1
|
Cai Y, Li J, Fan K, Zhang D, Lu H, Chen G. Downregulation of chloride voltage-gated channel 7 contributes to hyperalgesia following spared nerve injury. J Biol Chem 2024; 300:107779. [PMID: 39276933 PMCID: PMC11490881 DOI: 10.1016/j.jbc.2024.107779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
Alterations in anion balance potential, along with the involvement of cation-chloride cotransporters, play pivotal roles in the development of hyperalgesia after peripheral nerve injury. Chloride voltage-gated channel seven (CLCN7) is the predominant member of the CLC protein family. Investigations on CLCN7 have focused primarily on its involvement in osteosclerosis and lysosomal storage disorders; nevertheless, its contribution to neuropathic pain has not been determined. In this investigation, we noted high expression of CLCN7 in neurons situated within the spinal dorsal horns and dorsal root ganglions (DRGs). Immunofluorescence analysis revealed that CLCN7 was predominantly distributed among IB4-positive and CGRP-positive neurons. Furthermore, the expression of CLCN7 was observed to be mainly reduced in neurons within the spinal dorsal horns and in small- and medium-sized neurons located in the DRGs of spared nerve injury mice. Knockdown of CLCN7 via siRNA in the DRGs resulted in increased mechanical and thermal hyperalgesia in naïve mice. Furthermore, the excitability of cultured DRG neurons in vitro was augmented upon treatment with CLCN7 siRNA. These findings suggested that CLCN7 downregulation following SNI was crucial for the manifestation of mechanical and thermal hyperalgesia, highlighting potential targeting strategies for treating neuropathic pain.
Collapse
Affiliation(s)
- Yunyun Cai
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China
| | - Jiajie Li
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Kewei Fan
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China
| | - Dongmei Zhang
- Department of Rehabilitation Medicine, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China; Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit of Immunology, Nantong First People's Hospital, Nantong, Jiangsu, China
| | - Hongjian Lu
- Department of Rehabilitation Medicine, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China; Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit of Immunology, Nantong First People's Hospital, Nantong, Jiangsu, China; Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China.
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
2
|
Kim JS, Kehrl JH. Inhibition of WNK Kinases in NK Cells Disrupts Cellular Osmoregulation and Control of Tumor Metastasis. J Innate Immun 2024; 16:451-469. [PMID: 39265537 PMCID: PMC11521464 DOI: 10.1159/000540744] [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: 11/22/2023] [Accepted: 08/01/2024] [Indexed: 09/14/2024] Open
Abstract
INTRODUCTION The serine/threonine with-no-lysine (WNK) kinase family function in blood pressure control, electrolyte homeostasis, and cellular osmoregulation. These kinases and their downstream effectors are considered promising therapeutic targets in hypertension and stroke. However, the role of WNK kinases in immune cells remains poorly understood. METHODS Using the small-molecule WNK kinase inhibitors WNK463 and WNK-IN-11, we investigated how WNK kinase inhibition affects natural killer (NK) cell physiology. RESULTS WNK kinase inhibition with WNK463 or WNK-IN-11 significantly decreased IL-2-activated NK cell volume, motility, and cytolytic activity. Treatment of NK cells with these inhibitors induced autophagy by activating AMPK and inhibiting mTOR signaling. Moreover, WNK kinase inhibition increased phosphorylation of Akt and c-Myc by misaligning activity of activating kinases and inhibitory phosphatases. Treatment of tumor-bearing mice with WNK463 impaired tumor metastasis control by adoptively transferred NK cells. CONCLUSION The catalytic activity of WNK kinases has a critical role of multiple aspects of NK cell physiology and their pharmacologic inhibition negatively impacts NK cell function.
Collapse
Affiliation(s)
- Ji Sung Kim
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Boyd-Shiwarski CR, Shiwarski DJ, Subramanya AR. A New Phase for WNK Kinase Signaling Complexes as Biomolecular Condensates. Physiology (Bethesda) 2024; 39:0. [PMID: 38624245 PMCID: PMC11460533 DOI: 10.1152/physiol.00013.2024] [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: 02/22/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
The purpose of this review is to highlight transformative advances that have been made in the field of biomolecular condensates, with special emphasis on condensate material properties, physiology, and kinases, using the With-No-Lysine (WNK) kinases as a prototypical example. To convey how WNK kinases illustrate important concepts for biomolecular condensates, we start with a brief history, focus on defining features of biomolecular condensates, and delve into some examples of how condensates are implicated in cellular physiology (and pathophysiology). We then highlight how WNK kinases, through the action of "WNK droplets" that ubiquitously regulate intracellular volume and kidney-specific "WNK bodies" that are implicated in distal tubule salt reabsorption and potassium homeostasis, exemplify many of the defining features of condensates. Finally, this review addresses the controversies within this emerging field and questions to address.
Collapse
Affiliation(s)
- Cary R Boyd-Shiwarski
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Daniel J Shiwarski
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
4
|
Zhang S, Meor Azlan NF, Josiah SS, Zhou J, Zhou X, Jie L, Zhang Y, Dai C, Liang D, Li P, Li Z, Wang Z, Wang Y, Ding K, Wang Y, Zhang J. The role of SLC12A family of cation-chloride cotransporters and drug discovery methodologies. J Pharm Anal 2023; 13:1471-1495. [PMID: 38223443 PMCID: PMC10785268 DOI: 10.1016/j.jpha.2023.09.002] [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: 03/04/2023] [Revised: 06/20/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
The solute carrier family 12 (SLC12) of cation-chloride cotransporters (CCCs) comprises potassium chloride cotransporters (KCCs, e.g. KCC1, KCC2, KCC3, and KCC4)-mediated Cl- extrusion, and sodium potassium chloride cotransporters (N[K]CCs, NKCC1, NKCC2, and NCC)-mediated Cl- loading. The CCCs play vital roles in cell volume regulation and ion homeostasis. Gain-of-function or loss-of-function of these ion transporters can cause diseases in many tissues. In recent years, there have been considerable advances in our understanding of CCCs' control mechanisms in cell volume regulations, with many techniques developed in studying the functions and activities of CCCs. Classic approaches to directly measure CCC activity involve assays that measure the transport of potassium substitutes through the CCCs. These techniques include the ammonium pulse technique, radioactive or nonradioactive rubidium ion uptake-assay, and thallium ion-uptake assay. CCCs' activity can also be indirectly observed by measuring γ-aminobutyric acid (GABA) activity with patch-clamp electrophysiology and intracellular chloride concentration with sensitive microelectrodes, radiotracer 36Cl-, and fluorescent dyes. Other techniques include directly looking at kinase regulatory sites phosphorylation, flame photometry, 22Na+ uptake assay, structural biology, molecular modeling, and high-throughput drug screening. This review summarizes the role of CCCs in genetic disorders and cell volume regulation, current methods applied in studying CCCs biology, and compounds developed that directly or indirectly target the CCCs for disease treatments.
Collapse
Affiliation(s)
- Shiyao Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Sunday Solomon Josiah
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoxia Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Lingjun Jie
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Yanhui Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Cuilian Dai
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Dong Liang
- Aurora Discovery Inc., Foshan, Guangdong, 528300, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Qingdao, Shandong, 266021, China
| | - Zhengqiu Li
- School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Jinwei Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
5
|
Sapio MR, King DM, Staedtler ES, Maric D, Jahanipour J, Kurochkina NA, Manalo AP, Ghetti A, Mannes AJ, Iadarola MJ. Expression pattern analysis and characterization of the hereditary sensory and autonomic neuropathy 2 A (HSAN2A) gene with no lysine kinase (WNK1) in human dorsal root ganglion. Exp Neurol 2023; 370:114552. [PMID: 37793538 DOI: 10.1016/j.expneurol.2023.114552] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/20/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
Inherited painless neuropathies arise due to genetic insults that either block the normal signaling of or destroy the sensory afferent neurons in the dorsal root ganglion (DRG) responsible for transducing noxious stimuli. Complete loss of these neurons leads to profound insensitivity to all sensory modalities including pain. Hereditary sensory and autonomic neuropathy type 2 (HSNAII) is a rare genetic neuropathy characterized by a progressive distal early onset sensory loss. This syndrome is caused by autosomal recessive mutations in the with-no-lysine protein kinase 1 (WNK1) serine-threonine kinase gene. Of interest, disease-associated mutations are found in the large exon, termed "HSN2," which encodes a 498 amino acid domain C-terminal to the kinase domain. These mutations lead to truncation of the HSN2-containing proteins through the addition of an early stop codon (nonsense mutation) leading to loss of the C-terminal domains of this large protein. The present study evaluates the transcripts, gene structure, and protein structure of HSN2-containing WNK1 splice variants in DRG and spinal cord in order to establish the basal expression patterns of WNK1 and HSN2-containing WNK1 splice variants using multiplex fluorescent situ hybridization. We hypothesized that these transcripts would be enriched in pain-sensing DRG neurons, and, potentially, that enrichment in nociceptive neurons was responsible for the painless phenotypes observed. However, our in-depth analyses revealed that the HSN2-WNK1 splice variants were ubiquitously expressed but were not enriched in tachykinin 1-expressing C-fiber neurons, a class of neurons with a highly nociceptive character. We subsequently identified other subpopulations of DRG neurons with higher levels of HSN2-WNK1 expression, including mechanosensory large fibers. These data are inconsistent with the hypothesis that this transcript is enriched in nociceptive fibers, and instead suggest it may be related to general axon maintenance, or that nociceptive fibers are more sensitive to the genetic insult. These findings clarify the molecular and cellular expression pattern of this painless neuropathy gene in human tissue.
