1
|
Pan YZ, Talifu Z, Wang XX, Ke H, Zhang CJ, Xu X, Yang DG, Yu Y, Du LJ, Gao F, Li JJ. Combined use of CLP290 and bumetanide alleviates neuropathic pain and its mechanism after spinal cord injury in rats. CNS Neurosci Ther 2024; 30:e70045. [PMID: 39267289 PMCID: PMC11393004 DOI: 10.1111/cns.70045] [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: 04/11/2023] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/17/2024] Open
Abstract
AIM We aimed to explore whether the combination of CLP290 and bumetanide maximally improves neuropathic pain following spinal cord injury (SCI) and its possible molecular mechanism. METHODS Rats were randomly divided into five groups: Sham, SCI + vehicle, SCI + CLP290, SCI + bumetanide, and SCI + combination (CLP290 + bumetanide). Drug administration commenced on the 7th day post-injury (7 dpi) and continued for 14 days. All rats underwent behavioral assessments for 56 days to comprehensively evaluate the effects of interventions on mechanical pain, thermal pain, cold pain, motor function, and other relevant parameters. Electrophysiological assessments, immunoblotting, and immunofluorescence detection were performed at different timepoints post-injury, with a specific focus on the expression and changes of KCC2 and NKCC1 proteins in the lumbar enlargement of the spinal cord. RESULTS CLP290 and bumetanide alleviated SCI-associated hypersensitivity and locomotor function, with the combination providing enhanced recovery. The combined treatment group exhibited the most significant improvement in restoring Rate-Dependent Depression (RDD) levels. In the combined treatment group and the two individual drug administration groups, the upregulation of potassium chloride cotransporter 2 (K+-Cl-cotransporter 2, KCC2) expression and downregulation of sodium potassium chloride cotransporter 1 (Na+-K+-Cl-cotransporter 1, NKCC1) expression in the lumbar enlargement area resulted in a significant increase in the KCC2/NKCC1 ratio compared to the SCI + vehicle group, with the most pronounced improvement seen in the combined treatment group. Compared to the SCI + vehicle group, the SCI + bumetanide group showed no significant paw withdrawal thermal latency (PWTL) improvement at 21 and 35 dpi, but a notable enhancement at 56 dpi. In contrast, the SCI + CLP290 group significantly improved PWTL at 21 days, with non-significant changes at 35 and 56 days. At 21 dpi, KCC2 expression was marginally higher in monotherapy groups versus SCI + vehicle, but not significantly. At 56 dpi, only the SCI + bumetanide group showed a significant difference in KCC2 expression compared to the control group. CONCLUSION Combined application of CLP290 and bumetanide effectively increases the ratio of KCC2/NKCC1, restores RDD levels, enhances GABAA receptor-mediated inhibitory function in the spinal cord, and relieves neuropathic pain in SCI; Bumetanide significantly improves neuropathic pain in the long term, whereas CLP290 demonstrates a notable short-term effect.
Collapse
Affiliation(s)
- Yun-Zhu Pan
- Rehabilitation Medicine Department, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Zuliyaer Talifu
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
- Chinese Institute of Rehabilitation Science, Beijing, China
| | - Xiao-Xin Wang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
| | - Han Ke
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
| | - Chun-Jia Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
| | - Xin Xu
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
| | - De-Gang Yang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yan Yu
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liang-Jie Du
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Jian-Jun Li
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Chinese Institute of Rehabilitation Science, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| |
Collapse
|
2
|
Kok M, Brodsky JL. The biogenesis of potassium transporters: implications of disease-associated mutations. Crit Rev Biochem Mol Biol 2024; 59:154-198. [PMID: 38946646 PMCID: PMC11444911 DOI: 10.1080/10409238.2024.2369986] [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: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
The concentration of intracellular and extracellular potassium is tightly regulated due to the action of various ion transporters, channels, and pumps, which reside primarily in the kidney. Yet, potassium transporters and cotransporters play vital roles in all organs and cell types. Perhaps not surprisingly, defects in the biogenesis, function, and/or regulation of these proteins are linked to range of catastrophic human diseases, but to date, few drugs have been approved to treat these maladies. In this review, we discuss the structure, function, and activity of a group of potassium-chloride cotransporters, the KCCs, as well as the related sodium-potassium-chloride cotransporters, the NKCCs. Diseases associated with each of the four KCCs and two NKCCs are also discussed. Particular emphasis is placed on how these complex membrane proteins fold and mature in the endoplasmic reticulum, how non-native forms of the cotransporters are destroyed in the cell, and which cellular factors oversee their maturation and transport to the cell surface. When known, we also outline how the levels and activities of each cotransporter are regulated. Open questions in the field and avenues for future investigations are further outlined.
Collapse
Affiliation(s)
- Morgan Kok
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Ju YH, Cho J, Park JY, Kim H, Hong EB, Park KD, Lee CJ, Chung E, Kim HI, Nam MH. Tonic excitation by astrocytic GABA causes neuropathic pain by augmenting neuronal activity and glucose metabolism. Exp Mol Med 2024; 56:1193-1205. [PMID: 38760512 PMCID: PMC11148027 DOI: 10.1038/s12276-024-01232-z] [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: 11/18/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 05/19/2024] Open
Abstract
Neuropathic pain is a debilitating condition caused by the hyperexcitability of spinal dorsal horn neurons and is often characterized by allodynia. Although neuron-independent mechanisms of hyperexcitability have been investigated, the contribution of astrocyte-neuron interactions remains unclear. Here, we show evidence of reactive astrocytes and their excessive GABA release in the spinal dorsal horn, which paradoxically leads to the tonic excitation of neighboring neurons in a neuropathic pain model. Using multiple electrophysiological methods, we demonstrated that neuronal hyperexcitability is attributed to both increased astrocytic GABA synthesis via monoamine oxidase B (MAOB) and the depolarized reversal potential of GABA-mediated currents (EGABA) via the downregulation of the neuronal K+/Cl- cotransporter KCC2. Furthermore, longitudinal 2-deoxy-2-[18F]-fluoro-D-glucose microPET imaging demonstrated increased regional glucose metabolism in the ipsilateral dorsal horn, reflecting neuronal hyperexcitability. Importantly, inhibiting MAOB restored the entire astrocytic GABA-mediated cascade and abrogated the increased glucose metabolism and mechanical allodynia. Overall, astrocytic GABA-mediated tonic excitation is critical for neuronal hyperexcitability, leading to mechanical allodynia and neuropathic pain.
