has conventionally been difficult to achieve. After briefly discussing the challenges of modulating PPIs using small molecules, this current frontier review that follows subsequently expounds on approaches for circumventing such difficulties in the context of developing small molecule modulators of PPIs between transmembrane ion channels and their auxiliary proteins. In addition to broadly discussing such approaches, the implementation of such approaches is specifically discussed in the context of developing small molecule modulators between the CTD of Nav channels and auxiliary proteins. Developing allosteric modulators of ion channels by targeting their PPI interfaces with auxiliary proteins represents an innovative and promising strategy in ion channel drug discovery that could expand the "druggable genome" and usher in first-in-class PPI-targeting therapeutics for a multitude of channelopathies.
Collapse
Affiliation(s)
- Nolan M Dvorak
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Paul A Wadsworth
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Pingyuan Wang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, 77555, United States
| |
Collapse
|
24
|
Foxo1 selectively regulates static mechanical pain by interacting with Nav1.7. Pain 2021; 162:490-502. [PMID: 32868747 DOI: 10.1097/j.pain.0000000000002055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
ABSTRACT Mechanical allodynia is a debilitating condition for millions of patients with chronic pain. Mechanical allodynia can manifest in distinct forms, including brush-evoked dynamic and filament-evoked static allodynia. In the nervous system, the forkhead protein Foxo1 plays a critical role in neuronal structures and functions. However, the role of Foxo1 in the somatosensory signal remains unclear. Here, we found that Foxo1 selectively regulated static mechanical pain. Foxo1 knockdown decreased sensitivity to static mechanical stimuli in normal rats and attenuated static mechanical allodynia in rat models for neuropathic, inflammatory, and chemotherapy pain. Conversely, Foxo1 overexpression selectively enhanced sensitivity to static mechanical stimuli and provoked static mechanical allodynia. Furthermore, Foxo1 interacted with voltage-gated sodium Nav1.7 channels and increased the Nav1.7 current density by accelerating activation rather than by changing the expression of Nav1.7 in dorsal root ganglia neurons. In addition, the serum level of Foxo1 was found to be increased in chronic pain patients and to be positively correlated with the severity of chronic pain. Altogether, our findings suggest that serum Foxo1 level could be used as a biological marker for prediction and diagnosis of chronic pain. Moreover, selective blockade of Foxo1/Nav1.7 interaction may offer a new therapeutic approach in patients with mechanical pain.
Collapse
|
25
|
Wang K, Wang S, Chen Y, Wu D, Hu X, Lu Y, Wang L, Bao L, Li C, Zhang X. Single-cell transcriptomic analysis of somatosensory neurons uncovers temporal development of neuropathic pain. Cell Res 2021; 31:904-918. [PMID: 33692491 DOI: 10.1038/s41422-021-00479-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 01/06/2021] [Indexed: 01/22/2023] Open
Abstract
Peripheral nerve injury could lead to chronic neuropathic pain. Understanding transcriptional changes induced by nerve injury could provide fundamental insights into the complex pathogenesis of neuropathic pain. Gene expression profiles of dorsal root ganglia (DRG) in neuropathic pain condition have been studied. However, little is known about transcriptomic changes in individual DRG neurons after peripheral nerve injury. Here we performed single-cell RNA sequencing on dissociated mouse DRG cells after spared nerve injury (SNI). In addition to DRG neuron types that are found under physiological conditions, we identified three SNI-induced neuronal clusters (SNIICs) characterized by the expression of Atf3/Gfra3/Gal (SNIIC1), Atf3/Mrgprd (SNIIC2) and Atf3/S100b/Gal (SNIIC3). These SNIICs originated from Cldn9+/Gal+, Mrgprd+ and Trappc3l+ DRG neurons, respectively. Interestingly, SNIIC2 switched to SNIIC1 by increasing Gal and reducing Mrgprd expression 2 days after nerve injury. Inferring the gene regulatory networks after nerve injury, we revealed that activated transcription factors Atf3 and Egr1 in SNIICs could enhance Gal expression while activated Cpeb1 in SNIIC2 might suppress Mrgprd expression within 2 days after SNI. Furthermore, we mined the transcriptomic changes in the development of neuropathic pain to identify potential analgesic targets. We revealed that cardiotrophin-like cytokine factor 1, which activates astrocytes in the dorsal horn of spinal cord, was upregulated in SNIIC1 neurons and contributed to SNI-induced mechanical allodynia. Therefore, our results provide a new landscape to understand the dynamic course of neuron type changes and their underlying molecular mechanisms during the development of neuropathic pain.
Collapse
Affiliation(s)
- Kaikai Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sashuang Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Chen
- Research Unit of Pain, Chinese Academy of Medical Sciences, Institute of Brain-Intelligence Science and Technology, Zhangjiang Lab, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 200031, China
| | - Dan Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinyu Hu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingjin Lu
- Research Unit of Pain, Chinese Academy of Medical Sciences, Institute of Brain-Intelligence Science and Technology, Zhangjiang Lab, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 200031, China.,Shanghai Clinical Research Center, Chinese Academy of Sciences, Xuhui Central Hospital, Shanghai, 200031, China
| | - Liping Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lan Bao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Changlin Li
- Research Unit of Pain, Chinese Academy of Medical Sciences, Institute of Brain-Intelligence Science and Technology, Zhangjiang Lab, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 200031, China. .,Shanghai Clinical Research Center, Chinese Academy of Sciences, Xuhui Central Hospital, Shanghai, 200031, China.
| | - Xu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Research Unit of Pain, Chinese Academy of Medical Sciences, Institute of Brain-Intelligence Science and Technology, Zhangjiang Lab, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 200031, China.
| |
Collapse
|
26
|
Xie MX, Cao XY, Zeng WA, Lai RC, Guo L, Wang JC, Xiao YB, Zhang X, Chen D, Liu XG, Zhang XL. ATF4 selectively regulates heat nociception and contributes to kinesin-mediated TRPM3 trafficking. Nat Commun 2021; 12:1401. [PMID: 33658516 PMCID: PMC7930092 DOI: 10.1038/s41467-021-21731-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 02/09/2021] [Indexed: 12/30/2022] Open
Abstract
Effective treatments for patients suffering from heat hypersensitivity are lacking, mostly due to our limited understanding of the pathogenic mechanisms underlying this disorder. In the nervous system, activating transcription factor 4 (ATF4) is involved in the regulation of synaptic plasticity and memory formation. Here, we show that ATF4 plays an important role in heat nociception. Indeed, loss of ATF4 in mouse dorsal root ganglion (DRG) neurons selectively impairs heat sensitivity. Mechanistically, we show that ATF4 interacts with transient receptor potential cation channel subfamily M member-3 (TRPM3) and mediates the membrane trafficking of TRPM3 in DRG neurons in response to heat. Loss of ATF4 also significantly decreases the current and KIF17-mediated trafficking of TRPM3, suggesting that the KIF17/ATF4/TRPM3 complex is required for the neuronal response to heat stimuli. Our findings unveil the non-transcriptional role of ATF4 in the response to heat stimuli in DRG neurons.