Collapse
Affiliation(s)
- Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | - Jahandar Jahanipour
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | | | - Allison P Manalo
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Zhang H, Li A, Liu YF, Sun ZM, Jin BX, Lin JP, Yang Y, Yao YX. Spinal TAOK2 contributes to neuropathic pain via cGAS-STING activation in rats. iScience 2023; 26:107792. [PMID: 37720090 PMCID: PMC10502416 DOI: 10.1016/j.isci.2023.107792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 05/25/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
Thousand and one amino acid kinase 2 (TAOK2) is a member of the mammalian sterile 20 kinase family and is implicated in neurodevelopmental disorders; however, its role in neuropathic pain remains unknown. Here, we found that TAOK2 was enriched and activated after chronic constriction injury (CCI) in the rat spinal dorsal horn. Meanwhile, cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling was also activated with hyperalgesia. Silencing TAOK2 reversed hyperalgesia and suppressed the activation of cGAS-STING signaling induced by CCI, while pharmacological activation of TAOK2 induced pain hypersensitivity and upregulation of cGAS-STING signaling in naive rats. Furthermore, pharmacological inhibition or gene silencing of cGAS-STING signaling attenuated CCI-induced hyperalgesia. Taken together, these data demonstrate that the activation of spinal TAOK2 contributes to CCI-induced hyperalgesia via cGAS-STING signaling activation, providing new molecular targets for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Ang Li
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
- Department of Anesthesia, People’s Hospital of Guizhou Province, Guiyang, Guizhou 550025, China
| | - Yu-Fan Liu
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Zhong-Ming Sun
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Bing-Xin Jin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jia-Piao Lin
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yan Yang
- Department of Neurobiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
- Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yong-Xing Yao
- Department of Anesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
7
|
Fu X, Zhang Y, Zhang R. Regulatory role of PI3K/Akt/WNK1 signal pathway in mouse model of bone cancer pain. Sci Rep 2023; 13:14321. [PMID: 37652923 PMCID: PMC10471765 DOI: 10.1038/s41598-023-40182-w] [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: 04/29/2023] [Accepted: 08/06/2023] [Indexed: 09/02/2023] Open
Abstract
In the advanced stage of cancer, the pain caused by bone metastasis is unbearable, but the mechanism of bone cancer pain (BCP) is very complicated and remains unclear. In this study, we used 4T1 mouse breast cancer cells to establish a bone cancer pain model to study the mechanism of BCP. Then the paw withdrawal mechanical threshold (PWMT) and the hematoxylin-eosin staining were used to reflect the erosion of cancer cells on tibia tissue. We also determined the role of proinflammatory factors (TNF-α, IL-17, etc.) in BCP by the enzyme-linked immunosorbent assay in mouse serum. When GSK690693, a new Akt inhibitor, was given and the absence of intermediate signal dominated by Akt is found, pain may be relieved by blocking the transmission of pain signal and raising the PWMT. In addition, we also found that GSK690693 inhibited the phosphorylation of Akt protein, resulting in a significant decrease in with-nolysinekinases 1 (WNK1) expression in the spinal cord tissue. In the BCP model, we confirmed that GSK690693 has a relieving effect on BCP, which may play an analgesic effect through PI3K-WNK1 signal pathway. At the same time, there is a close relationship between inflammatory factors and PI3K-WNK1 signal pathway. The PI3K/Akt pathway in the dorsal horn of the mouse spinal cord activates the downstream WNK1 protein, which promotes the release of inflammatory cytokines, which leads to the formation of BCP in mice. Inhibition of Akt can reduce the levels of IL-17 and TNF-α, cut off the downstream WNK1 protein signal receiving pathway, increase the PWMT and relieve BCP in mice. To clarify the analgesic target of BCP, to provide reference and theoretical support for the clinical effective treatment of BCP and the development of new high-efficiency analgesics.
Collapse
Affiliation(s)
- Xiao Fu
- Department of Anesthesiology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
- Inner Mongolia Medical University, Hohhot, 010110, China
| | - Yanhong Zhang
- Department of Anesthesiology, Peking University Cancer Hospital Inner Mongolia Hospital/Cancer Hospital Affiliated to Inner Mongolia Medical University, Hohhot, 010020, China.
| | - Rui Zhang
- Department of Anesthesiology, Peking University Cancer Hospital Inner Mongolia Hospital/Cancer Hospital Affiliated to Inner Mongolia Medical University, Hohhot, 010020, China
| |
Collapse
|
8
|
Espinosa-Juárez JV, Chiquete E, Estañol B, Aceves JDJ. Optogenetic and Chemogenic Control of Pain Signaling: Molecular Markers. Int J Mol Sci 2023; 24:10220. [PMID: 37373365 DOI: 10.3390/ijms241210220] [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: 04/11/2023] [Revised: 06/05/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Pain is a complex experience that involves physical, emotional, and cognitive aspects. This review focuses specifically on the physiological processes underlying pain perception, with a particular emphasis on the various types of sensory neurons involved in transmitting pain signals to the central nervous system. Recent advances in techniques like optogenetics and chemogenetics have allowed researchers to selectively activate or inactivate specific neuronal circuits, offering a promising avenue for developing more effective pain management strategies. The article delves into the molecular targets of different types of sensory fibers such as channels, for example, TRPV1 in C-peptidergic fiber, TRPA1 in C-non-peptidergic receptors expressed differentially as MOR and DOR, and transcription factors, and their colocalization with the vesicular transporter of glutamate, which enable researchers to identify specific subtypes of neurons within the pain pathway and allows for selective transfection and expression of opsins to modulate their activity.
Collapse
Affiliation(s)
- Josue Vidal Espinosa-Juárez
- Escuela de Ciencias Químicas Sede Ocozocoautla, Universidad Autónoma de Chiapas, Ocozocoautla de Espinosa 29140, Mexico
| | - Erwin Chiquete
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Bruno Estañol
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - José de Jesús Aceves
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| |
Collapse
|
9
|
Pressey JC, de Saint-Rome M, Raveendran VA, Woodin MA. Chloride transporters controlling neuronal excitability. Physiol Rev 2023; 103:1095-1135. [PMID: 36302178 DOI: 10.1152/physrev.00025.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Synaptic inhibition plays a crucial role in regulating neuronal excitability, which is the foundation of nervous system function. This inhibition is largely mediated by the neurotransmitters GABA and glycine that activate Cl--permeable ion channels, which means that the strength of inhibition depends on the Cl- gradient across the membrane. In neurons, the Cl- gradient is primarily mediated by two secondarily active cation-chloride cotransporters (CCCs), NKCC1 and KCC2. CCC-mediated regulation of the neuronal Cl- gradient is critical for healthy brain function, as dysregulation of CCCs has emerged as a key mechanism underlying neurological disorders including epilepsy, neuropathic pain, and autism spectrum disorder. This review begins with an overview of neuronal chloride transporters before explaining the dependent relationship between these CCCs, Cl- regulation, and inhibitory synaptic transmission. We then discuss the evidence for how CCCs can be regulated, including by activity and their protein interactions, which underlie inhibitory synaptic plasticity. For readers who may be interested in conducting experiments on CCCs and neuronal excitability, we have included a section on techniques for estimating and recording intracellular Cl-, including their advantages and limitations. Although the focus of this review is on neurons, we also examine how Cl- is regulated in glial cells, which in turn regulate neuronal excitability through the tight relationship between this nonneuronal cell type and synapses. Finally, we discuss the relatively extensive and growing literature on how CCC-mediated neuronal excitability contributes to neurological disorders.