Collapse
Affiliation(s)
- Yeon Ha Ju
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jongwook Cho
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Ji-Young Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Eun-Bin Hong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Euiheon Chung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyoung-Ihl Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
- Department of Neurosurgery, Presbyterian Medical Center, Jeonju, 54987, Republic of Korea.
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
4
|
McMoneagle E, Zhou J, Zhang S, Huang W, Josiah SS, Ding K, Wang Y, Zhang J. Neuronal K +-Cl - cotransporter KCC2 as a promising drug target for epilepsy treatment. Acta Pharmacol Sin 2024; 45:1-22. [PMID: 37704745 PMCID: PMC10770335 DOI: 10.1038/s41401-023-01149-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023] Open
Abstract
Epilepsy is a prevalent neurological disorder characterized by unprovoked seizures. γ-Aminobutyric acid (GABA) serves as the primary fast inhibitory neurotransmitter in the brain, and GABA binding to the GABAA receptor (GABAAR) regulates Cl- and bicarbonate (HCO3-) influx or efflux through the channel pore, leading to GABAergic inhibition or excitation, respectively. The neuron-specific K+-Cl- cotransporter 2 (KCC2) is essential for maintaining a low intracellular Cl- concentration, ensuring GABAAR-mediated inhibition. Impaired KCC2 function results in GABAergic excitation associated with epileptic activity. Loss-of-function mutations and altered expression of KCC2 lead to elevated [Cl-]i and compromised synaptic inhibition, contributing to epilepsy pathogenesis in human patients. KCC2 antagonism studies demonstrate the necessity of limiting neuronal hyperexcitability within the brain, as reduced KCC2 functioning leads to seizure activity. Strategies focusing on direct (enhancing KCC2 activation) and indirect KCC2 modulation (altering KCC2 phosphorylation and transcription) have proven effective in attenuating seizure severity and exhibiting anti-convulsant properties. These findings highlight KCC2 as a promising therapeutic target for treating epilepsy. Recent advances in understanding KCC2 regulatory mechanisms, particularly via signaling pathways such as WNK, PKC, BDNF, and its receptor TrkB, have led to the discovery of novel small molecules that modulate KCC2. Inhibiting WNK kinase or utilizing newly discovered KCC2 agonists has demonstrated KCC2 activation and seizure attenuation in animal models. This review discusses the role of KCC2 in epilepsy and evaluates its potential as a drug target for epilepsy treatment by exploring various strategies to regulate KCC2 activity.
Collapse
Affiliation(s)
- Erin McMoneagle
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK
| | - Jin 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
| | - Shiyao Zhang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital Xiamen University, School of Medicine, Xiamen University, Xiang'an Nan Lu, Xiamen, 361102, China
| | - Weixue Huang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Sunday Solomon Josiah
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK
| | - 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.
| | - 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.
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Hatherly Laboratories, Streatham Campus, Exeter, EX4 4PS, UK.
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital Xiamen University, School of Medicine, Xiamen University, Xiang'an Nan Lu, Xiamen, 361102, China.
- 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
|
Sun C, Deng J, Ma Y, Meng F, Cui X, Li M, Li J, Li J, Yin P, Kong L, Zhang L, Tang P. The dual role of microglia in neuropathic pain after spinal cord injury: Detrimental and protective effects. Exp Neurol 2023; 370:114570. [PMID: 37852469 DOI: 10.1016/j.expneurol.2023.114570] [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: 07/04/2023] [Revised: 09/21/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition that is frequently accompanied by neuropathic pain, resulting in significant physical and psychological harm to a vast number of individuals globally. Despite the high prevalence of neuropathic pain following SCI, the precise underlying mechanism remains incompletely understood. Microglia are a type of innate immune cell that are present in the central nervous system (CNS). They have been observed to have a significant impact on neuropathic pain following SCI. This article presents a comprehensive overview of recent advances in understanding the role of microglia in the development of neuropathic pain following SCI. Specifically, the article delves into the detrimental and protective effects of microglia on neuropathic pain following SCI, as well as the mechanisms underlying their interconversion. Furthermore, the article provides a thorough overview of potential avenues for future research in this area.