Collapse
Affiliation(s)
- Man-Xiu Xie
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Xian-Ying Cao
- College of Food Science and Technology, Hainan University, 58 Renmin Avenue, Haikou, China
- State Key Laboratory of Marine Resources Utilization of South China Sea, 58 Renmin Avenue, Haikou, China
| | - Wei-An Zeng
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Ren-Chun Lai
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Lan Guo
- College of Food Science and Technology, Hainan University, 58 Renmin Avenue, Haikou, China
| | - Jun-Chao Wang
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, China
| | - Yi-Bin Xiao
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, China
| | - Xi Zhang
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, China
| | - Di Chen
- College of Food Science and Technology, Hainan University, 58 Renmin Avenue, Haikou, China
| | - Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, China.
| | - Xiao-Long Zhang
- Medical Research Center of Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Rd. 2, Guangzhou, China.
| |
Collapse
|
27
|
Luo H, Cai B, Pan J, Shi HX, Wang KK, Zhong YQ, Lu YJ, Bao L, Zhang X, Li KC. FXYD6 promotes thermal nociception by regulating TRPV1. Mol Pain 2021. [PMCID: PMC7887684 DOI: 10.1177/1744806921992249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
FXYD6, an unnecessary auxiliary subunit of Na+,K+-ATPase, is expressed in the nervous system. However, its functions remain largely unclear. In the present study, we find that FXYD6 is involved in the thermal nociception. FXYD6 was mainly expressed in small-diameter DRG neurons expressing transient receptor potential channel V1 (TRPV1). In the SNS-Cre/Fxyd6F/F mice, loss of FXYD6 in these sensory neurons impaired the behavioral responses to noxious heat stimulus and intraplantar injection of capsaicin. The capsaicin-induced and TRPV1-mediated currents were decreased in the FXYD6–deficient DRG neurons. Heterologous expression of FXYD6 could increase the TRPV1 capsaicin-sensitive currents in HEK293 cells. Furthermore, we found that the negatively charged PGDEE motif in C-terminal of FXYD6 is required for the FXYD6/TRPV1 interaction and FXYD6-mediated enhancement of TRPV1. Disrupting the FXYD6/TRPV1 interaction with the TAT-PGDEE peptide could elevate the threshold of thermal nociception. Therefore, FXYD6 maintains the thermal nociception via interacting with TRPV1 channel in nociceptors.
Collapse
Affiliation(s)
- Hao Luo
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bing Cai
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Institute of Brain-Intelligence Science and Technology, Zhangjiang Laboratory, Shanghai, China
| | - Jing Pan
- Institute of Brain-Intelligence Science and Technology, Zhangjiang Laboratory, Shanghai, China
| | - Hai-Xiang Shi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kai-Kai Wang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan-Qing Zhong
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying-Jin Lu
- Institute of Brain-Intelligence Science and Technology, Zhangjiang Laboratory, Shanghai, China
| | - Lan Bao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xu Zhang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Institute of Brain-Intelligence Science and Technology, Zhangjiang Laboratory, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research Center, Chinese Academy of Sciences/XuHui Central Hospital, Shanghai, China
| | - Kai-Cheng Li
- Institute of Brain-Intelligence Science and Technology, Zhangjiang Laboratory, Shanghai, China
- Shanghai Clinical Research Center, Chinese Academy of Sciences/XuHui Central Hospital, Shanghai, China
| |
Collapse
|
28
|
Schörnig M, Ju X, Fast L, Ebert S, Weigert A, Kanton S, Schaffer T, Nadif Kasri N, Treutlein B, Peter BM, Hevers W, Taverna E. Comparison of induced neurons reveals slower structural and functional maturation in humans than in apes. eLife 2021; 10:59323. [PMID: 33470930 PMCID: PMC7870144 DOI: 10.7554/elife.59323] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022] Open
Abstract
We generated induced excitatory neurons (iNeurons, iNs) from chimpanzee, bonobo, and human stem cells by expressing the transcription factor neurogenin-2 (NGN2). Single-cell RNA sequencing showed that genes involved in dendrite and synapse development are expressed earlier during iNs maturation in the chimpanzee and bonobo than the human cells. In accordance, during the first 2 weeks of differentiation, chimpanzee and bonobo iNs showed repetitive action potentials and more spontaneous excitatory activity than human iNs, and extended neurites of higher total length. However, the axons of human iNs were slightly longer at 5 weeks of differentiation. The timing of the establishment of neuronal polarity did not differ between the species. Chimpanzee, bonobo, and human neurites eventually reached the same level of structural complexity. Thus, human iNs develop slower than chimpanzee and bonobo iNs, and this difference in timing likely depends on functions downstream of NGN2.
Collapse
Affiliation(s)
- Maria Schörnig
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Xiangchun Ju
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Luise Fast
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Sebastian Ebert
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Anne Weigert
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Sabina Kanton
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Theresa Schaffer
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Nael Nadif Kasri
- Department of Human Genetics and Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboudumc, Nijmegen, Netherlands
| | - Barbara Treutlein
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | | | - Wulf Hevers
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Elena Taverna
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| |
Collapse
|
29
|
Joglekar A, Prjibelski A, Mahfouz A, Collier P, Lin S, Schlusche AK, Marrocco J, Williams SR, Haase B, Hayes A, Chew JG, Weisenfeld NI, Wong MY, Stein AN, Hardwick SA, Hunt T, Wang Q, Dieterich C, Bent Z, Fedrigo O, Sloan SA, Risso D, Jarvis ED, Flicek P, Luo W, Pitt GS, Frankish A, Smit AB, Ross ME, Tilgner HU. A spatially resolved brain region- and cell type-specific isoform atlas of the postnatal mouse brain. Nat Commun 2021; 12:463. [PMID: 33469025 PMCID: PMC7815907 DOI: 10.1038/s41467-020-20343-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/27/2020] [Indexed: 01/19/2023] Open
Abstract
Splicing varies across brain regions, but the single-cell resolution of regional variation is unclear. We present a single-cell investigation of differential isoform expression (DIE) between brain regions using single-cell long-read sequencing in mouse hippocampus and prefrontal cortex in 45 cell types at postnatal day 7 ( www.isoformAtlas.com ). Isoform tests for DIE show better performance than exon tests. We detect hundreds of DIE events traceable to cell types, often corresponding to functionally distinct protein isoforms. Mostly, one cell type is responsible for brain-region specific DIE. However, for fewer genes, multiple cell types influence DIE. Thus, regional identity can, although rarely, override cell-type specificity. Cell types indigenous to one anatomic structure display distinctive DIE, e.g. the choroid plexus epithelium manifests distinct transcription-start-site usage. Spatial transcriptomics and long-read sequencing yield a spatially resolved splicing map. Our methods quantify isoform expression with cell-type and spatial resolution and it contributes to further our understanding of how the brain integrates molecular and cellular complexity.