Collapse
Affiliation(s)
- Jessica C Pressey
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Miranda de Saint-Rome
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vineeth A Raveendran
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Melanie A Woodin
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Liedtke W. Long March Toward Safe and Effective Analgesia by Enhancing Gene Expression of Kcc2: First Steps Taken. Front Mol Neurosci 2022; 15:865600. [PMID: 35645734 PMCID: PMC9137411 DOI: 10.3389/fnmol.2022.865600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
Low intraneuronal chloride in spinal cord dorsal horn pain relay neurons is critical for physiologic transmission of primary pain afferents because low intraneuronal chloride dictates whether GABA-ergic and glycin-ergic neurotransmission is inhibitory. If the neuronal chloride elevates to pathologic levels, then spinal cord primary pain relay becomes leaky and exhibits the behavioral hallmarks of pathologic pain, namely hypersensitivity and allodynia. Low chloride in spinal cord dorsal horn neurons is maintained by proper gene expression of Kcc2 and sustained physiologic function of the KCC2 chloride extruding electroneutral transporter. Peripheral nerve injury and other forms of neural injury evoke greatly diminished Kcc2 gene expression and subsequent corruption of inhibitory neurotransmission in the spinal cord dorsal horn, thus causing derailment of the gate function for pain. Here I review key discoveries that have helped us understand these fundamentals, and focus on recent insights relating to the discovery of Kcc2 gene expression enhancing compounds via compound screens in neurons. One such study characterized the kinase inhibitor, kenpaullone, more in-depth, revealing its function as a robust and long-lasting analgesic in preclinical models of nerve injury and cancer bone pain, also elucidating its mechanism of action via GSK3β inhibition, diminishing delta-catenin phosphorylation, and facilitating its nuclear transfer and subsequent enhancement of Kcc2 gene expression by de-repressing Kaiso epigenetic transcriptional regulator. Future directions re Kcc2 gene expression enhancement are discussed, namely combination with other analgesics and analgesic methods, such as spinal cord stimulation and electroacupuncture, gene therapy, and leveraging Kcc2 gene expression-enhancing nanomaterials.
Collapse
|
11
|
Xiang Y, Dai J, Xu L, Li X, Jiang J, Xu J. Research progress in immune microenvironment regulation of muscle atrophy induced by peripheral nerve injury. Life Sci 2021; 287:120117. [PMID: 34740577 DOI: 10.1016/j.lfs.2021.120117] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/18/2021] [Accepted: 10/28/2021] [Indexed: 01/08/2023]
Abstract
Denervated skeletal muscular atrophy is primarily characterized by loss of muscle strength and mass and an unideal functional recovery of the muscle after extended denervation. This review emphasizes the interaction between the immune system and the denervated skeletal muscle. Immune cells such as neutrophils, macrophages and T-cells are activated and migrate to denervated muscle, where they release a high concentration of cytokines and chemokines. The migration of these immune cells, the transformation of different functional immune cell subtypes, and the cytokine network in the immune microenvironment may be involved in the regulatory process of muscle atrophy or repair. However, the exact mechanisms of the interaction between these immune cells and immune molecules in skeletal muscles are unclear. In this paper, the immune microenvironment regulation of muscle atrophy induced by peripheral nerve injury is reviewed.
Collapse
Affiliation(s)
- Yaoxian Xiang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai, China; Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China
| | - Junxi Dai
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai, China; Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China
| | - Lei Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai, China; Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China
| | - Xiaokang Li
- Natl Res Inst Child Hlth & Dev, Div Transplantat Immunol, Tokyo, Japan
| | - Junjian Jiang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai, China; Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China.
| | - Jianguang Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai, China; Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai, China; School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
12
|
Kipnis PA, Kadam SD. Novel Concepts for the Role of Chloride Cotransporters in Refractory Seizures. Aging Dis 2021; 12:1056-1069. [PMID: 34221549 PMCID: PMC8219493 DOI: 10.14336/ad.2021.0129] [Citation(s) in RCA: 3] [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: 12/12/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Epilepsy is associated with a multitude of acquired or genetic neurological disorders characterized by a predisposition to spontaneous recurrent seizures. An estimated 15 million patients worldwide have ongoing seizures despite optimal management and are classified as having refractory epilepsy. Early-life seizures like those caused by perinatal hypoxic ischemic encephalopathy (HIE) remain a clinical challenge because although transient, they are difficult to treat and associated with poor neurological outcomes. Pediatric epilepsy syndromes are consistently associated with intellectual disability and neurocognitive comorbidities. HIE and arterial ischemic stroke are the most common causes of seizures in term neonates and account for 7.5-20% of neonatal seizures. Standard first-line treatments such as phenobarbital (PB) and phenytoin fail to curb seizures in ~50% of neonates. In the long-term, HIE can result in hippocampal sclerosis and temporal lobe epilepsy (TLE), which is the most common adult epilepsy, ~30% of which is associated with refractory seizures. For patients with refractory TLE seizures, a viable option is the surgical resection of the epileptic foci. Novel insights gained from investigating the developmental role of Cl- cotransporter function have helped to elucidate some of the mechanisms underlying the emergence of refractory seizures in both HIE and TLE. KCC2 as the chief Cl- extruder in neurons is critical for enabling strong hyperpolarizing synaptic inhibition in the brain and has been implicated in the pathophysiology underlying both conditions. More recently, KCC2 function has become a novel therapeutic target to combat refractory seizures.
Collapse
Affiliation(s)
- Pavel A Kipnis
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
| | - Shilpa D Kadam
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
13
|
Ghosh S, Tourtellotte WG. The Complex Clinical and Genetic Landscape of Hereditary Peripheral Neuropathy. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 16:487-509. [PMID: 33497257 DOI: 10.1146/annurev-pathol-030320-100822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hereditary peripheral neuropathy (HPN) is a complex group of neurological disorders caused by mutations in genes expressed by neurons and Schwann cells. The inheritance of a single mutation or multiple mutations in several genes leads to disease phenotype. Patients exhibit symptoms during development, at an early age or later in adulthood. Most of the mechanistic understanding about these neuropathies comes from animal models and histopathological analyses of postmortem human tissues. Diagnosis is often very complex due to the heterogeneity and overlap in symptoms and the frequent overlap between various genes and different mutations they possess. Some symptoms in HPN are common through different subtypes such as axonal degeneration, demyelination, and loss of motor and sensory neurons, leading to similar physiologic abnormalities. Recent advances in gene-targeted therapies, genetic engineering, and next-generation sequencing have augmented our understanding of the underlying pathogenetic mechanisms of HPN.
Collapse
Affiliation(s)
- Soumitra Ghosh
- Department of Pathology and Laboratory Medicine, Neurology, and Neurological Surgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA;
| | - Warren G Tourtellotte
- Department of Pathology and Laboratory Medicine, Neurology, and Neurological Surgery, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA;
| |
Collapse
|
14
|
Josiah SS, Meor Azlan NF, Zhang J. Targeting the WNK-SPAK/OSR1 Pathway and Cation-Chloride Cotransporters for the Therapy of Stroke. Int J Mol Sci 2021; 22:1232. [PMID: 33513812 PMCID: PMC7865768 DOI: 10.3390/ijms22031232] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/05/2023] Open
Abstract
Stroke is one of the major culprits responsible for morbidity and mortality worldwide, and the currently available pharmacological strategies to combat this global disease are scanty. Cation-chloride cotransporters (CCCs) are expressed in several tissues (including neurons) and extensively contribute to the maintenance of numerous physiological functions including chloride homeostasis. Previous studies have implicated two CCCs, the Na+-K+-Cl- and K+-Cl- cotransporters (NKCCs and KCCs) in stroke episodes along with their upstream regulators, the with-no-lysine kinase (WNKs) family and STE20/SPS1-related proline/alanine rich kinase (SPAK) or oxidative stress response kinase (OSR1) via a signaling pathway. As the WNK-SPAK/OSR1 pathway reciprocally regulates NKCC and KCC, a growing body of evidence implicates over-activation and altered expression of NKCC1 in stroke pathology whilst stimulation of KCC3 during and even after a stroke event is neuroprotective. Both inhibition of NKCC1 and activation of KCC3 exert neuroprotection through reduction in intracellular chloride levels and thus could be a novel therapeutic strategy. Hence, this review summarizes the current understanding of functional regulations of the CCCs implicated in stroke with particular focus on NKCC1, KCC3, and WNK-SPAK/OSR1 signaling and discusses the current and potential pharmacological treatments for stroke.