Collapse
Affiliation(s)
- Chang Sun
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; Department of Orthopedics, Air Force Medical Center, PLA, Beijing, China
| | - Junhao Deng
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Yifei Ma
- School of Medicine, Nankai University, Tianjin, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiang Cui
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Ming Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jiantao Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Jia Li
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Lingjie Kong
- State Key Laboratory of Precision Measurement Technology and Instruments, Department of Precision Instruments, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China; National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China.
| |
Collapse
|
6
|
Warfield R, Robinson JA, Podgorski RM, Miller AD, Burdo TH. Neuroinflammation in the Dorsal Root Ganglia and Dorsal Horn Contributes to Persistence of Nociceptor Sensitization in SIV-Infected Antiretroviral Therapy-Treated Macaques. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2017-2030. [PMID: 37734588 PMCID: PMC10699130 DOI: 10.1016/j.ajpath.2023.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023]
Abstract
Despite the development of antiretroviral therapy (ART), HIV-associated distal sensory polyneuropathy remains prevalent. Using SIV-infected rhesus macaques, this study examined molecular mechanisms of peripheral and central sensitization to infer chronic pain from HIV infection. Previous studies identified atrophy in nociceptive neurons during SIV infection, which was associated with monocyte infiltration into the dorsal root ganglia (DRG). However, the sensory signaling mechanism connecting this pathology to symptoms remains unclear, especially because pain persists after resolution of high viremia and inflammation with ART. We hypothesized that residual DRG and dorsal horn neuroinflammation contributes to nociceptive sensitization. Using three cohorts of macaques [uninfected (SIV-), SIV-infected (SIV+), and SIV infected with ART (SIV+/ART)], this study showed an increase in the cellular and cytokine inflammatory profiles in the DRG of SIV+/ART macaques compared with uninfected animals. It found significant increase in the expression of nociceptive ion channels, TRPV1, and TRPA1 among DRG neurons in SIV+/ART compared with uninfected animals. SIV-infected and SIV+/ART animals showed reduced innervation of the nonpeptidergic nociceptors into the dorsal horn compared with uninfected animals. Finally, there were a significantly higher number of CD68+ cells in the dorsal horn of SIV+/ART macaques compared with uninfected animals. In summary, these data demonstrate that neuroinflammation, characteristics of nociceptor sensitization, and central terminal atrophy persists in SIV+/ART animals.
Collapse
Affiliation(s)
- Rebecca Warfield
- Department of Microbiology, Immunology, and Inflammation, Center for NeuroVirology and Gene Editing, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Jake A Robinson
- Department of Microbiology, Immunology, and Inflammation, Center for NeuroVirology and Gene Editing, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Rachel M Podgorski
- Department of Microbiology, Immunology, and Inflammation, Center for NeuroVirology and Gene Editing, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Andrew D Miller
- Section of Anatomic Pathology, Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York
| | - Tricia H Burdo
- Department of Microbiology, Immunology, and Inflammation, Center for NeuroVirology and Gene Editing, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania.
| |
Collapse
|
7
|
Calvo PM, de la Cruz RR, Pastor AM, Alvarez FJ. Preservation of KCC2 expression in axotomized abducens motoneurons and its enhancement by VEGF. Brain Struct Funct 2023; 228:967-984. [PMID: 37005931 PMCID: PMC10428176 DOI: 10.1007/s00429-023-02635-w] [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: 11/08/2022] [Accepted: 03/23/2023] [Indexed: 04/04/2023]
Abstract
The potassium chloride cotransporter 2 (KCC2) is the main Cl- extruder in neurons. Any alteration in KCC2 levels leads to changes in Cl- homeostasis and, consequently, in the polarity and amplitude of inhibitory synaptic potentials mediated by GABA or glycine. Axotomy downregulates KCC2 in many different motoneurons and it is suspected that interruption of muscle-derived factors maintaining motoneuron KCC2 expression is in part responsible. In here, we demonstrate that KCC2 is expressed in all oculomotor nuclei of cat and rat, but while trochlear and oculomotor motoneurons downregulate KCC2 after axotomy, expression is unaltered in abducens motoneurons. Exogenous application of vascular endothelial growth factor (VEGF), a neurotrophic factor expressed in muscle, upregulated KCC2 in axotomized abducens motoneurons above control levels. In parallel, a physiological study using cats chronically implanted with electrodes for recording abducens motoneurons in awake animals, demonstrated that inhibitory inputs related to off-fixations and off-directed saccades in VEGF-treated axotomized abducens motoneurons were significantly higher than in control, but eye-related excitatory signals in the on direction were unchanged. This is the first report of lack of KCC2 regulation in a motoneuron type after injury, proposing a role for VEGF in KCC2 regulation and demonstrating the link between KCC2 and synaptic inhibition in awake, behaving animals.
Collapse
Affiliation(s)
- Paula M Calvo
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012, Seville, Spain
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA
| | - Rosa R de la Cruz
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012, Seville, Spain
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012, Seville, Spain
| | | |
Collapse
|
8
|
Natale CA, Christie MJ, Aubrey KR. Spinal glycinergic currents are reduced in a rat model of neuropathic pain following partial nerve ligation but not chronic constriction injury. J Neurophysiol 2023; 129:333-341. [PMID: 36541621 DOI: 10.1152/jn.00451.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Animal models have consistently indicated that central sensitization and the development of chronic neuropathic pain are linked to changes to inhibitory signaling in the dorsal horn of the spinal cord. However, replication of data investigating the cellular mechanisms that underlie these changes remains a challenge and there is still a lack of understanding about what aspects of spinal inhibitory transmission most strongly contribute to the disease. Here, we compared the effect of two different sciatic nerve injuries commonly used to generate rodent models of neuropathic pain on spinal glycinergic signaling. Using whole cell patch-clamp electrophysiology in spinal slices, we recorded from neurons in the lamina II of the dorsal horn and evoked inhibitory postsynaptic currents with a stimulator in lamina III, where glycinergic cell bodies are concentrated. We found that glycine inputs onto radial neurons were reduced following partial nerve ligation (PNL) of the sciatic nerve, consistent with a previous report. However, this finding was not replicated in animals that underwent chronic constriction injury (CCI) to the same nerve region. To limit the between-experiment variability, we kept the rat species, sex, and age consistent and had a single investigator carry out the surgeries. These data show that PNL and CCI cause divergent spinal signaling outcomes in the cord and add to the body of evidence suggesting that treatments for neuropathic pain should be triaged according to nerve injury or cellular dysfunction rather than the symptoms of the disease.NEW & NOTEWORTHY Neuropathic pain models are used in preclinical research to investigate the mechanisms underlying allodynia, a common symptom of neuropathic pain, and to test, develop, and validate therapies for persistent pain. We demonstrate that a glycinergic dysfunction is consistently associated with partial nerve ligation but not the chronic constriction injury model. This suggests that the cellular effects produced by each injury are distinct and that data from different neuropathic pain models should be considered separately.