Collapse
Affiliation(s)
- Anoushka Joglekar
- Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Andrey Prjibelski
- Center for Algorithmic Biotechnology, Institute of Translational Biomedicine, St. Petersburg State University, St Petersburg, Russia
| | - Ahmed Mahfouz
- Department of Human Genetics, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, 2628 XE, The Netherlands
| | - Paul Collier
- Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Susan Lin
- Graduate Program in Neuroscience, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Anna Katharina Schlusche
- Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Jordan Marrocco
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | | | - Bettina Haase
- The Vertebrate Genomes Lab, The Rockefeller University, New York, NY, USA
| | | | | | | | - Man Ying Wong
- Brain and Mind Research Institute and Appel Alzheimer's Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Simon A Hardwick
- Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Toby Hunt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Qi Wang
- Section of Bioinformatics and Systems Cardiology, University Hospital, 96120, Heidelberg, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, University Hospital, 96120, Heidelberg, Germany
| | | | - Olivier Fedrigo
- The Vertebrate Genomes Lab, The Rockefeller University, New York, NY, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Davide Risso
- Department of Statistical Sciences, University of Padova, Padova, Italy
| | - Erich D Jarvis
- The Vertebrate Genomes Lab, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Wenjie Luo
- Brain and Mind Research Institute and Appel Alzheimer's Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Geoffrey S Pitt
- Graduate Program in Neuroscience, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Adam Frankish
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| | - M Elizabeth Ross
- Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA
| | - Hagen U Tilgner
- Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
30
|
Wang Q, Yang J, Wang H, Shan B, Yin C, Yu H, Zhang X, Dong Z, Yu Y, Zhao R, Liu B, Zhang H, Wang C. Fibroblast growth factor 13 stabilizes microtubules to promote Na + channel function in nociceptive DRG neurons and modulates inflammatory pain. J Adv Res 2020; 31:97-111. [PMID: 34194835 PMCID: PMC8240113 DOI: 10.1016/j.jare.2020.12.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/16/2020] [Accepted: 12/14/2020] [Indexed: 11/28/2022] Open
Abstract
Introduction Fibroblast growth factor homologous factors (FHFs), among other fibroblast growth factors, are increasingly found to be important regulators of ion channel functions. Although FHFs have been link to several neuronal diseases and arrhythmia, its role in inflammatory pain still remains unclear. Objectives This study aimed to investigate the role and mechanism of FGF13 in inflammatory pain. Methods Fgf13 conditional knockout mice were generated and CFA-induced chronic inflammatory pain model was established to measure the pain threshold. Immunostaining, western blot and quantitative real-time reverse transcription PCR (qRT-PCR) were performed to detect the expression of FGF13 in CFA-induced inflammatory pain. Whole-cell patch clamp recording was used to record the action potential firing properties and sodium currents of DRG neurons. Results Conditional knockout of Fgf13 in dorsal root ganglion (DRG) neurons (Fgf13-/Y) led to attenuated pain responses induced by complete Freund's adjuvant (CFA). FGF13 was expressed predominantly in small-diameter DRG neurons. CFA treatment resulted in an increased expression of FGF13 proteins as well as an increased excitability in nociceptive DRG neurons which was inhibited when FGF13 was absent. The role of FGF13 in neuronal excitability of DRG was linked to its modulation of voltage-gated Na+ channels mediated by microtubules. Overexpression of FGF13, but not FGF13 mutant which lacks the ability to bind and stabilize microtubules, rescued the decreased neuronal excitability and Na+ current density in DRG neurons of Fgf13-/Y mice. Conclusion This study revealed that FGF13 could stabilize microtubules to modulate sodium channel function in DRG neurons and modulate inflammatory pain. This study provides a novel mechanism for FGF13 modulation of sodium channel function and suggests that FGF13 might be a novel target for inflammatory pain treatment.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Jing Yang
- Department of Physiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Handong Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Bin Shan
- Department of Pharmacy, The Forth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Chengyu Yin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Hang Yu
- College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xuerou Zhang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Zishan Dong
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yulou Yu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Ran Zhao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Boyi Liu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
31
|
FGF13 Is Required for Histamine-Induced Itch Sensation by Interaction with Na V1.7. J Neurosci 2020; 40:9589-9601. [PMID: 33172979 DOI: 10.1523/jneurosci.0599-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/17/2023] Open
Abstract
Itch can be induced by activation of small-diameter DRG neurons, which express abundant intracellular fibroblast growth factor 13 (FGF13). Although FGF13 is revealed to be essential for heat nociception, its role in mediating itch remains to be investigated. Here, we reported that loss of FGF13 in mouse DRG neurons impaired the histamine-induced scratching behavior. Calcium imaging showed that the percentage of histamine-responsive DRG neurons was largely decreased in FGF13-deficient mice; and consistently, electrophysiological recording exhibited that histamine failed to evoke action potential firing in most DRG neurons from these mice. Given that the reduced histamine-evoked neuronal response was caused by knockdown of FGF13 but not by FGF13A deficiency, FGF13B was supposed to mediate this process. Furthermore, overexpression of histamine Type 1 receptor H1R, but not H2R, H3R, nor H4R, increased the percentage of histamine-responsive DRG neurons, and the scratching behavior in FGF13-deficient mice was highly reduced by selective activation of H1R, suggesting that H1R is mainly required for FGF13-mediated neuronal response and scratching behavior induced by histamine. However, overexpression of H1R failed to rescue the histamine-evoked neuronal response in FGF13-deficient mice. Histamine enhanced the FGF13 interaction with NaV1.7. Disruption of this interaction by a membrane-permeable competitive peptide, GST-Flag-NaV1.7CT-TAT, reduced the percentage of histamine-responsive DRG neurons, and impaired the histamine-induced scratching, indicating that the FGF13/NaV1.7 interaction is a key molecular determinant in the histamine-induced itch sensation. Therefore, our study reveals a novel role of FGF13 in mediating itch sensation via the interaction of NaV1.7 in the peripheral nervous system.SIGNIFICANCE STATEMENT Scratching induced by itch brings serious tissue damage in chronic itchy diseases, and targeting itch-sensing molecules is crucial for its therapeutic intervention. Here, we reveal that FGF13 is required for the neuronal excitation and scratching behavior induced by histamine. We further provide the evidence that the histamine-evoked neuronal response is mainly mediated by histamine Type 1 receptor H1R, and is largely attenuated in FGF13-deficent mice. Importantly, we identify that histamine enhances the FGF13/NaV1.7 interaction, and disruption of this interaction reduces histamine-evoked neuronal excitation and highly impairs histamine-induced scratching behavior. Additionally, we also find that FGF13 is involved in 5-hydroxytryptamine-induced scratching behavior and hapten 1-fluoro-2,4-dinitrobenzene-induced chronic itch.
Collapse
|
32
|
Wu K, Yue J, Shen K, He J, Zhu G, Liu S, Zhang C, Yang H. Increased expression of fibroblast growth factor 13 in cortical lesions of the focal cortical dysplasia. Brain Res Bull 2020; 168:36-44. [PMID: 33285262 DOI: 10.1016/j.brainresbull.2020.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 02/08/2023]
Abstract
Focal cortical dysplasias (FCDs) are well recognized as important causes of medically intractable epilepsy in both children and adults. To explore the potential role of fibroblast growth factor 13 (FGF13) in intractable epilepsy caused by FCDs, we examined the expression of FGF13 in cortical lesions from 23 patients with FCD type Ia (FCDIa), 24 patients with FCD type IIa (FCDIIa), and 12 patients with FCD type IIb (FCDIIb), and we compared the results with the FGF13 expression levels in control cortex (CTX) brain tissues from 12 nonepileptic normal subjects. Both the mRNA levels and protein levels of FGF13 were significantly higher in the cortical lesions from patients with FCD than in the control cortices. The immunohistochemical results showed that strong FGF13 immunoreactivity was observed in misshapen cells, including neuronal microcolumns, hypertrophic neurons, dysmorphic neurons, and most balloon cells. Moreover, double-label immunofluorescence analyses confirmed that FGF13 was mainly localized in neurons and nearly absent in glia-like cells. Taken together, our results suggest that the overexpression of FGF13 in FCDs and the cell-specific distribution patterns of FGF13 in misshapen neurons in FCDs could potentially contribute to intractable epilepsy caused by FCDs.