Collapse
Affiliation(s)
| | | | - Jinwei Zhang
- Hatherly Laboratories, Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Exeter EX4 4PS, UK; (S.S.J.); (N.F.M.A.)
| |
Collapse
|
15
|
Li J, Luan F, Song J, Dong J, Shang M. Clinical Efficacy of Controlled-Release Morphine Tablets Combined with Celecoxib in Pain Management and the Effects on WNK1 Expression. Clinics (Sao Paulo) 2021; 76:e1907. [PMID: 33503173 PMCID: PMC7798123 DOI: 10.6061/clinics/2021/e1907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/29/2020] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES This study was designed to evaluate the clinical efficacy of controlled-release morphine tablets combined with celecoxib in relieving osteocarcinoma-related pain and the effects of the combination on WNK1 expression. METHODS A total of 110 patients with osteocarcinoma-related pain were selected and divided into two groups based on the treatment administered, including the control group (treated with controlled-release morphine tablets alone) and the study group (treated with a combination of controlled-release morphine tablets and celecoxib). We compared the treatment efficacy, pain level (visual analog scale (VAS)), time of onset of breakthrough pain (BTP), dose of morphine, incidence of adverse events, quality of life (QOL) score, and With-no-lysine 1 (WNK1) expression in the peripheral blood (PB) as determined with qRT-PCR before and after treatment, of the two groups. RESULTS The total effective rate of the study group was higher than that of the control group, while the VAS score, time of onset of BTP, dose of morphine, incidence of adverse events, QOL score, and relative WNK1 expression in the PB were lower than those of the control group (p<0.05). CONCLUSION Combination treatment with controlled-release morphine tablets and celecoxib can be extensively used in the clinical setting because it effectively improves the symptoms, QOL score, and adverse effects in patients with osteocarcinoma-related pain.
Collapse
Affiliation(s)
- Jian Li
- Department of Joint Surgery, the Fourth People's Hospital of Jinan, Jinan, China
| | - Fanghai Luan
- Department of Orthopedic Surgery, the Fourth People's Hospital of Jinan, Jinan, China
| | - Jiangfeng Song
- Department of Orthopedic, Ju County People's Hospital, Rizhao, China
| | - Jianhua Dong
- Department of Orthopedic, Ju County People's Hospital, Rizhao, China
| | - Mingfu Shang
- Department of Spinal Cord Repairing, 960 Hospital of the Joint Logistics Support Force of PLA, Jinan, China
- *Corresponding author. E-mail:
| |
Collapse
|
16
|
Murillo-de-Ozores AR, Chávez-Canales M, de los Heros P, Gamba G, Castañeda-Bueno M. Physiological Processes Modulated by the Chloride-Sensitive WNK-SPAK/OSR1 Kinase Signaling Pathway and the Cation-Coupled Chloride Cotransporters. Front Physiol 2020; 11:585907. [PMID: 33192599 PMCID: PMC7606576 DOI: 10.3389/fphys.2020.585907] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
The role of Cl- as an intracellular signaling ion has been increasingly recognized in recent years. One of the currently best described roles of Cl- in signaling is the modulation of the With-No-Lysine (K) (WNK) - STE20-Proline Alanine rich Kinase (SPAK)/Oxidative Stress Responsive Kinase 1 (OSR1) - Cation-Coupled Cl- Cotransporters (CCCs) cascade. Binding of a Cl- anion to the active site of WNK kinases directly modulates their activity, promoting their inhibition. WNK activation due to Cl- release from the binding site leads to phosphorylation and activation of SPAK/OSR1, which in turn phosphorylate the CCCs. Phosphorylation by WNKs-SPAK/OSR1 of the Na+-driven CCCs (mediating ions influx) promote their activation, whereas that of the K+-driven CCCs (mediating ions efflux) promote their inhibition. This results in net Cl- influx and feedback inhibition of WNK kinases. A wide variety of alterations to this pathway have been recognized as the cause of several human diseases, with manifestations in different systems. The understanding of WNK kinases as Cl- sensitive proteins has allowed us to better understand the mechanistic details of regulatory processes involved in diverse physiological phenomena that are reviewed here. These include cell volume regulation, potassium sensing and intracellular signaling in the renal distal convoluted tubule, and regulation of the neuronal response to the neurotransmitter GABA.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Chávez-Canales
- Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paola de los Heros
- Unidad de Investigación UNAM-INC, Research Division, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
17
|
Hughes DI, Todd AJ. Central Nervous System Targets: Inhibitory Interneurons in the Spinal Cord. Neurotherapeutics 2020; 17:874-885. [PMID: 33029722 PMCID: PMC7641291 DOI: 10.1007/s13311-020-00936-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Pain is a percept of critical importance to our daily survival. In most cases, it serves both an adaptive function by helping us respond appropriately in a potentially hostile environment and also a protective role by alerting us to tissue damage. Normally, it is evoked by the activation of peripheral nociceptive nerve endings and the subsequent relay of information to distinct cortical and sub-cortical regions, but under pathological conditions that result in chronic pain, it can become spontaneous. Given that one in three chronic pain patients do not respond to the treatments currently available, the need for more effective analgesics is evident. Two principal obstacles to the development of novel analgesic therapies are our limited understanding of how neuronal circuits that comprise these pain pathways transmit and modulate sensory information under normal circumstances and how these circuits change under pathological conditions leading to chronic pain states. In this review, we focus on the role of inhibitory interneurons in setting pain thresholds and, in particular, how disinhibition in the spinal dorsal horn can lead to aberrant sensory processing associated with chronic pain states.
Collapse
Affiliation(s)
- David I Hughes
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland.
| | - Andrew J Todd
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland
| |
Collapse
|
18
|
Duy PQ, He M, He Z, Kahle KT. Preclinical insights into therapeutic targeting of KCC2 for disorders of neuronal hyperexcitability. Expert Opin Ther Targets 2020; 24:629-637. [PMID: 32336175 DOI: 10.1080/14728222.2020.1762174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Epilepsy is a common neurological disorder of neuronal hyperexcitability that begets recurrent and unprovoked seizures. The lack of a truly satisfactory pharmacotherapy for epilepsy highlights the clinical urgency for the discovery of new drug targets. To that end, targeting the electroneutral K+/Cl- cotransporter KCC2 has emerged as a novel therapeutic strategy for the treatment of epilepsy. AREAS COVERED We summarize the roles of KCC2 in the maintenance of synaptic inhibition and the evidence linking KCC2 dysfunction to epileptogenesis. We also discuss preclinical proof-of-principle studies that demonstrate that augmentation of KCC2 function can reduce seizure activity. Moreover, potential strategies to modulate KCC2 activity for therapeutic benefit are highlighted. EXPERT OPINION Although KCC2 is a promising drug target, questions remain before clinical translation. It is unclear whether increasing KCC2 activity can reverse epileptogenesis, the ultimate curative goal for epilepsy therapy that extends beyond seizure reduction. Furthermore, the potential adverse effects associated with increased KCC2 function have not been studied. Continued investigations into the neurobiology of KCC2 will help to translate promising preclinical insights into viable therapeutic avenues that leverage fundamental properties of KCC2 to treat medically intractable epilepsy and other disorders of failed synaptic inhibition with attendant neuronal hyperexcitability.
Collapse
Affiliation(s)
- Phan Q Duy
- Department of Neurosurgery, Yale University School of Medicine , New Haven, CT, USA.,Medical Scientist Training Program, Yale University School of Medicine , New Haven, CT, USA
| | - Miao He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School , Boston, MA, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School , Boston, MA, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale University School of Medicine , New Haven, CT, USA.,Department of Genetics, Yale University School of Medicine , New Haven, CT, USA.,Departments of Pediatrics and Cellular & Molecular Physiology, Yale University School of Medicine , New Haven, CT, USA.,Yale-Rockefeller NIH Centers for Mendelian Genomics, Yale University , New Haven, CT, USA.,Yale Stem Cell Center, Yale School of Medicine , New Haven, CT, USA
| |
Collapse
|
19
|
Miller RE, Tran PB, Ishihara S, Syx D, Ren D, Miller RJ, Valdes AM, Malfait AM. Microarray analyses of the dorsal root ganglia support a role for innate neuro-immune pathways in persistent pain in experimental osteoarthritis. Osteoarthritis Cartilage 2020; 28:581-592. [PMID: 31982564 PMCID: PMC7214125 DOI: 10.1016/j.joca.2020.01.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Following destabilization of the medial meniscus (DMM), mice develop experimental osteoarthritis (OA) and associated pain behaviors that are dependent on the stage of disease. We aimed to describe changes in gene expression in knee-innervating dorsal root ganglia (DRG) after surgery, in order to identify molecular pathways associated with three pre-defined pain phenotypes: "post-surgical pain", "early-stage OA pain", and "persistent OA pain". DESIGN We performed DMM or sham surgery in 10-week old male C57BL/6 mice and harvested L3-L5 DRG 4, 8, and 16 weeks after surgery or from age-matched naïve mice (n = 3/group). RNA was extracted and an Affymetrix Mouse Transcriptome Array 1.0 was performed. Three pain phenotypes were defined: "post-surgical pain" (sham and DMM 4-week vs 14-week old naïve), "early OA pain" (DMM 4-week vs sham 4-week), and "persistent OA pain" (DMM 8- and 16-week vs naïve and sham 8- and 16-week). 'Top hit' genes were defined as P < 0.001. Pathway analysis (Ingenuity Pathway Analysis) was conducted using differentially expressed genes defined as P < 0.05. In addition, we performed qPCR for Ngf and immunohistochemistry for F4/80+ macrophages in the DRG. RESULTS For each phenotype, top hit genes identified a small number of differentially expressed genes, some of which have been previously associated with pain (7/67 for "post-surgical pain"; 2/14 for "early OA pain"; 8/37 for "persistent OA pain"). Overlap between groups was limited, with 8 genes differentially regulated (P < 0.05) in all three phenotypes. Pathway analysis showed that in the persistent OA pain phase many of the functions of differentially regulated genes are related to immune cell recruitment and activation. Genes previously linked to OA pain (CX3CL1, CCL2, TLR1, and NGF) were upregulated in this phenotype and contributed to activation of the neuroinflammation canonical pathway. In separate sets of mice, we confirmed that Ngf was elevated in the DRG 8 weeks after DMM (P = 0.03), and numbers of F4/80+ macrophages were increased 16 weeks after DMM (P = 0.002 vs Sham). CONCLUSION These transcriptomics findings support the idea that distinct molecular pathways discriminate early from persistent OA pain. Pathway analysis suggests neuroimmune interactions in the DRG contribute to initiation and maintenance of pain in OA.