Collapse
Affiliation(s)
- Claudia A Natale
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Macdonald J Christie
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Karin R Aubrey
- Pain Management Research, Kolling Institute, Royal North Shore Hospital, St Leonards, New South Wales, Australia.,Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
9
|
Petroianu GA, Aloum L, Adem A. Neuropathic pain: Mechanisms and therapeutic strategies. Front Cell Dev Biol 2023; 11:1072629. [PMID: 36727110 PMCID: PMC9884983 DOI: 10.3389/fcell.2023.1072629] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
The physiopathology and neurotransmission of pain are of an owe inspiring complexity. Our ability to satisfactorily suppress neuropathic or other forms of chronic pain is limited. The number of pharmacodynamically distinct and clinically available medications is low and the successes achieved modest. Pain Medicine practitioners are confronted with the ethical dichotomy imposed by Hippocrates: On one hand the mandate of primum non nocere, on the other hand, the promise of heavenly joys if successful divinum est opus sedare dolorem. We briefly summarize the concepts associated with nociceptive pain from nociceptive input (afferents from periphery), modulatory output [descending noradrenergic (NE) and serotoninergic (5-HT) fibers] to local control. The local control is comprised of the "inflammatory soup" at the site of pain origin and synaptic relay stations, with an ATP-rich environment promoting inflammation and nociception while an adenosine-rich environment having the opposite effect. Subsequently, we address the transition from nociceptor pain to neuropathic pain (independent of nociceptor activation) and the process of sensitization and pain chronification (transient pain progressing into persistent pain). Having sketched a model of pain perception and processing we attempt to identify the sites and modes of action of clinically available drugs used in chronic pain treatment, focusing on adjuvant (co-analgesic) medication.
Collapse
|
10
|
Wu Y, Wang F. Inhibition of NKCC1 in spinal dorsal horn and dorsal root ganglion results in alleviation of neuropathic pain in rats with spinal cord contusion. Mol Pain 2023; 19:17448069231159855. [PMID: 36760008 PMCID: PMC9950615 DOI: 10.1177/17448069231159855] [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] [Indexed: 02/11/2023] Open
Abstract
Previous studies have confirmed the relationship between chloride homeostasis and pain. However, the role of sodium potassium chloride co-transporter isoform 1 (NKCC1) in dorsal horn and dorsal root ganglion neurons (DRGs) in spinal cord injury (SCI)-induced neuropathic pain (NP) remains inconclusive. Therefore, we aimed to explore whether suppression of NKCC1 in the spinal cord and DRGs alleviate the NP of adult rats with thoracic spinal cord contusion. Thirty adult female Sprague-Dawley rats (8 week-old, weighing 250-280 g) were randomly divided into three groups with ten animals in each group (sham, SCI, and bumetanide groups). The paw withdrawal mechanical threshold and paw withdrawal thermal latency were recorded before injury (baseline) and on post-injury days 14, 21, 28, and 35. At the end of experiment, western blotting (WB) analysis, quantitative real-time Polymerase Chain Reaction (PCR) and immunofluorescence were performed to quantify NKCC1 expression. Our results revealed that NKCC1 protein expression in the spinal cord and DRGs was significantly up-regulated in rats with SCI. Intraperitoneal treatment of bumetanide (an NKCC1 inhibitor) reversed the expression of NKCC1 in the dorsal horn and DRGs and ameliorated mechanical ectopic pain and thermal hypersensitivities in the SCI rats. Our study demonstrated the occurrence of NKCC1 overexpression in the spinal cord and DRGs in a rodent model of NP and indicated that changes in the peripheral nervous system also play a major role in promoting pain sensitization after SCI.