Collapse
Affiliation(s)
- Kefu Wu
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jiong Yue
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Kaifeng Shen
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jiaojiang He
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gang Zhu
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shiyong Liu
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Chunqing Zhang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China.
| | - Hui Yang
- Epilepsy Research Center of PLA, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
33
|
Huang Y, Lu Y, Zhao X, Zhang J, Zhang F, Chen Y, Bi L, Gu J, Jiang Z, Wu X, Li Q, Liu Y, Shen J, Liu X. Cytokine activin C ameliorates chronic neuropathic pain in peripheral nerve injury rodents by modulating the TRPV1 channel. Br J Pharmacol 2020; 177:5642-5657. [PMID: 33095918 PMCID: PMC7707095 DOI: 10.1111/bph.15284] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/06/2020] [Accepted: 09/25/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE The cytokine activin C is mainly expressed in small-diameter dorsal root ganglion (DRG) neurons and suppresses inflammatory pain. However, the effects of activin C in neuropathic pain remain elusive. EXPERIMENTAL APPROACH Male rats and wild-type and TRPV1 knockout mice with peripheral nerve injury - sciatic nerve axotomy and spinal nerve ligation in rats; chronic constriction injury (CCI) in mice - provided models of chronic neuropathic pain. Ipsilateral lumbar (L)4-5 DRGs were assayed for activin C expression. Chronic neuropathic pain animals were treated with intrathecal or locally pre-administered activin C or the vehicle. Nociceptive behaviours and pain-related markers in L4-5 DRGs and spinal cord were evaluated. TRPV1 channel modulation by activin C was measured. KEY RESULTS Following peripheral nerve injury, expression of activin βC subunit mRNA and activin C protein was markedly up-regulated in L4-5 DRGs of animals with axotomy, SNL or CCI. [Correction added on 26 November 2020, after first online publication: The preceding sentence has been corrected in this current version.] Intrathecal activin C dose-dependently inhibited neuropathic pain in spinal nerve ligated rats. Local pre-administration of activin C decreased neuropathic pain, macrophage infiltration into ipsilateral L4-5 DRGs and microglial reaction in L4-5 spinal cords of mice with CCI. In rat DRG neurons, activin C enhanced capsaicin-induced TRPV1 currents. Pre-treatment with activin C reduced capsaicin-evoked acute hyperalgesia and normalized capsaicin-evoked persistent hypothermia in mice. Finally, the analgesic effect of activin C was abolished in TRPV1 knockout mice with CCI. CONCLUSION AND IMPLICATIONS Activin C inhibits neuropathic pain by modulating TRPV1 channels, revealing potential analgesic applications in chronic neuropathic pain therapy.
Collapse
Affiliation(s)
- Ya‐Kun Huang
- School of PharmacyNantong UniversityNantongChina
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Yu‐Gang Lu
- Department of Anesthesiology, Shanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Xin Zhao
- Department of GeriatricsRenji Hospital, School of Medicine, Shanghai Jiaotong UniversityShanghaiChina
| | - Jing‐Bing Zhang
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | | | - Yong Chen
- School of PharmacyNantong UniversityNantongChina
| | - Ling‐Bo Bi
- School of PharmacyNantong UniversityNantongChina
| | - Jia‐Hui Gu
- School of PharmacyNantong UniversityNantongChina
| | - Zuo‐Jie Jiang
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Xiao‐Man Wu
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Qing‐Yi Li
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Yanli Liu
- College of Pharmaceutical ScienceSoochow UniversitySuzhouChina
| | - Jian‐Xin Shen
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Xing‐Jun Liu
- School of PharmacyNantong UniversityNantongChina
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| |
Collapse
|
34
|
Park DS, Shekhar A, Santucci J, Redel-Traub G, Solinas S, Mintz S, Lin X, Chang EW, Narke D, Xia Y, Goldfarb M, Fishman GI. Ionic Mechanisms of Impulse Propagation Failure in the FHF2-Deficient Heart. Circ Res 2020; 127:1536-1548. [PMID: 32962518 DOI: 10.1161/circresaha.120.317349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE FHFs (fibroblast growth factor homologous factors) are key regulators of sodium channel (NaV) inactivation. Mutations in these critical proteins have been implicated in human diseases including Brugada syndrome, idiopathic ventricular arrhythmias, and epileptic encephalopathy. The underlying ionic mechanisms by which reduced Nav availability in Fhf2 knockout (Fhf2KO) mice predisposes to abnormal excitability at the tissue level are not well defined. OBJECTIVE Using animal models and theoretical multicellular linear strands, we examined how FHF2 orchestrates the interdependency of sodium, calcium, and gap junctional conductances to safeguard cardiac conduction. METHODS AND RESULTS Fhf2KO mice were challenged by reducing calcium conductance (gCaV) using verapamil or by reducing gap junctional conductance (Gj) using carbenoxolone or by backcrossing into a cardiomyocyte-specific Cx43 (connexin 43) heterozygous background. All conditions produced conduction block in Fhf2KO mice, with Fhf2 wild-type (Fhf2WT) mice showing normal impulse propagation. To explore the ionic mechanisms of block in Fhf2KO hearts, multicellular linear strand models incorporating FHF2-deficient Nav inactivation properties were constructed and faithfully recapitulated conduction abnormalities seen in mutant hearts. The mechanisms of conduction block in mutant strands with reduced gCaV or diminished Gj are very different. Enhanced Nav inactivation due to FHF2 deficiency shifts dependence onto calcium current (ICa) to sustain electrotonic driving force, axial current flow, and action potential (AP) generation from cell-to-cell. In the setting of diminished Gj, slower charging time from upstream cells conspires with accelerated Nav inactivation in mutant strands to prevent sufficient downstream cell charging for AP propagation. CONCLUSIONS FHF2-dependent effects on Nav inactivation ensure adequate sodium current (INa) reserve to safeguard against numerous threats to reliable cardiac impulse propagation.
Collapse
Affiliation(s)
- David S Park
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| | - Akshay Shekhar
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine.,Regeneron Pharmaceuticals, Tarrytown, NY (A.S.)
| | - John Santucci
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| | - Gabriel Redel-Traub
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| | - Sergio Solinas
- University of Zurich, Institute of Neuroinformatics, Switzerland (S.S.).,Hunter College of City University, Department of Biological Sciences, New York (S.S., M.G.)
| | - Shana Mintz
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| | - Xianming Lin
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| | - Ernest Whanwook Chang
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| | - Deven Narke
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| | - Yuhe Xia
- Department of Population Health (Y.X.), New York University School of Medicine
| | - Mitchell Goldfarb
- Hunter College of City University, Department of Biological Sciences, New York (S.S., M.G.)
| | - Glenn I Fishman
- The Leon H. Charney Division of Cardiology (D.S.P., A.S., J.S., G.R.-T., S.M., X.L., E.W.C., D.N., G.I.F.), New York University School of Medicine
| |
Collapse
|
35
|
Alles SR, Gomez K, Moutal A, Khanna R. Putative roles of SLC7A5 (LAT1) transporter in pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100050. [PMID: 32715162 PMCID: PMC7369351 DOI: 10.1016/j.ynpai.2020.100050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022]
Abstract
Large amino acid transporter 1 (LAT1), also known as SLC7A5, is an essential amino acid transporter that forms a heterodimeric complex with the glycoprotein cell-surface antigen heavy chain (4F2hc (CD98, SLC3A2)). Within nociceptive pathways, LAT1 is expressed in the dorsal root ganglia and spinal cord. Although LAT1 expression is upregulated following spinal cord injury, little is known about LAT1 in neuropathic pain. To date, only circumstantial evidence supports LAT1/4F2hc's role in pain. Notably, LAT1's expression and regulation link it to key cell types and pathways implicated in pain. Transcriptional regulation of LAT1 expression occurs via the Wnt/frizzled/β-catenin signal transduction pathway, which has been shown to be involved in chronic pain. The LAT1/4F2hc complex may also be involved in pain pathways related to T- and B-cells. LAT1's expression induces activation of the mammalian target of rapamycin (mTOR) signaling axis, which is involved in inflammation and neuropathic pain. Similarly, hypoxia and cancer induce activation of hypoxia-inducible factor 2 alpha, promoting not only LAT1's expression but also mTORC1's activation. Perhaps the strongest evidence linking LAT1 to pain is its interactions with key voltage-gated ion channels connected to nociception, namely the voltage-gated potassium channels Kv1.1 and Kv1.2 and the voltage-gated sodium channel Nav1.7. Through functional regulation of these channels, LAT1 may play a role in governing the excitatory to inhibitory ratio which is altered in chronic neuropathic pain states. Remarkably, the most direct role for LAT1 in pain is to mediate the influx of gabapentin and pregabalin, two first-line neuropathic pain drugs, that indirectly inhibit high voltage-activated calcium channel auxiliary subunit α2δ-1. In this review, we discuss the expression, regulation, relevant signaling pathways, and protein interactions of LAT1 that may link it to the development and/or maintenance of pain. We hypothesize that LAT1 expressed in nociceptive pathways may be a viable new target in pain.