Collapse
Affiliation(s)
- Rachel E. Miller
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, Chicago IL
| | - Phuong B. Tran
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, Chicago IL
| | - Shingo Ishihara
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, Chicago IL
| | - Delfien Syx
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, Chicago IL
| | | | - Ana M. Valdes
- Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham UK
| | - Anne-Marie Malfait
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, Chicago IL
| |
Collapse
|
20
|
Beverungen H, Klaszky SC, Klaszky M, Côté MP. Rehabilitation Decreases Spasticity by Restoring Chloride Homeostasis through the Brain-Derived Neurotrophic Factor-KCC2 Pathway after Spinal Cord Injury. J Neurotrauma 2020; 37:846-859. [PMID: 31578924 PMCID: PMC7071070 DOI: 10.1089/neu.2019.6526] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Activity-based therapy is routinely integrated in rehabilitation programs to facilitate functional recovery after spinal cord injury (SCI). Among its beneficial effects is a reduction of hyperreflexia and spasticity, which affects ∼75% of the SCI population. Unlike current anti-spastic pharmacological treatments, rehabilitation attenuates spastic symptoms without causing an active depression in spinal excitability, thus avoiding further interference with motor recovery. Understanding how activity-based therapies contribute to decrease spasticity is critical to identifying new pharmacological targets and to optimize rehabilitation programs. It was recently demonstrated that a decrease in the expression of KCC2, a neuronal Cl- extruder, contributes to the development spasticity in SCI rats. Although exercise can decrease spinal hyperexcitability and increase KCC2 expression on lumbar motoneurons after SCI, a causal effect remains to be established. Activity-dependent processes include an increase in brain-derived neurotrophic factor (BDNF) expression. Interestingly, BDNF is a regulator of KCC2 but also a potent modulator of spinal excitability. Therefore, we hypothesized that after SCI, the activity-dependent increase in KCC2 expression: 1) functionally contributes to reduce hyperreflexia, and 2) is regulated by BDNF. SCI rats chronically received VU0240551 (KCC2 blocker) or TrkB-IgG (BDNF scavenger) during the daily rehabilitation sessions and the frequency-dependent depression of the H-reflex, a monitor of hyperreflexia, was recorded 4 weeks post-injury. Our results suggest that the activity-dependent increase in KCC2 functionally contributes to H-reflex recovery and critically depends on BDNF activity. This study provides a new perspective in understanding how exercise impacts hyperreflexia by identifying the biological basis of the recovery of function.
Collapse
Affiliation(s)
- Henrike Beverungen
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Samantha Choyke Klaszky
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Michael Klaszky
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Marie-Pascale Côté
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Gao JL, Peng K, Shen MW, Hou YH, Qian XB, Meng XW, Ji FH, Wang LN, Yang JP. Suppression of WNK1-SPAK/OSR1 Attenuates Bone Cancer Pain by Regulating NKCC1 and KCC2. THE JOURNAL OF PAIN 2019; 20:1416-1428. [PMID: 31085334 DOI: 10.1016/j.jpain.2019.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/15/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023]
Abstract
Our preliminary experiment indicated the activation of with-nolysine kinases 1 (WNK1) in bone cancer pain (BCP) rats. This study aimed to investigate the underlying mechanisms via which WNK1 contributed to BCP. A rat model of BCP was induced by Walker-256 tumor cell implantation. WNK1 expression and distribution in the lumbar spinal cord dorsal horn and dorsal root ganglion were examined. SPS1-related proline/alanine-rich kinase (SPAK), oxidative stress-responsive kinase 1 (OSR1), sodium-potassium-chloride cotransporter 1 (NKCC1), and potassium-chloride cotransporter 2 (KCC2) expression were assessed. Pain behaviors including mechanical allodynia and movement-evoked pain were measured. BCP rats exhibited significant mechanical allodynia, with increased WNK1 expression in the dorsal horn and dorsal root ganglion neurons, elevated SPAK/OSR1 and NKCC1 expression in the dorsal root ganglion, and decreased KCC2 expression in the dorsal horn. WNK1 knock-down by small interfering alleviated mechanical allodynia and movement-evoked pain, inhibited WNK1-SPAK/OSR1-NKCC1 activities, and restored KCC2 expression. In addition, closantel (a WNK1-SPAK/OSR1 inhibitor) improved pain behaviors, downregulated SPAK/OSR1 and NKCC1 expression, and upregulated KCC2 expression in BCP rats. Activation of WNK1-SPAK/OSR1 signaling contributed to BCP in rats by modulating NKCC1 and KCC2 expression. Therefore, suppression of WNK1-SPAK/OSR1 may serve as a potential target for BCP therapy. PERSPECTIVE: Our findings demonstrated that the WNK1-SPAK/OSR1 signaling contributed to BCP in rats via regulating NKCC1 and KCC2. Suppressing this pathway reduced pain behaviors. Based on these findings, the WNK1-SPAK/OSR1 signaling may be a potential target for BCP therapy.
Collapse
Affiliation(s)
- Jian-Ling Gao
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Meng-Wei Shen
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Department of Anesthesiology, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, China
| | - Yong-Heng Hou
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Bo Qian
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Wen Meng
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fu-Hai Ji
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li-Na Wang
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jian-Ping Yang
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
22
|
Duy PQ, David WB, Kahle KT. Identification of KCC2 Mutations in Human Epilepsy Suggests Strategies for Therapeutic Transporter Modulation. Front Cell Neurosci 2019; 13:515. [PMID: 31803025 PMCID: PMC6873151 DOI: 10.3389/fncel.2019.00515] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/01/2019] [Indexed: 11/28/2022] Open
Abstract
Epilepsy is a common neurological disorder characterized by recurrent and unprovoked seizures thought to arise from impaired balance between neuronal excitation and inhibition. Our understanding of the neurophysiological mechanisms that render the brain epileptogenic remains incomplete, reflected by the lack of satisfactory treatments that can effectively prevent epileptic seizures without significant drug-related adverse effects. Type 2 K+-Cl− cotransporter (KCC2), encoded by SLC12A5, is important for chloride homeostasis and neuronal excitability. KCC2 dysfunction attenuates Cl− extrusion and impairs GABAergic inhibition, and can lead to neuronal hyperexcitability. Converging lines of evidence from human genetics have secured the link between KCC2 dysfunction and the development of epilepsy. Here, we review KCC2 mutations in human epilepsy and discuss potential therapeutic strategies based on the functional impact of these mutations. We suggest that a strategy of augmenting KCC2 activity by antagonizing its critical inhibitory phosphorylation sites may be a particularly efficacious method of facilitating Cl− extrusion and restoring GABA inhibition to treat medication-refractory epilepsy and other seizure disorders.