Collapse
Affiliation(s)
- Yao Wu
- School of Rehabilitation Medicine, 12517Capital Medical University, Beijing, China.,Department of Spine Surgery, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Fangyong Wang
- School of Rehabilitation Medicine, 12517Capital Medical University, Beijing, China.,Department of Spine Surgery, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| |
Collapse
|
11
|
Talifu Z, Pan Y, Gong H, Xu X, Zhang C, Yang D, Gao F, Yu Y, Du L, Li J. The role of KCC2 and NKCC1 in spinal cord injury: From physiology to pathology. Front Physiol 2022; 13:1045520. [PMID: 36589461 PMCID: PMC9799334 DOI: 10.3389/fphys.2022.1045520] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The balance of ion concentrations inside and outside the cell is an essential homeostatic mechanism in neurons and serves as the basis for a variety of physiological activities. In the central nervous system, NKCC1 and KCC2, members of the SLC12 cation-chloride co-transporter (CCC) family, participate in physiological and pathophysiological processes by regulating intracellular and extracellular chloride ion concentrations, which can further regulate the GABAergic system. Over recent years, studies have shown that NKCC1 and KCC2 are essential for the maintenance of Cl- homeostasis in neural cells. NKCC1 transports Cl- into cells while KCC2 transports Cl- out of cells, thereby regulating chloride balance and neuronal excitability. An imbalance of NKCC1 and KCC2 after spinal cord injury will disrupt CI- homeostasis, resulting in the transformation of GABA neurons from an inhibitory state into an excitatory state, which subsequently alters the spinal cord neural network and leads to conditions such as spasticity and neuropathic pain, among others. Meanwhile, studies have shown that KCC2 is also an essential target for motor function reconstruction after spinal cord injury. This review mainly introduces the physiological structure and function of NKCC1 and KCC2 and discusses their pathophysiological roles after spinal cord injury.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yunzhu Pan
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Han Gong
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Chunjia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Degang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liangjie Du
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| | - Jianjun Li
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| |
Collapse
|
12
|
Yeo M, Zhang Q, Ding L, Shen X, Chen Y, Liedtke W. Spinal cord dorsal horn sensory gate in preclinical models of chemotherapy-induced painful neuropathy and contact dermatitis chronic itch becomes less leaky with Kcc2 gene expression-enhancing treatments. Front Mol Neurosci 2022; 15:911606. [PMID: 36504679 PMCID: PMC9731339 DOI: 10.3389/fnmol.2022.911606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 10/05/2022] [Indexed: 11/25/2022] Open
Abstract
Low intraneuronal chloride in spinal cord dorsal horn (SCDH) pain relay neurons is of critical relevance for physiological transmission of primary sensory afferents because low intraneuronal chloride dictates GABA-ergic and glycin-ergic neurotransmission to be inhibitory. If neuronal chloride rises to unphysiological levels, the primary sensory gate in the spinal cord dorsal horn becomes corrupted, with resulting behavioral hallmarks of hypersensitivity and allodynia, for example in pathological pain. Low chloride in spinal cord dorsal horn neurons relies on the robust gene expression of Kcc2 and sustained transporter function of the KCC2 chloride-extruding electroneutral transporter. Based on a recent report where we characterized the GSK3-inhibitory small molecule, kenpaullone, as a Kcc2 gene expression-enhancer that potently repaired diminished Kcc2 expression and KCC2 transporter function in SCDH pain relay neurons, we extend our recent findings by reporting (i) effective pain control in a preclinical model of taxol-induced painful peripheral neuropathy that was accomplished by topical application of a TRPV4/TRPA1 dual-inhibitory compound (compound 16-8), and was associated with the repair of diminished Kcc2 gene expression in the SCDH; and (ii) potent functioning of kenpaullone as an antipruritic in a DNFB contact dermatitis preclinical model. These observations suggest that effective peripheral treatment of chemotherapy-induced painful peripheral neuropathy impacts the pain-transmitting neural circuit in the SCDH in a beneficial manner by enhancing Kcc2 gene expression, and that chronic pruritus might be relayed in the primary sensory gate of the spinal cord, following similar principles as pathological pain, specifically relating to the critical functioning of Kcc2 gene expression and the KCC2 transporter function.
Collapse
Affiliation(s)
- Michele Yeo
- Departments of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Qiaojuan Zhang
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| | - LeAnne Ding
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| | - Xiangjun Shen
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States
| | - Yong Chen
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States,*Correspondence: Yong Chen
| | - Wolfgang Liedtke
- Departments of Neurology, Duke University Medical Center, Durham, NC, United States,Wolfgang Liedtke
| |
Collapse
|
13
|
Prael III FJ, Kim K, Du Y, Spitznagel BD, Sulikowski GA, Delpire E, Weaver CD. Discovery of Small Molecule KCC2 Potentiators Which Attenuate In Vitro Seizure-Like Activity in Cultured Neurons. Front Cell Dev Biol 2022; 10:912812. [PMID: 35813195 PMCID: PMC9263442 DOI: 10.3389/fcell.2022.912812] [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: 04/04/2022] [Accepted: 06/02/2022] [Indexed: 01/14/2023] Open
Abstract
KCC2 is a K+-Cl- cotransporter that is expressed in neurons throughout the central nervous system. Deficits in KCC2 activity have been implicated in a variety of neurological disorders, including epilepsy, chronic pain, autism spectrum disorders, and Rett syndrome. Therefore, it has been hypothesized that pharmacological potentiation of KCC2 activity could provide a treatment for these disorders. To evaluate the therapeutic potential of pharmacological KCC2 potentiation, drug-like, selective KCC2 potentiators are required. Unfortunately, the lack of such tools has greatly hampered the investigation of the KCC2 potentiation hypothesis. Herein, we describe the discovery and characterization of a new class of small-molecule KCC2 potentiator. This newly discovered class exhibits KCC2-dependent activity and a unique mechanistic profile relative to previously reported small molecules. Furthermore, we demonstrate that KCC2 potentiation by this new class of KCC2 potentiator attenuates seizure-like activity in neuronal-glial co-cultures. Together, our results provide evidence that pharmacological KCC2 potentiation, by itself, is sufficient to attenuate neuronal excitability in an in vitro model that is sensitive to anti-epileptic drugs. Our findings and chemical tools are important for evaluating the promise of KCC2 as a therapeutic target and could lay a foundation for the development of KCC2-directed therapeutics for multiple neurological disorders.