Collapse
Affiliation(s)
- Sascha R.A. Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, United States
| | - Kimberly Gomez
- Department of Pharmacology, University of Arizona, United States
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, United States
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, United States
- Department of Anesthesiology, College of Medicine, The University of Arizona, Tucson, AZ 85724, United States
- BIO5 Institute, University of Arizona, 1657 East Helen Street Tucson, AZ 85719, United States
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, United States
- Regulonix Holding Inc., Tucson, AZ, United States
| |
Collapse
|
36
|
Hemokinin-1 Gene Expression Is Upregulated in Trigeminal Ganglia in an Inflammatory Orofacial Pain Model: Potential Role in Peripheral Sensitization. Int J Mol Sci 2020; 21:ijms21082938. [PMID: 32331300 PMCID: PMC7215309 DOI: 10.3390/ijms21082938] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/12/2020] [Accepted: 04/19/2020] [Indexed: 12/19/2022] Open
Abstract
A large percentage of primary sensory neurons in the trigeminal ganglia (TG) contain neuropeptides such as tachykinins or calcitonin gene-related peptide. Neuropeptides released from the central terminals of primary afferents sensitize the secondary nociceptive neurons in the trigeminal nucleus caudalis (TNC), but also activate glial cells contributing to neuroinflammation and consequent sensitization in chronic orofacial pain and migraine. In the present study, we investigated the newest member of the tachykinin family, hemokinin-1 (HK-1) encoded by the Tac4 gene in the trigeminal system. HK-1 had been shown to participate in inflammation and hyperalgesia in various models, but its role has not been investigated in orofacial pain or headache. In the complete Freund’s adjuvant (CFA)-induced inflammatory orofacial pain model, we showed that Tac4 expression increased in the TG in response to inflammation. Duration-dependent Tac4 upregulation was associated with the extent of the facial allodynia. Tac4 was detected in both TG neurons and satellite glial cells (SGC) by the ultrasensitive RNAscope in situ hybridization. We also compared gene expression changes of selected neuronal and glial sensitization and neuroinflammation markers between wild-type and Tac4-deficient (Tac4-/-) mice. Expression of the SGC/astrocyte marker in the TG and TNC was significantly lower in intact and saline/CFA-treated Tac4-/- mice. The procedural stress-related increase of the SGC/astrocyte marker was also strongly attenuated in Tac4-/- mice. Analysis of TG samples with a mouse neuroinflammation panel of 770 genes revealed that regulation of microglia and cytotoxic cell-related genes were significantly different in saline-treated Tac4-/- mice compared to their wild-types. It is concluded that HK-1 may participate in neuron-glia interactions both under physiological and inflammatory conditions and mediate pain in the trigeminal system.
Collapse
|
37
|
Tower J, Pomatto LCD, Davies KJA. Sex differences in the response to oxidative and proteolytic stress. Redox Biol 2020; 31:101488. [PMID: 32201219 PMCID: PMC7212483 DOI: 10.1016/j.redox.2020.101488] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 12/16/2022] Open
Abstract
Sex differences in diseases involving oxidative and proteolytic stress are common, including greater ischemic heart disease, Parkinson disease and stroke in men, and greater Alzheimer disease in women. Sex differences are also observed in stress response of cells and tissues, where female cells are generally more resistant to heat and oxidative stress-induced cell death. Studies implicate beneficial effects of estrogen, as well as cell-autonomous effects including superior mitochondrial function and increased expression of stress response genes in female cells relative to male cells. The p53 and forkhead box (FOX)-family genes, heat shock proteins (HSPs), and the apoptosis and autophagy pathways appear particularly important in mediating sex differences in stress response.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA90089, USA.
| | - Laura C D Pomatto
- National Institute on General Medical Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kelvin J A Davies
- Molecular and Computational Biology Program, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA90089, USA; Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, USA
| |
Collapse
|
38
|
Tong DL, Kempsell KE, Szakmany T, Ball G. Development of a Bioinformatics Framework for Identification and Validation of Genomic Biomarkers and Key Immunopathology Processes and Controllers in Infectious and Non-infectious Severe Inflammatory Response Syndrome. Front Immunol 2020; 11:380. [PMID: 32318053 PMCID: PMC7147506 DOI: 10.3389/fimmu.2020.00380] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as dysregulated host response caused by systemic infection, leading to organ failure. It is a life-threatening condition, often requiring admission to an intensive care unit (ICU). The causative agents and processes involved are multifactorial but are characterized by an overarching inflammatory response, sharing elements in common with severe inflammatory response syndrome (SIRS) of non-infectious origin. Sepsis presents with a range of pathophysiological and genetic features which make clinical differentiation from SIRS very challenging. This may reflect a poor understanding of the key gene inter-activities and/or pathway associations underlying these disease processes. Improved understanding is critical for early differential recognition of sepsis and SIRS and to improve patient management and clinical outcomes. Judicious selection of gene biomarkers suitable for development of diagnostic tests/testing could make differentiation of sepsis and SIRS feasible. Here we describe a methodologic framework for the identification and validation of biomarkers in SIRS, sepsis and septic shock patients, using a 2-tier gene screening, artificial neural network (ANN) data mining technique, using previously published gene expression datasets. Eight key hub markers have been identified which may delineate distinct, core disease processes and which show potential for informing underlying immunological and pathological processes and thus patient stratification and treatment. These do not show sufficient fold change differences between the different disease states to be useful as primary diagnostic biomarkers, but are instrumental in identifying candidate pathways and other associated biomarkers for further exploration.
Collapse
Affiliation(s)
- Dong Ling Tong
- Artificial Intelligence Laboratory, Faculty of Engineering and Computing, First City University College, Petaling Jaya, Malaysia.,School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Karen E Kempsell
- Public Health England, National Infection Service, Porton Down, Salisbury, United Kingdom
| | - Tamas Szakmany
- Department of Anaesthesia Intensive Care and Pain Medicine, Division of Population Medicine, Cardiff University, Cardiff, United Kingdom
| | - Graham Ball
- School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
39
|
Johnstone CN, Pattison AD, Harrison PF, Powell DR, Lock P, Ernst M, Anderson RL, Beilharz TH. FGF13 promotes metastasis of triple-negative breast cancer. Int J Cancer 2020; 147:230-243. [PMID: 31957002 DOI: 10.1002/ijc.32874] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 12/01/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer (TNBC) represents 10-20% of all human ductal adenocarcinomas and has a poor prognosis relative to other subtypes, due to the high propensity to develop distant metastases. Hence, new molecular targets for therapeutic intervention are needed for TNBC. We recently conducted a rigorous phenotypic and genomic characterization of four isogenic populations of MDA-MB-231 human triple-negative breast cancer cells that possess a range of intrinsic spontaneous metastatic capacities in vivo, ranging from nonmetastatic (MDA-MB-231_ATCC) to highly metastatic to lung, liver, spleen and spine (MDA-MB-231_HM). Gene expression profiling of primary tumours by RNA-Seq identified the fibroblast growth factor homologous factor, FGF13, as highly upregulated in aggressively metastatic MDA-MB-231_HM tumours. Clinically, higher FGF13 mRNA expression was associated with significantly worse relapse free survival in both luminal A and basal-like human breast cancers but was not associated with other clinical variables and was not upregulated in primary tumours relative to normal mammary gland. Stable FGF13 depletion restricted in vitro colony forming ability in MDA-MB-231_HM TNBC cells but not in oestrogen receptor (ER)-positive MCF-7 or MDA-MB-361 cells. However, despite augmenting MDA-MB-231_HM cell migration and invasion in vitro, FGF13 suppression almost completely blocked the spontaneous metastasis of MDA-MB-231_HM orthotopic xenografts to both lung and liver while having negligible impact on primary tumour growth. Together, these data indicate that FGF13 may represent a therapeutic target for blocking metastatic outgrowth of certain TNBCs. Further evaluation of the roles of individual FGF13 protein isoforms in progression of the different subtypes of breast cancer is warranted.