Collapse
Affiliation(s)
- Phan Q Duy
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States.,Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, United States
| | - Wyatt B David
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States.,Department of Genetics, Yale University School of Medicine, New Haven, CT, United States.,Departments of Pediatrics and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States.,Yale-Rockefeller NIH Centers for Mendelian Genomics, Yale University, New Haven, CT, United States.,Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
23
|
Gradwell MA, Callister RJ, Graham BA. Reviewing the case for compromised spinal inhibition in neuropathic pain. J Neural Transm (Vienna) 2019; 127:481-503. [PMID: 31641856 DOI: 10.1007/s00702-019-02090-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 09/30/2019] [Indexed: 12/20/2022]
Abstract
A striking and debilitating property of the nervous system is that damage to this tissue can cause chronic intractable pain, which persists long after resolution of the initial insult. This neuropathic form of pain can arise from trauma to peripheral nerves, the spinal cord, or brain. It can also result from neuropathies associated with disease states such as diabetes, human immunodeficiency virus/AIDS, herpes, multiple sclerosis, cancer, and chemotherapy. Regardless of the origin, treatments for neuropathic pain remain inadequate. This continues to drive research into the underlying mechanisms. While the literature shows that dysfunction in numerous loci throughout the CNS can contribute to chronic pain, the spinal cord and in particular inhibitory signalling in this region have remained major research areas. This review focuses on local spinal inhibition provided by dorsal horn interneurons, and how such inhibition is disrupted during the development and maintenance of neuropathic pain.
Collapse
Affiliation(s)
- M A Gradwell
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW, Australia
| | - R J Callister
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW, Australia
| | - B A Graham
- School of Biomedical Sciences and Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW, Australia.
| |
Collapse
|
24
|
Watanabe M, Zhang J, Mansuri MS, Duan J, Karimy JK, Delpire E, Alper SL, Lifton RP, Fukuda A, Kahle KT. Developmentally regulated KCC2 phosphorylation is essential for dynamic GABA-mediated inhibition and survival. Sci Signal 2019; 12:eaaw9315. [PMID: 31615901 DOI: 10.1126/scisignal.aaw9315.(#,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2024]
Abstract
Despite its importance for γ-aminobutyric acid (GABA) inhibition and involvement in neurodevelopmental disease, the regulatory mechanisms of the K+/Cl- cotransporter KCC2 (encoded by SLC12A5) during maturation of the central nervous system (CNS) are not entirely understood. Here, we applied quantitative phosphoproteomics to systematically map sites of KCC2 phosphorylation during CNS development in the mouse. KCC2 phosphorylation at Thr906 and Thr1007, which inhibits KCC2 activity, underwent dephosphorylation in parallel with the GABA excitatory-inhibitory sequence in vivo. Knockin mice expressing the homozygous phosphomimetic KCC2 mutations T906E/T1007E (Kcc2E/E ), which prevented the normal developmentally regulated dephosphorylation of these sites, exhibited early postnatal death from respiratory arrest and a marked absence of cervical spinal neuron respiratory discharges. Kcc2E/E mice also displayed disrupted lumbar spinal neuron locomotor rhythmogenesis and touch-evoked status epilepticus associated with markedly impaired KCC2-dependent Cl- extrusion. These data identify a previously unknown phosphorylation-dependent KCC2 regulatory mechanism during CNS development that is essential for dynamic GABA-mediated inhibition and survival.
Collapse
Affiliation(s)
- Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - M Shahid Mansuri
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jingjing Duan
- Human Aging Research Institute, School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Seth L Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
- The Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan.
- Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
25
|
Watanabe M, Zhang J, Mansuri MS, Duan J, Karimy JK, Delpire E, Alper SL, Lifton RP, Fukuda A, Kahle KT. Developmentally regulated KCC2 phosphorylation is essential for dynamic GABA-mediated inhibition and survival. Sci Signal 2019; 12:eaaw9315. [PMID: 31615901 PMCID: PMC7219477 DOI: 10.1126/scisignal.aaw9315] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Despite its importance for γ-aminobutyric acid (GABA) inhibition and involvement in neurodevelopmental disease, the regulatory mechanisms of the K+/Cl- cotransporter KCC2 (encoded by SLC12A5) during maturation of the central nervous system (CNS) are not entirely understood. Here, we applied quantitative phosphoproteomics to systematically map sites of KCC2 phosphorylation during CNS development in the mouse. KCC2 phosphorylation at Thr906 and Thr1007, which inhibits KCC2 activity, underwent dephosphorylation in parallel with the GABA excitatory-inhibitory sequence in vivo. Knockin mice expressing the homozygous phosphomimetic KCC2 mutations T906E/T1007E (Kcc2E/E ), which prevented the normal developmentally regulated dephosphorylation of these sites, exhibited early postnatal death from respiratory arrest and a marked absence of cervical spinal neuron respiratory discharges. Kcc2E/E mice also displayed disrupted lumbar spinal neuron locomotor rhythmogenesis and touch-evoked status epilepticus associated with markedly impaired KCC2-dependent Cl- extrusion. These data identify a previously unknown phosphorylation-dependent KCC2 regulatory mechanism during CNS development that is essential for dynamic GABA-mediated inhibition and survival.
Collapse
Affiliation(s)
- Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - M Shahid Mansuri
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jingjing Duan
- Human Aging Research Institute, School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Seth L Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
- The Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan.
- Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
26
|
Goutierre M, Al Awabdh S, Donneger F, François E, Gomez-Dominguez D, Irinopoulou T, Menendez de la Prida L, Poncer JC. KCC2 Regulates Neuronal Excitability and Hippocampal Activity via Interaction with Task-3 Channels. Cell Rep 2019; 28:91-103.e7. [PMID: 31269453 DOI: 10.1016/j.celrep.2019.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/18/2019] [Accepted: 05/30/2019] [Indexed: 10/26/2022] Open
Abstract
KCC2 regulates neuronal transmembrane chloride gradients and thereby controls GABA signaling in the brain. KCC2 downregulation is observed in numerous neurological and psychiatric disorders. Paradoxical, excitatory GABA signaling is usually assumed to contribute to abnormal network activity underlying the pathology. We tested this hypothesis and explored the functional impact of chronic KCC2 downregulation in the rat dentate gyrus. Although the reversal potential of GABAA receptor currents is depolarized in KCC2 knockdown neurons, this shift is compensated by depolarization of the resting membrane potential. This reflects downregulation of leak potassium currents. We show KCC2 interacts with Task-3 (KCNK9) channels and is required for their membrane expression. Increased neuronal excitability upon KCC2 suppression altered dentate gyrus rhythmogenesis, which could be normalized by chemogenetic hyperpolarization. Our data reveal KCC2 downregulation engages complex synaptic and cellular alterations beyond GABA signaling that perturb network activity thus offering additional targets for therapeutic intervention.
Collapse
Affiliation(s)
- Marie Goutierre
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Sana Al Awabdh
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Florian Donneger
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Emeline François
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Daniel Gomez-Dominguez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
| | - Theano Irinopoulou
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | | | - Jean Christophe Poncer
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France.
| |
Collapse
|
27
|
Huang H, Song S, Banerjee S, Jiang T, Zhang J, Kahle KT, Sun D, Zhang Z. The WNK-SPAK/OSR1 Kinases and the Cation-Chloride Cotransporters as Therapeutic Targets for Neurological Diseases. Aging Dis 2019; 10:626-636. [PMID: 31165006 PMCID: PMC6538211 DOI: 10.14336/ad.2018.0928] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 09/28/2018] [Indexed: 02/05/2023] Open
Abstract
In recent years, cation-chloride cotransporters (CCCs) have drawn attention in the medical neuroscience research. CCCs include the family of Na+-coupled Cl- importers (NCC, NKCC1, and NKCC2), K+-coupled Cl- exporters (KCCs), and possibly polyamine transporters (CCC9) and CCC interacting protein (CIP1). For decades, CCCs have been the targets of several commonly used diuretic drugs, including hydrochlorothiazide, furosemide, and bumetanide. Genetic mutations of NCC and NKCC2 cause congenital renal tubular disorders and lead to renal salt-losing hypotension, secondary hyperreninemia, and hypokalemic metabolic alkalosis. New studies reveal that CCCs along with their regulatory WNK (Kinase with no lysine (K)), and SPAK (Ste20-related proline-alanine-rich kinase)/OSR1(oxidative stress-responsive kinase-1) are essential for regulating cell volume and maintaining ionic homeostasis in the nervous system, especially roles of the WNK-SPAK-NKCC1 signaling pathway in ischemic brain injury and hypersecretion of cerebrospinal fluid in post-hemorrhagic hydrocephalus. In addition, disruption of Cl- exporter KCC2 has an effect on synaptic inhibition, which may be involved in developing pain, epilepsy, and possibly some neuropsychiatric disorders. Interference with KCC3 leads to peripheral nervous system neuropathy as well as axon and nerve fiber swelling and psychosis. The WNK-SPAK/OSR1-CCCs complex emerges as therapeutic targets for multiple neurological diseases. This review will highlight these new findings.