Collapse
Affiliation(s)
- Francis J. Prael III
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States
| | - Kwangho Kim
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States,Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Yu Du
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | | | - Gary A. Sulikowski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States,Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, United States,Department of Chemistry, Vanderbilt University, Nashville, TN, United States,*Correspondence: C. David Weaver,
| |
Collapse
|
14
|
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
|
15
|
Wong ML, Widerstrom-Noga E, Field-Fote EC. Effects of whole-body vibration on neuropathic pain and the relationship between pain and spasticity in persons with spinal cord injury. Spinal Cord 2022; 60:963-970. [PMID: 35468994 DOI: 10.1038/s41393-022-00806-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Whole-body vibration (WBV) appears to modulate reflex hyperexcitability and spasticity. Due to common underlying neural mechanisms between spasticity and neuropathic pain, WBV may also reduce chronic pain after spinal cord injury (SCI). Our objective was to determine whether there are dose-related changes in pain following WBV and to examine the relationships between neuropathic pain and reflex excitability. STUDY DESIGN Secondary analysis of a sub-population (participants with neuropathic pain, n = 16) from a larger trial comparing the effects of two different doses of WBV on spasticity in persons with SCI. SETTING Hospital/Rehabilitation Center in Atlanta, GA, USA. METHODS Participants were randomized to 8-bout or 16-bout WBV groups. Both groups received ten sessions of sham intervention, followed by ten sessions of WBV. Primary measures included the Neuropathic Pain Symptom Inventory (NPSI) for pain symptom severity and H-reflex paired-pulse depression (PPD) for reflex excitability. RESULTS Mean change in NPSI scores were not significantly different between the groups (7 ± 6; p = 0.29; ES = 0.57); however, 8-bouts of WBV were consistently beneficial for participants with high neuropathic pain symptom severity (NPSI total score >30), while 16-bouts of WBV appeared to increase pain in some individuals with high NPSI scores. A baseline NPSI cut score of 30 predicted PPD response (sensitivity = 1.0, specificity = 0.83), with higher NPSI scores associated with decreased PPD in response to WBV. CONCLUSIONS WBV in moderate doses appears to decrease neuropathic pain symptoms and improve reflex modulation. However, at higher doses neuropathic pain symptoms may be aggravated. Lower baseline NPSI scores were associated with improved reflex modulation.
Collapse
Affiliation(s)
- Marlon L Wong
- Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA. .,Department of Physical Therapy, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Eva Widerstrom-Noga
- Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA.,Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Edelle C Field-Fote
- Shepherd Center, Crawford Research Institute, Atlanta, GA, USA.,Emory University School of Medicine, Division of Physical Therapy, Atlanta, GA, USA.,Georgia Institute of Technology, School of Biological Sciences, Atlanta, GA, USA
| |
Collapse
|
16
|
Ba X, Ran C, Guo W, Guo J, Zeng Q, Liu T, Sun W, Xiao L, Xiong D, Huang Y, Jiang C, Hao Y. Three-Day Continuous Oxytocin Infusion Attenuates Thermal and Mechanical Nociception by Rescuing Neuronal Chloride Homeostasis via Upregulation KCC2 Expression and Function. Front Pharmacol 2022; 13:845018. [PMID: 35401174 PMCID: PMC8988046 DOI: 10.3389/fphar.2022.845018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Oxytocin (OT) and its receptor are promising targets for the treatment and prevention of the neuropathic pain. In the present study, we compared the effects of a single and continuous intrathecal infusion of OT on nerve injury-induced neuropathic pain behaviours in mice and further explore the mechanisms underlying their analgesic properties. We found that three days of continuous intrathecal OT infusion alleviated subsequent pain behaviours for 14 days, whereas a single OT injection induced a transient analgesia for 30 min, suggesting that only continuous intrathecal OT attenuated the establishment and development of neuropathic pain behaviours. Supporting this behavioural finding, continuous intrathecal infusion, but not short-term incubation of OT, reversed the nerve injury-induced depolarizing shift in Cl- reversal potential via restoring the function and expression of spinal K+-Cl- cotransporter 2 (KCC2), which may be caused by OT-induced enhancement of GABA inhibitory transmission. This result suggests that only continuous use of OT may reverse the pathological changes caused by nerve injury, thereby mechanistically blocking the establishment and development of pain. These findings provide novel evidence relevant for advancing understanding of the effects of continuous OT administration on the pathophysiology of pain.
Collapse
Affiliation(s)
- Xiyuan Ba
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Chenqiu Ran
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Wenjun Guo
- Department of Pain Medicine, Shenzhen, China
| | - Jing Guo
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen, China
| | - Qian Zeng
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Donglin Xiong
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Yelan Huang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Changyu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital, Shenzhen, China
| | - Yue Hao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
17
|
Serranilla M, Woodin MA. Striatal Chloride Dysregulation and Impaired GABAergic Signaling Due to Cation-Chloride Cotransporter Dysfunction in Huntington’s Disease. Front Cell Neurosci 2022; 15:817013. [PMID: 35095429 PMCID: PMC8795088 DOI: 10.3389/fncel.2021.817013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Intracellular chloride (Cl–) levels in mature neurons must be tightly regulated for the maintenance of fast synaptic inhibition. In the mature central nervous system (CNS), synaptic inhibition is primarily mediated by gamma-amino butyric acid (GABA), which binds to Cl– permeable GABAA receptors (GABAARs). The intracellular Cl– concentration is primarily maintained by the antagonistic actions of two cation-chloride cotransporters (CCCs): Cl–-importing Na+-K+-Cl– co-transporter-1 (NKCC1) and Cl– -exporting K+-Cl– co-transporter-2 (KCC2). In mature neurons in the healthy brain, KCC2 expression is higher than NKCC1, leading to lower levels of intracellular Cl–, and Cl– influx upon GABAAR activation. However, in neurons of the immature brain or in neurological disorders such as epilepsy and traumatic brain injury, impaired KCC2 function and/or enhanced NKCC1 expression lead to intracellular Cl– accumulation and GABA-mediated excitation. In Huntington’s disease (HD), KCC2- and NKCC1-mediated Cl–-regulation are also altered, which leads to GABA-mediated excitation and contributes to the development of cognitive and motor impairments. This review summarizes the role of Cl– (dys)regulation in the healthy and HD brain, with a focus on the basal ganglia (BG) circuitry and CCCs as potential therapeutic targets in the treatment of HD.