Collapse
Affiliation(s)
- Cameron N Johnstone
- Cancer Research Division, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Department of Clinical Pathology, University of Melbourne, Parkville, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Andrew D Pattison
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Paul F Harrison
- Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| | - David R Powell
- Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| | - Peter Lock
- LIMS Bioimaging Facility, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Robin L Anderson
- Cancer Research Division, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Department of Clinical Pathology, University of Melbourne, Parkville, VIC, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Traude H Beilharz
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| |
Collapse
|
40
|
Wang F, Bélanger E, Côté SL, Desrosiers P, Prescott SA, Côté DC, De Koninck Y. Sensory Afferents Use Different Coding Strategies for Heat and Cold. Cell Rep 2019; 23:2001-2013. [PMID: 29768200 DOI: 10.1016/j.celrep.2018.04.065] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 02/22/2018] [Accepted: 04/13/2018] [Indexed: 11/24/2022] Open
Abstract
Primary afferents transduce environmental stimuli into electrical activity that is transmitted centrally to be decoded into corresponding sensations. However, it remains unknown how afferent populations encode different somatosensory inputs. To address this, we performed two-photon Ca2+ imaging from thousands of dorsal root ganglion (DRG) neurons in anesthetized mice while applying mechanical and thermal stimuli to hind paws. We found that approximately half of all neurons are polymodal and that heat and cold are encoded very differently. As temperature increases, more heating-sensitive neurons are activated, and most individual neurons respond more strongly, consistent with graded coding at population and single-neuron levels, respectively. In contrast, most cooling-sensitive neurons respond in an ungraded fashion, inconsistent with graded coding and suggesting combinatorial coding, based on which neurons are co-activated. Although individual neurons may respond to multiple stimuli, our results show that different stimuli activate distinct combinations of diversely tuned neurons, enabling rich population-level coding.
Collapse
Affiliation(s)
- Feng Wang
- CERVO Brain Research Centre, Québec Mental Health Institute, Quebec City, QC, Canada
| | - Erik Bélanger
- CERVO Brain Research Centre, Québec Mental Health Institute, Quebec City, QC, Canada; Center for Optics, Photonics and Lasers (COPL), Laval University, Quebec City, QC, Canada
| | - Sylvain L Côté
- CERVO Brain Research Centre, Québec Mental Health Institute, Quebec City, QC, Canada
| | - Patrick Desrosiers
- CERVO Brain Research Centre, Québec Mental Health Institute, Quebec City, QC, Canada; Department of Physics, Physical Engineering, and Optics, Laval University, Quebec City, QC, Canada
| | - Steven A Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Physiology and the Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Daniel C Côté
- CERVO Brain Research Centre, Québec Mental Health Institute, Quebec City, QC, Canada; Center for Optics, Photonics and Lasers (COPL), Laval University, Quebec City, QC, Canada; Department of Physics, Physical Engineering, and Optics, Laval University, Quebec City, QC, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Québec Mental Health Institute, Quebec City, QC, Canada; Center for Optics, Photonics and Lasers (COPL), Laval University, Quebec City, QC, Canada; Department of Psychiatry and Neuroscience, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
41
|
Sinden DS, Holman CD, Bare CJ, Sun X, Gade AR, Cohen DE, Pitt GS. Knockout of the X-linked Fgf13 in the hypothalamic paraventricular nucleus impairs sympathetic output to brown fat and causes obesity. FASEB J 2019; 33:11579-11594. [PMID: 31339804 PMCID: PMC6994920 DOI: 10.1096/fj.201901178r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor (FGF)13, a nonsecreted, X-linked, FGF homologous factor, is differentially expressed in adipocytes in response to diet, yet Fgf13's role in metabolism has not been explored. Heterozygous Fgf13 knockouts fed normal chow and housed at 22°C showed hyperactivity accompanying reduced core temperature and obesity when housed at 30°C. Those heterozygous knockouts showed defects in thermogenesis even at 30°C and an inability to protect core temperature. Surprisingly, we detected trivial FGF13 in adipose of wild-type mice fed normal chow and no obesity in adipose-specific heterozygous knockouts housed at 30°C, and we detected an intact brown fat response through exogenous β3 agonist stimulation, suggesting a defect in sympathetic drive to brown adipose tissue. In contrast, hypothalamic-specific ablation of Fgf13 recapitulated weight gain at 30°C. Norepinephrine turnover in brown fat was reduced at both housing temperatures. Thus, our data suggest that impaired CNS regulation of sympathetic activation of brown fat underlies obesity and thermogenesis in Fgf13 heterozygous knockouts fed normal chow.-Sinden, D. S., Holman, C. D., Bare, C. J., Sun, X., Gade, A. R., Cohen, D. E., Pitt, G. S. Knockout of the X-linked Fgf13 in the hypothalamic paraventricular nucleus impairs sympathetic output to brown fat and causes obesity.
Collapse
Affiliation(s)
- Daniel S. Sinden
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Corey D. Holman
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Curtis J. Bare
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Xiaolu Sun
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Aravind R. Gade
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - David E. Cohen
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
42
|
Effraim PR, Huang J, Lampert A, Stamboulian S, Zhao P, Black JA, Dib-Hajj SD, Waxman SG. Fibroblast growth factor homologous factor 2 (FGF-13) associates with Nav1.7 in DRG neurons and alters its current properties in an isoform-dependent manner. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100029. [PMID: 31223136 PMCID: PMC6565799 DOI: 10.1016/j.ynpai.2019.100029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 11/29/2022]
Abstract
Fibroblast Growth Factor Homologous Factors (FHF) constitute a subfamily of FGF proteins with four prototypes (FHF1-4; also known as FGF11-14). FHF proteins have been shown to bind directly to the membrane-proximal segment of the C-terminus in voltage-gated sodium channels (Nav), and regulate current density, availability, and frequency-dependent inhibition of sodium currents. Members of the FHF2 subfamily, FHF2A and FHF2B, differ in the length and sequence of their N-termini, and, importantly, differentially regulate Nav1.6 gating properties. Using immunohistochemistry, we show that FHF2 isoforms are expressed in adult dorsal root ganglion (DRG) neurons where they co-localize with Nav1.6 and Nav1.7. FHF2A and FHF2B show differential localization in neuronal compartments in DRG neurons, and levels of expression of FHF2 factors are down-regulated following sciatic nerve axotomy. Because Nav1.7 in nociceptors plays a critical role in pain, we reasoned that its interaction with FHF2 isoforms might regulate its current properties. Using whole-cell patch clamp in heterologous expression systems, we show that the expression of FHF2A in HEK293 cell line stably expressing Nav1.7 channels causes no change in activation, whereas FHF2B depolarizes activation. Both FHF2 isoforms depolarize fast-inactivation. Additionally, FHF2A causes an accumulation of inactivated channels at all frequencies tested due to a slowing of recovery from inactivation, whereas FHF2B has little effect on these properties of Nav1.7. Measurements of the Nav1.7 current in DRG neurons in which FHF2 levels are knocked down confirmed the effects of FHF2A on repriming, and FHF2B on activation, however FHF2A and B did not have an effect on fast inactivation. Our data demonstrates that FHF2 does indeed regulate the current properties of Nav1.7 and does so in an isoform and cell-specific manner.