Collapse
Affiliation(s)
- Huachen Huang
- Department of Neurology, The First Affiliate Hospital, Harbin Medical University, Harbin, Heilongjiang, China.
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Suneel Banerjee
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Tong Jiang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, EX4 4PS, UK.
| | - Kristopher T. Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, USA.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pittsburgh, PA, USA.
- Correspondence should be addressed to: Dr. Dandan Sun, Department of Neurology, University of Pittsburgh, Pittsburgh, USA. . Dr. Zhongling Zhang, The First Affiliated Hospital, Harbin Medical University, China.
| | - Zhongling Zhang
- Department of Neurology, The First Affiliate Hospital, Harbin Medical University, Harbin, Heilongjiang, China.
- Correspondence should be addressed to: Dr. Dandan Sun, Department of Neurology, University of Pittsburgh, Pittsburgh, USA. . Dr. Zhongling Zhang, The First Affiliated Hospital, Harbin Medical University, China.
| |
Collapse
|
28
|
Inquimbert P, Moll M, Latremoliere A, Tong CK, Whang J, Sheehan GF, Smith BM, Korb E, Athié MCP, Babaniyi O, Ghasemlou N, Yanagawa Y, Allis CD, Hof PR, Scholz J. NMDA Receptor Activation Underlies the Loss of Spinal Dorsal Horn Neurons and the Transition to Persistent Pain after Peripheral Nerve Injury. Cell Rep 2018; 23:2678-2689. [PMID: 29847798 PMCID: PMC6276118 DOI: 10.1016/j.celrep.2018.04.107] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/12/2018] [Accepted: 04/26/2018] [Indexed: 12/31/2022] Open
Abstract
Peripheral nerve lesions provoke apoptosis in the dorsal horn of the spinal cord. The cause of cell death, the involvement of neurons, and the relevance for the processing of somatosensory information are controversial. Here, we demonstrate in a mouse model of sciatic nerve injury that glutamate-induced neurodegeneration and loss of γ-aminobutyric acid (GABA)ergic interneurons in the superficial dorsal horn promote the transition from acute to chronic neuropathic pain. Conditional deletion of Grin1, the essential subunit of N-methyl-d-aspartate-type glutamate receptors (NMDARs), protects dorsal horn neurons from excitotoxicity and preserves GABAergic inhibition. Mice deficient in functional NMDARs exhibit normal nociceptive responses and acute pain after nerve injury, but this initial increase in pain sensitivity is reversible. Eliminating NMDARs fully prevents persistent pain-like behavior. Reduced pain in mice lacking proapoptotic Bax confirmed the significance of neurodegeneration. We conclude that NMDAR-mediated neuron death contributes to the development of chronic neuropathic pain.
Collapse
Affiliation(s)
- Perrine Inquimbert
- Centre National de la Recherche Scientifique, UPR 3212, Institut des Neurosciences Cellulaires et Intégratives and Université de Strasbourg, 67084 Strasbourg, France; F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Moll
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA; Institute of Pharmacology, Heidelberg University, 69120 Heidelberg, Germany
| | - Alban Latremoliere
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Chi-Kun Tong
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - John Whang
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Gregory F Sheehan
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Brendan M Smith
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Erica Korb
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY 10065, USA
| | - Maria C P Athié
- Department of Structural and Functional Biology, State University of Campinas, Campinas, SP 13083-865, Brazil
| | - Olusegun Babaniyi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Nader Ghasemlou
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - C David Allis
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY 10065, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joachim Scholz
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
29
|
Sato A, Shibuya H. Glycogen synthase kinase 3ß functions as a positive effector in the WNK signaling pathway. PLoS One 2018; 13:e0193204. [PMID: 29494638 PMCID: PMC5832235 DOI: 10.1371/journal.pone.0193204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/02/2018] [Indexed: 12/21/2022] Open
Abstract
The with no lysine (WNK) protein kinase family is conserved among many species. Some mutations in human WNK gene are associated with pseudohypoaldosteronism type II, a form of hypertension, and hereditary sensory and autonomic neuropathy type 2A. In kidney, WNK regulates the activity of STE20/SPS1-related, proline alanine-rich kinase and/or oxidative-stress responsive 1, which in turn regulate ion co-transporters. The misregulation of this pathway is involved in the pathogenesis of pseudohypoaldosteronism type II. In the neural system, WNK is involved in the specification of the cholinergic neuron, but the pathogenesis of hereditary sensory and autonomic neuropathy type 2A is still unknown. To better understand the WNK pathway, we isolated WNK-associated genes using Drosophila. We identified Glycogen synthase kinase 3ß (GSK3ß)/Shaggy (Sgg) as a candidate gene that was shown to interact with the WNK signaling pathway in both Drosophila and mammalian cells. Furthermore, GSK3ß was involved in neural specification downstream of WNK. These results suggest that GSK3ß/Sgg functions as a positive effector in the WNK signaling pathway.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Molecular Cell Biology and Joint Usage/Research Center for Intractable Diseases, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Shibuya
- Department of Molecular Cell Biology and Joint Usage/Research Center for Intractable Diseases, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
30
|
Woolf CJ. Pain amplification-A perspective on the how, why, when, and where of central sensitization. ACTA ACUST UNITED AC 2018. [DOI: 10.1111/jabr.12124] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Clifford J. Woolf
- FM Kirby Neurobiology Center; Boston Children's Hospital; Boston MA USA
- Department of Neurobiology; Harvard Medical School; Boston MA USA
| |
Collapse
|
31
|
Annecchino LA, Schultz SR. Progress in automating patch clamp cellular physiology. Brain Neurosci Adv 2018; 2:2398212818776561. [PMID: 32166142 PMCID: PMC7058203 DOI: 10.1177/2398212818776561] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/19/2018] [Indexed: 12/30/2022] Open
Abstract
Patch clamp electrophysiology has transformed research in the life sciences over the last few decades. Since their inception, automatic patch clamp platforms have evolved considerably, demonstrating the capability to address both voltage- and ligand-gated channels, and showing the potential to play a pivotal role in drug discovery and biomedical research. Unfortunately, the cell suspension assays to which early systems were limited cannot recreate biologically relevant cellular environments, or capture higher order aspects of synaptic physiology and network dynamics. In vivo patch clamp electrophysiology has the potential to yield more biologically complex information and be especially useful in reverse engineering the molecular and cellular mechanisms of single-cell and network neuronal computation, while capturing important aspects of human disease mechanisms and possible therapeutic strategies. Unfortunately, it is a difficult procedure with a steep learning curve, which has restricted dissemination of the technique. Luckily, in vivo patch clamp electrophysiology seems particularly amenable to robotic automation. In this review, we document the development of automated patch clamp technology, from early systems based on multi-well plates through to automated planar-array platforms, and modern robotic platforms capable of performing two-photon targeted whole-cell electrophysiological recordings in vivo.
Collapse
Affiliation(s)
- Luca A. Annecchino
- Centre for Neurotechnology and Department of Bioengineering, Imperial College London, London, UK
| | - Simon R. Schultz
- Centre for Neurotechnology and Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
32
|
Heubl M, Zhang J, Pressey JC, Al Awabdh S, Renner M, Gomez-Castro F, Moutkine I, Eugène E, Russeau M, Kahle KT, Poncer JC, Lévi S. GABA A receptor dependent synaptic inhibition rapidly tunes KCC2 activity via the Cl --sensitive WNK1 kinase. Nat Commun 2017; 8:1776. [PMID: 29176664 PMCID: PMC5701213 DOI: 10.1038/s41467-017-01749-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/13/2017] [Indexed: 02/08/2023] Open
Abstract
The K+-Cl- co-transporter KCC2 (SLC12A5) tunes the efficacy of GABAA receptor-mediated transmission by regulating the intraneuronal chloride concentration [Cl-]i. KCC2 undergoes activity-dependent regulation in both physiological and pathological conditions. The regulation of KCC2 by synaptic excitation is well documented; however, whether the transporter is regulated by synaptic inhibition is unknown. Here we report a mechanism of KCC2 regulation by GABAA receptor (GABAAR)-mediated transmission in mature hippocampal neurons. Enhancing GABAAR-mediated inhibition confines KCC2 to the plasma membrane, while antagonizing inhibition reduces KCC2 surface expression by increasing the lateral diffusion and endocytosis of the transporter. This mechanism utilizes Cl- as an intracellular secondary messenger and is dependent on phosphorylation of KCC2 at threonines 906 and 1007 by the Cl--sensing kinase WNK1. We propose this mechanism contributes to the homeostasis of synaptic inhibition by rapidly adjusting neuronal [Cl-]i to GABAAR activity.