Collapse
|
18
|
Löscher W, Kaila K. CNS pharmacology of NKCC1 inhibitors. Neuropharmacology 2021; 205:108910. [PMID: 34883135 DOI: 10.1016/j.neuropharm.2021.108910] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022]
Abstract
The Na-K-2Cl cotransporter NKCC1 and the neuron-specific K-Cl cotransporter KCC2 are considered attractive CNS drug targets because altered neuronal chloride regulation and consequent effects on GABAergic signaling have been implicated in numerous CNS disorders. While KCC2 modulators are not yet clinically available, the loop diuretic bumetanide has been used off-label in attempts to treat brain disorders and as a tool for NKCC1 inhibition in preclinical models. Bumetanide is known to have anticonvulsant and neuroprotective effects under some pathophysiological conditions. However, as shown in several species from neonates to adults (mice, rats, dogs, and by extrapolation in humans), at the low clinical doses of bumetanide approved for diuresis, this drug has negligible access into the CNS, reaching levels that are much lower than what is needed to inhibit NKCC1 in cells within the brain parenchyma. Several drug discovery strategies have been initiated over the last ∼15 years to develop brain-permeant compounds that, ideally, should be selective for NKCC1 to eliminate the diuresis mediated by inhibition of renal NKCC2. The strategies employed to improve the pharmacokinetic and pharmacodynamic properties of NKCC1 blockers include evaluation of other clinically approved loop diuretics; development of lipophilic prodrugs of bumetanide; development of side-chain derivatives of bumetanide; and unbiased high-throughput screening approaches of drug discovery based on large chemical compound libraries. The main outcomes are that (1), non-acidic loop diuretics such as azosemide and torasemide may have advantages as NKCC1 inhibitors vs. bumetanide; (2), bumetanide prodrugs lead to significantly higher brain levels than the parent drug and have lower diuretic activity; (3), the novel bumetanide side-chain derivatives do not exhibit any functionally relevant improvement of CNS accessibility or NKCC1 selectivity vs. bumetanide; (4) novel compounds discovered by high-throughput screening may resolve some of the inherent problems of bumetanide, but as yet this has not been achieved. Thus, further research is needed to optimize the design of brain-permeant NKCC1 inhibitors. In parallel, a major challenge is to identify the mechanisms whereby various NKCC1-expressing cellular targets of these drugs within (e.g., neurons, oligodendrocytes or astrocytes) and outside the brain parenchyma (e.g., the blood-brain barrier, the choroid plexus, and the endocrine system), as well as molecular off-target effects, might contribute to their reported therapeutic and adverse effects.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany.
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Finland
| |
Collapse
|
19
|
Yeo M, Chen Y, Jiang C, Chen G, Wang K, Chandra S, Bortsov A, Lioudyno M, Zeng Q, Wang P, Wang Z, Busciglio J, Ji RR, Liedtke W. Repurposing cancer drugs identifies kenpaullone which ameliorates pathologic pain in preclinical models via normalization of inhibitory neurotransmission. Nat Commun 2021; 12:6208. [PMID: 34707084 PMCID: PMC8551327 DOI: 10.1038/s41467-021-26270-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
Inhibitory GABA-ergic neurotransmission is fundamental for the adult vertebrate central nervous system and requires low chloride concentration in neurons, maintained by KCC2, a neuroprotective ion transporter that extrudes intracellular neuronal chloride. To identify Kcc2 gene expression‑enhancing compounds, we screened 1057 cell growth-regulating compounds in cultured primary cortical neurons. We identified kenpaullone (KP), which enhanced Kcc2/KCC2 expression and function in cultured rodent and human neurons by inhibiting GSK3ß. KP effectively reduced pathologic pain-like behavior in mouse models of nerve injury and bone cancer. In a nerve-injury pain model, KP restored Kcc2 expression and GABA-evoked chloride reversal potential in the spinal cord dorsal horn. Delta-catenin, a phosphorylation-target of GSK3ß in neurons, activated the Kcc2 promoter via KAISO transcription factor. Transient spinal over-expression of delta-catenin mimicked KP analgesia. Our findings of a newly repurposed compound and a novel, genetically-encoded mechanism that each enhance Kcc2 gene expression enable us to re-normalize disrupted inhibitory neurotransmission through genetic re-programming.
Collapse
Affiliation(s)
- Michele Yeo
- Department of Neurology, Duke University Medical Center, Durham, NC, USA.
| | - Yong Chen
- Department of Neurology, Duke University Medical Center, Durham, NC, USA.
| | - Changyu Jiang
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Gang Chen
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Kaiyuan Wang
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Sharat Chandra
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Andrey Bortsov
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Maria Lioudyno
- Department of Neurobiology & Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, CA, USA
| | - Qian Zeng
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Peng Wang
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Zilong Wang
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA
| | - Jorge Busciglio
- Department of Neurobiology & Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, CA, USA
| | - Ru-Rong Ji
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | - Wolfgang Liedtke
- Department of Neurology, Duke University Medical Center, Durham, NC, USA.
- Department of Anesthesiology (Center for Translational Pain Medicine), Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
- Duke Neurology Clinics for Headache, Head-Pain and Trigeminal Sensory Disorders, Duke University Medical Center, Durham, NC, USA.