Collapse
Affiliation(s)
- Philip R. Effraim
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Jianying Huang
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Severine Stamboulian
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Joel A. Black
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D. Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
43
|
Cao S, Li Q, Hou J, Li Z, Cao X, Liu X, Qin B. Intrathecal TRPM8 blocking attenuates cold hyperalgesia via PKC and NF-κB signaling in the dorsal root ganglion of rats with neuropathic pain. J Pain Res 2019; 12:1287-1296. [PMID: 31114308 PMCID: PMC6497852 DOI: 10.2147/jpr.s197168] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/27/2019] [Indexed: 12/23/2022] Open
Abstract
Background: TRPM8 channel plays central roles in the sensitization of nociceptive transduction and is thought as one of the potential targets for the treatment of neuropathic pain. However, the specific molecular mechanisms are still less clear. Methods: Sciatic chronic constriction injury (CCI) rats were intrathecally administered with AMTB (TRPM8-selective antagonist) or PDTC (nuclear factor-kappa B (NF-κB) inhibitor). Cold-, thermal- and mechanical-pain thresholds were examined in CCI and sham-operated rats before and after intrathecal administration of AMTB or PDTC. Protein expression levels of TRPM8 and NF-κB p65, p-PKC/PKC value and p-PKA/PKA value in the CCI ipsilateral L4-6 dorsal root ganglions (DRGs) were analyzed. In addition, the co-expression of TRPM8 and NF-κB was evaluated in DRG. Results: Intrathecal injection of AMTB decreased the cold hypersensitivity and aggravated the thermal-hyperalgesia in the next 2 weeks after CCI surgery. The protein expression of TRPM8 and NF-κB p65 in the ipsilateral DRGs significantly increased after CCI surgery, which can be reversed by intrathecal administration of AMTB. The PKC, PKA, p-PKC/PKC and p-PKA/PKA values showed significantly increase after CCI surgery, while intrathecal AMTB administration offset the expression increase of PKC, p-PKC and p-PKC/PKC but PKA or p-PKA/PKA in the DRG. NF-κB inhibitor not only efficiently increased the cold-, thermal-pain threshold of CCI rats, but also enhanced AMTB’s anti-cold pain effect although exerted no anti-thermal hyperalgesia effect compared with TRPM8 blockade group. Immunofluorescence results showed co-expression of TRPM8 and NF-κB in DRG neurons. Conclusion: TRPM8 channels in DRGs participate in the pathogenesis of cold and thermal hyperalgesia (not mechanical allodynia) in rats with neuropathic pain, which could be regulated by PKC (not PKA) and NF-κB signaling. TRPM8 channel, PKC and NF-κB are potential targets for cold hyperalgesia treatment in neuropathic pain patients.
Collapse
Affiliation(s)
- Song Cao
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Qingmei Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Jingfeng Hou
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Zhourui Li
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Xinya Cao
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Xiaohong Liu
- Department of Physiology, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Bangyong Qin
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China
| |
Collapse
|
44
|
Schneider ER, Anderson EO, Feketa VV, Mastrotto M, Nikolaev YA, Gracheva EO, Bagriantsev SN. A Cross-Species Analysis Reveals a General Role for Piezo2 in Mechanosensory Specialization of Trigeminal Ganglia from Tactile Specialist Birds. Cell Rep 2019; 26:1979-1987.e3. [PMID: 30784581 PMCID: PMC6420409 DOI: 10.1016/j.celrep.2019.01.100] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/20/2018] [Accepted: 01/25/2019] [Indexed: 12/22/2022] Open
Abstract
A major challenge in biology is to link cellular and molecular variations with behavioral phenotypes. Here, we studied somatosensory neurons from a panel of bird species from the family Anatidae, known for their tactile-based foraging behavior. We found that tactile specialists exhibit a proportional expansion of neuronal mechanoreceptors in trigeminal ganglia. The expansion of mechanoreceptors occurs via neurons with intermediately and slowly inactivating mechanocurrent. Such neurons contain the mechanically gated Piezo2 ion channel whose expression positively correlates with the expression of factors responsible for the development and function of mechanoreceptors. Conversely, Piezo2 expression negatively correlates with expression of molecules mediating the detection of temperature and pain, suggesting that the expansion of Piezo2-containing mechanoreceptors with prolonged mechanocurrent occurs at the expense of thermoreceptors and nociceptors. Our study suggests that the trade-off between neuronal subtypes is a general mechanism of tactile specialization at the level of somatosensory system.
Collapse
Affiliation(s)
- Eve R Schneider
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Evan O Anderson
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Viktor V Feketa
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Marco Mastrotto
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Yury A Nikolaev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA.
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar St., New Haven, CT 06510, USA.
| |
Collapse
|
45
|
Chew LA, Bellampalli SS, Dustrude ET, Khanna R. Mining the Na v1.7 interactome: Opportunities for chronic pain therapeutics. Biochem Pharmacol 2019; 163:9-20. [PMID: 30699328 DOI: 10.1016/j.bcp.2019.01.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/24/2019] [Indexed: 12/14/2022]
Abstract
The peripherally expressed voltage-gated sodium NaV1.7 (gene SCN9A) channel boosts small stimuli to initiate firing of pain-signaling dorsal root ganglia (DRG) neurons and facilitates neurotransmitter release at the first synapse within the spinal cord. Mutations in SCN9A produce distinct human pain syndromes. Widely acknowledged as a "gatekeeper" of pain, NaV1.7 has been the focus of intense investigation but, to date, no NaV1.7-selective drugs have reached the clinic. Elegant crystallographic studies have demonstrated the potential of designing highly potent and selective NaV1.7 compounds but their therapeutic value remains untested. Transcriptional silencing of NaV1.7 by a naturally expressed antisense transcript has been reported in rodents and humans but whether this represents a viable opportunity for designing NaV1.7 therapeutics is currently unknown. The demonstration that loss of NaV1.7 function is associated with upregulation of endogenous opioids and potentiation of mu- and delta-opioid receptor activities, suggests that targeting only NaV1.7 may be insufficient for analgesia. However, the link between opioid-dependent analgesic mechanisms and function of sodium channels and intracellular sodium-dependent signaling remains controversial. Thus, additional new targets - regulators, modulators - are needed. In this context, we mine the literature for the known interactome of NaV1.7 with a focus on protein interactors that affect the channel's trafficking or link it to opioid signaling. As a case study, we present antinociceptive evidence of allosteric regulation of NaV1.7 by the cytosolic collapsin response mediator protein 2 (CRMP2). Throughout discussions of these possible new targets, we offer thoughts on the therapeutic implications of modulating NaV1.7 function in chronic pain.
Collapse
Affiliation(s)
- Lindsey A Chew
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Shreya S Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Erik T Dustrude
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA; Graduate Interdisciplinary Program in Neuroscience, College of Medicine, University of Arizona, Tucson, AZ, USA; The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ 85724, USA.
| |
Collapse
|
46
|
Favuzzi E, Deogracias R, Marques-Smith A, Maeso P, Jezequel J, Exposito-Alonso D, Balia M, Kroon T, Hinojosa AJ, F Maraver E, Rico B. Distinct molecular programs regulate synapse specificity in cortical inhibitory circuits. Science 2019; 363:413-417. [PMID: 30679375 DOI: 10.1126/science.aau8977] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022]
Abstract
How neuronal connections are established and organized into functional networks determines brain function. In the mammalian cerebral cortex, different classes of GABAergic interneurons exhibit specific connectivity patterns that underlie their ability to shape temporal dynamics and information processing. Much progress has been made toward parsing interneuron diversity, yet the molecular mechanisms by which interneuron-specific connectivity motifs emerge remain unclear. In this study, we investigated transcriptional dynamics in different classes of interneurons during the formation of cortical inhibitory circuits in mouse. We found that whether interneurons form synapses on the dendrites, soma, or axon initial segment of pyramidal cells is determined by synaptic molecules that are expressed in a subtype-specific manner. Thus, cell-specific molecular programs that unfold during early postnatal development underlie the connectivity patterns of cortical interneurons.