Collapse
Affiliation(s)
- Martin Heubl
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Jinwei Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, Scotland
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, EX4 4PS, UK
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, NIH-Yale Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Jessica C Pressey
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Sana Al Awabdh
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Marianne Renner
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Ferran Gomez-Castro
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Imane Moutkine
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Emmanuel Eugène
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Marion Russeau
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, NIH-Yale Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Jean Christophe Poncer
- Inserm UMR-S 839, 75005, Paris, France
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France
- Institut du Fer à Moulin, 75005, Paris, France
| | - Sabine Lévi
- Inserm UMR-S 839, 75005, Paris, France.
- Université Pierre & Marie Curie, Sorbonne Universités, 75005, Paris, France.
- Institut du Fer à Moulin, 75005, Paris, France.
| |
Collapse
|
33
|
Bell DC, Dallas ML. Using automated patch clamp electrophysiology platforms in pain-related ion channel research: insights from industry and academia. Br J Pharmacol 2017. [PMID: 28622411 DOI: 10.1111/bph.13916] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Automated patch clamp (APC) technology was first developed at the turn of the millennium. The increased throughput it afforded promised a new paradigm in ion channel recordings, offering the potential to overcome the time-consuming, low-throughput bottleneck, arising from manual patch clamp investigations. This has relevance to the fast-paced development of novel therapies for chronic pain. This review highlights the advances in technology, using select examples that have facilitated APC usage in both industry and academia. It covers both first generation and the latest developments in second-generation platforms. In addition, it also provides an overview of the pain research field and how APC platforms have furthered our understanding of ion channel research and the development of pharmacological tools and therapeutics. APC platforms have much to offer to the ion channel research community, and this review highlights areas of best practice for both academia and industry. The impact of APC platforms and the prospects of ion channel research and improved therapeutics for chronic pain will be evaluated. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
| | - Mark L Dallas
- School of Pharmacy, University of Reading, Reading, UK
| |
Collapse
|
34
|
Ding XD, Wang W, Ding ZG, Liu YP, Zhong J, Chen HX. Impact of Botox-A SNAP-25 protein expression and the mechanism of inhibitory neurotransmitter imbalance in chronic sciatic nerve pain rat model. Exp Ther Med 2017; 13:2783-2786. [PMID: 28588664 PMCID: PMC5450774 DOI: 10.3892/etm.2017.4351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 03/16/2017] [Indexed: 12/17/2022] Open
Abstract
The Botox-A impact on the expression of SNAP-25 protein in rat chronic sciatic nerve pain model was assessed and the mechanism of inhibitory neurotransmitter imbalance was studied. A chronic constriction injury (CCI) model consisted of 30 healthy male rats. The rats were randomly divided into the sham-operated group, CCI group and BoNT/A intervention group, and during 1, 7 and 14 days we conducted mechanical withdrawal threshold (MWT) test and thermal withdrawal latency (TWL) test before and after operation. After 14 days, the animals were sacrificed. SNAP-25 protein expression level, mRNA subunit NR2B within excitatory neurotransmitter glutamate GLT and protein expression level, as well as GAT mRNA, the inhibitory GABA neurotransmitter transporter and protein expression level were studied by RT-polymerase chain reaction and western blot analysis. The difference between MWT and TWL at each point in time before and after operation showed no statistical significance (P>0.05) in the sham-operated group. For the CCI group at each time point, MWT and TWL were obviously lower than the sham-operated group and the difference was statistically significant (P<0.05) while the internal difference at each time point showed no statistical significance (P>0.05). The expression level of protein of SNAP-25 and NR2B mRNA in the CCI group was clearly higher than sham-operated group. Additionally, the expression level of GAT-1 mRNA and protein in CCI group was apparently lower than the sham-operated group. In conclusion, Botox-A helped reduce SNAP-25 within rat chronic sciatic nerve pain model thereby relieving pain.
Collapse
|
35
|
Abstract
WNK kinases, along with their upstream regulators (CUL3/KLHL3) and downstream targets (the SPAK/OSR1 kinases and the cation-Cl- cotransporters [CCCs]), comprise a signaling cascade essential for ion homeostasis in the kidney and nervous system. Recent work has furthered our understanding of the WNKs in epithelial transport, cell volume homeostasis, and GABA signaling, and uncovered novel roles for this pathway in immune cell function and cell proliferation.
Collapse
Affiliation(s)
- Masoud Shekarabi
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jinwei Zhang
- Departments of Neurosurgery, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06477, USA; MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Arjun R Khanna
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
| | - David H Ellison
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon 97239, USA; VA Portland Health Care System, Portland, OR 97239, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06477, USA.
| |
Collapse
|
36
|
Abstract
The mammalian nervous system encodes many different forms of pain, from those that arise as a result of short-term low-grade interactions with noxious thermal, chemical, or mechanical sources to more serious forms of pain induced by trauma and disease. In this Review, we highlight recent advances in our understanding of the neural circuits that encode these types of pain. Promising therapeutic strategies based on recent advances are also highlighted.
Collapse
Affiliation(s)
- Cedric Peirs
- Departments of Neurobiology and Otolaryngology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, BST3, Pittsburgh, PA 15213, USA
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Rebecca P Seal
- Departments of Neurobiology and Otolaryngology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, BST3, Pittsburgh, PA 15213, USA.
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
37
|
Zhang J, Deng X, Kahle KT. Leveraging unique structural characteristics of WNK kinases to achieve therapeutic inhibition. Sci Signal 2016; 9:e3. [PMID: 27811182 DOI: 10.1126/scisignal.aaj2227] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The with-no-lysine (K) WNK kinases are master regulators of the Na+-(K+)-Cl- cotransporters, including the renal-specific NCC and NKCC2 cotransporters. The discovery of WNK463, an orally bioavailable pan-WNK kinase inhibitor that exploits unique structural properties of the WNK catalytic domain to achieve high affinity and kinase selectivity, illustrates a strategy of leveraging distinct kinase features to develop specific inhibitors and validates the genetic predictions of the in vivo pharmacology of WNK inhibition.
Collapse
Affiliation(s)
- Jinwei Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kristopher T Kahle
- Department of Cellular and Molecular Physiology and Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
38
|
Moon HC, Lee YJ, Cho CB, Park YS. Suppressed GABAergic signaling in the zona incerta causes neuropathic pain in a thoracic hemisection spinal cord injury rat model. Neurosci Lett 2016; 632:55-61. [DOI: 10.1016/j.neulet.2016.08.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/19/2016] [Accepted: 08/20/2016] [Indexed: 01/01/2023]
|
39
|
Abstract
WNK (With-No-Lysine (K)) kinases are serine-threonine kinases characterized by an atypical placement of a catalytic lysine within the kinase domain. Mutations in human WNK1 or WNK4 cause an autosomal dominant syndrome of hypertension and hyperkalemia, reflecting the fact that WNK kinases are critical regulators of renal ion transport processes. Here, the role of WNKs in the regulation of ion transport processes in vertebrate and invertebrate renal function, cellular and organismal osmoregulation, and cell migration and cerebral edema will be reviewed, along with emerging literature demonstrating roles for WNKs in cardiovascular and neural development, Wnt signaling, and cancer. Conserved roles for these kinases across phyla are emphasized.
Collapse
Affiliation(s)
| | - Andreas Jenny
- Albert Einstein College of Medicine, New York, NY, United States.
| |
Collapse
|
40
|
Tang BL. (WNK)ing at death: With-no-lysine (Wnk) kinases in neuropathies and neuronal survival. Brain Res Bull 2016; 125:92-8. [PMID: 27131446 DOI: 10.1016/j.brainresbull.2016.04.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/11/2016] [Accepted: 04/24/2016] [Indexed: 12/22/2022]
Abstract
Members of With-no-lysine (WNK) family of serine-threonine kinase are key regulators of chloride ion transport in diverse cell types, controlling the activity and the surface expression of cation-chloride (Na(+)/K(+)-Cl(-)) co-transporters. Mutations in WNK1 and WNK4 are linked to a hereditary form of hypertension, and WNKs have been extensively investigated pertaining to their roles in renal epithelial ion homeostasis. However, some members of the WNK family and their splice isoforms are also expressed in the mammalian brain, and have been implicated in aspects of hereditary neuropathy as well as neuronal and glial survival. WNK2, which is exclusively enriched in neurons, is well known as an anti-proliferative tumor suppressor. WNK3, on the other hand, appears to promote cell survival as its inhibition enhances neuronal apoptosis. However, loss of WNK3 has been recently shown to reduce ischemia-associated brain damage. In this review, I surveyed the potentially context-dependent roles of WNKs in neurological disorders and neuronal survival.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.
| |
Collapse
|