Collapse
Affiliation(s)
- Emilia Favuzzi
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Rubén Deogracias
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - André Marques-Smith
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Patricia Maeso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Julie Jezequel
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - David Exposito-Alonso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Maddalena Balia
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Tim Kroon
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Antonio J Hinojosa
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Elisa F Maraver
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| |
Collapse
|
47
|
CRISPR/Cas9 editing of Nf1 gene identifies CRMP2 as a therapeutic target in neurofibromatosis type 1-related pain that is reversed by (S)-Lacosamide. Pain 2018; 158:2301-2319. [PMID: 28809766 DOI: 10.1097/j.pain.0000000000001002] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a rare autosomal dominant disease linked to mutations of the Nf1 gene. Patients with NF1 commonly experience severe pain. Studies on mice with Nf1 haploinsufficiency have been instructive in identifying sensitization of ion channels as a possible cause underlying the heightened pain suffered by patients with NF1. However, behavioral assessments of Nf1 mice have led to uncertain conclusions about the potential causal role of Nf1 in pain. We used the clustered regularly interspaced short palindromic repeats (CRISPR)-associated 9 (CRISPR/Cas9) genome editing system to create and mechanistically characterize a novel rat model of NF1-related pain. Targeted intrathecal delivery of guide RNA/Cas9 nuclease plasmid in combination with a cationic polymer was used to generate allele-specific C-terminal truncation of neurofibromin, the protein encoded by the Nf1 gene. Rats with truncation of neurofibromin, showed increases in voltage-gated calcium (specifically N-type or CaV2.2) and voltage-gated sodium (particularly tetrodotoxin-sensitive) currents in dorsal root ganglion neurons. These gains-of-function resulted in increased nociceptor excitability and behavioral hyperalgesia. The cytosolic regulatory protein collapsin response mediator protein 2 (CRMP2) regulates activity of these channels, and also binds to the targeted C-terminus of neurofibromin in a tripartite complex, suggesting a possible mechanism underlying NF1 pain. Prevention of CRMP2 phosphorylation with (S)-lacosamide resulted in normalization of channel current densities, excitability, as well as of hyperalgesia following CRISPR/Cas9 truncation of neurofibromin. These studies reveal the protein partners that drive NF1 pain and suggest that CRMP2 is a key target for therapeutic intervention.
Collapse
|
48
|
Bang S, Yoo J, Gong X, Liu D, Han Q, Luo X, Chang W, Chen G, Im ST, Kim YH, Strong JA, Zhang MZ, Zhang JM, Lee SY, Ji RR. Differential Inhibition of Nav1.7 and Neuropathic Pain by Hybridoma-Produced and Recombinant Monoclonal Antibodies that Target Nav1.7 : Differential activities of Nav1.7-targeting monoclonal antibodies. Neurosci Bull 2018; 34:22-41. [PMID: 29333591 PMCID: PMC5799132 DOI: 10.1007/s12264-018-0203-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022] Open
Abstract
The voltage-gated Na+ channel subtype Nav1.7 is important for pain and itch in rodents and humans. We previously showed that a Nav1.7-targeting monoclonal antibody (SVmab) reduces Na+ currents and pain and itch responses in mice. Here, we investigated whether recombinant SVmab (rSVmab) binds to and blocks Nav1.7 similar to SVmab. ELISA tests revealed that SVmab was capable of binding to Nav1.7-expressing HEK293 cells, mouse DRG neurons, human nerve tissue, and the voltage-sensor domain II of Nav1.7. In contrast, rSVmab showed no or weak binding to Nav1.7 in these tests. Patch-clamp recordings showed that SVmab, but not rSVmab, markedly inhibited Na+ currents in Nav1.7-expressing HEK293 cells. Notably, electrical field stimulation increased the blocking activity of SVmab and rSVmab in Nav1.7-expressing HEK293 cells. SVmab was more effective than rSVmab in inhibiting paclitaxel-induced mechanical allodynia. SVmab also bound to human DRG neurons and inhibited their Na+ currents. Finally, potential reasons for the differential efficacy of SVmab and rSVmab and future directions are discussed.
Collapse
Affiliation(s)
- Sangsu Bang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Jiho Yoo
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA
| | - Xingrui Gong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Di Liu
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Qingjian Han
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Xin Luo
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Wonseok Chang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology and Biophysics, College of Medicine, Eulji University, 143-5 Yongdu-Dong, Jung-Gu, Daejeon, 34824, Korea
| | - Gang Chen
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Sang-Taek Im
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Yong Ho Kim
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
| | - Ma-Zhong Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA.
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA.
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA.
| |
Collapse
|
49
|
Decreased expression of fibroblast growth factor 13 in early-onset preeclampsia is associated with the increased trophoblast permeability. Placenta 2018; 62:43-49. [DOI: 10.1016/j.placenta.2017.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
|
50
|
Kanellopoulos AH, Koenig J, Huang H, Pyrski M, Millet Q, Lolignier S, Morohashi T, Gossage SJ, Jay M, Linley JE, Baskozos G, Kessler BM, Cox JJ, Dolphin AC, Zufall F, Wood JN, Zhao J. Mapping protein interactions of sodium channel Na V1.7 using epitope-tagged gene-targeted mice. EMBO J 2018; 37:427-445. [PMID: 29335280 PMCID: PMC5793798 DOI: 10.15252/embj.201796692] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 11/30/2017] [Accepted: 12/05/2017] [Indexed: 11/24/2022] Open
Abstract
The voltage-gated sodium channel NaV1.7 plays a critical role in pain pathways. We generated an epitope-tagged NaV1.7 mouse that showed normal pain behaviours to identify channel-interacting proteins. Analysis of NaV1.7 complexes affinity-purified under native conditions by mass spectrometry revealed 267 proteins associated with Nav1.7 in vivo The sodium channel β3 (Scn3b), rather than the β1 subunit, complexes with Nav1.7, and we demonstrate an interaction between collapsing-response mediator protein (Crmp2) and Nav1.7, through which the analgesic drug lacosamide regulates Nav1.7 current density. Novel NaV1.7 protein interactors including membrane-trafficking protein synaptotagmin-2 (Syt2), L-type amino acid transporter 1 (Lat1) and transmembrane P24-trafficking protein 10 (Tmed10) together with Scn3b and Crmp2 were validated by co-immunoprecipitation (Co-IP) from sensory neuron extract. Nav1.7, known to regulate opioid receptor efficacy, interacts with the G protein-regulated inducer of neurite outgrowth (Gprin1), an opioid receptor-binding protein, demonstrating a physical and functional link between Nav1.7 and opioid signalling. Further information on physiological interactions provided with this normal epitope-tagged mouse should provide useful insights into the many functions now associated with the NaV1.7 channel.
Collapse
Affiliation(s)
| | - Jennifer Koenig
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Honglei Huang
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Martina Pyrski
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Queensta Millet
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Stéphane Lolignier
- Molecular Nociception Group, WIBR, University College London, London, UK
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Toru Morohashi
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Samuel J Gossage
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Maude Jay
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - John E Linley
- Molecular Nociception Group, WIBR, University College London, London, UK
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | - Benedikt M Kessler
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford, UK
| | - James J Cox
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - John N Wood
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Jing Zhao
- Molecular Nociception Group, WIBR, University College London, London, UK
| |
Collapse
|