- Duke Anesthesiology Clinics for Innovative Pain Therapy, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
20
|
The Alteration of Chloride Homeostasis/GABAergic Signaling in Brain Disorders: Could Oxidative Stress Play a Role? Antioxidants (Basel) 2021; 10:antiox10081316. [PMID: 34439564 PMCID: PMC8389245 DOI: 10.3390/antiox10081316] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022] Open
Abstract
In neuronal precursors and immature neurons, the depolarizing (excitatory) effect of γ-Aminobutyric acid (GABA) signaling is associated with elevated [Cl−]i; as brain cells mature, a developmental switch occurs, leading to the decrease of [Cl−]i and to the hyperpolarizing (inhibitory) effect of GABAergic signaling. [Cl−]i is controlled by two chloride co-transporters: NKCC1, which causes Cl− to accumulate into the cells, and KCC2, which extrudes it. The ontogenetic upregulation of the latter determines the above-outlined switch; however, many other factors contribute to the correct [Cl−]i in mature neurons. The dysregulation of chloride homeostasis is involved in seizure generation and has been associated with schizophrenia, Down’s Syndrome, Autism Spectrum Disorder, and other neurodevelopmental disorders. Recently, much effort has been put into developing new drugs intended to inhibit NKCC1 activity, while no attention has been paid to the origin of [Cl−]i dysregulation. Our study examines the pathophysiology of Cl− homeostasis and focuses on the impact of oxidative stress (OS) and inflammation on the activity of Cl− co-transporters, highlighting the relevance of OS in numerous brain abnormalities and diseases. This hypothesis supports the importance of primary prevention during pregnancy. It also integrates the therapeutic framework addressed to restore normal GABAergic signaling by counteracting the alteration in chloride homeostasis in central nervous system (CNS) cells, aiming at limiting the use of drugs that potentially pose a health risk.
Collapse
|
21
|
Bilchak JN, Yeakle K, Caron G, Malloy D, Côté MP. Enhancing KCC2 activity decreases hyperreflexia and spasticity after chronic spinal cord injury. Exp Neurol 2021; 338:113605. [PMID: 33453210 PMCID: PMC7904648 DOI: 10.1016/j.expneurol.2021.113605] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 01/09/2021] [Indexed: 02/03/2023]
Abstract
After spinal cord injury (SCI), the majority of individuals develop spasticity, a debilitating condition involving involuntary movements, co-contraction of antagonistic muscles, and hyperreflexia. By acting on GABAergic and Ca2+-dependent signaling, current anti-spastic medications lead to serious side effects, including a drastic decrease in motoneuronal excitability which impairs motor function and rehabilitation efforts. Exercise, in contrast, decreases spastic symptoms without decreasing motoneuron excitability. These functional improvements coincide with an increase in expression of the chloride co-transporter KCC2 in lumbar motoneurons. Thus, we hypothesized that spastic symptoms can be alleviated directly through restoration of chloride homeostasis and endogenous inhibition by increasing KCC2 activity. Here, we used the recently developed KCC2 enhancer, CLP257, to evaluate the effects of acutely increasing KCC2 extrusion capability on spastic symptoms after chronic SCI. Sprague Dawley rats received a spinal cord transection at T12 and were either bike-trained or remained sedentary for 5 weeks. Increasing KCC2 activity in the lumbar enlargement improved the rate-dependent depression of the H-reflex and reduced both phasic and tonic EMG responses to muscle stretch in sedentary animals after chronic SCI. Furthermore, the improvements due to this pharmacological treatment mirror those of exercise. Together, our results suggest that pharmacologically increasing KCC2 activity is a promising approach to decrease spastic symptoms in individuals with SCI. By acting to directly restore endogenous inhibition, this strategy has potential to avoid severe side effects and improve the quality of life of affected individuals.
Collapse
Affiliation(s)
- Jadwiga N Bilchak
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Kyle Yeakle
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Guillaume Caron
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Dillon Malloy
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Injury Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, United States of America.
| |
Collapse
|
22
|
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
|
23
|
Kosaka Y, Yafuso T, Shimizu-Okabe C, Kim J, Kobayashi S, Okura N, Ando H, Okabe A, Takayama C. Development and persistence of neuropathic pain through microglial activation and KCC2 decreasing after mouse tibial nerve injury. Brain Res 2020; 1733:146718. [PMID: 32045595 DOI: 10.1016/j.brainres.2020.146718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 01/27/2020] [Accepted: 02/07/2020] [Indexed: 12/20/2022]
Abstract
Gamma-amino butyric acid (GABA) is an inhibitory neurotransmitter in the mature brain, but is excitatory during development and after motor nerve injury. This difference in GABAergic action depends on the intracellular chloride ion concentration ([Cl-]i), primarily regulated by potassium chloride co-transporter 2 (KCC2). To reveal precise processes of the neuropathic pain through changes in GABAergic action, we prepared tibial nerve ligation and severance models using male mice, and examined temporal relationships amongst changes in (1) the mechanical withdrawal threshold in the sural nerve area, (2) localization of the molecules involved in GABAergic transmission and its upstream signaling in the dorsal horn, and (3) histology of the tibial nerve. In the ligation model, tibial nerve degeneration disappeared by day 56, but mechanical allodynia, reduced KCC2 localization, and increased microglia density remained until day 90. Microglia density was higher in the tibial zone than the sural zone before day 21, but this result was inverted after day 28. In contrast, in the severance model, all above changes were detected until day 28, but were simultaneously and significantly recovered by day 90. These results suggested that in male mice, allodynia may be caused by reduced GABAergic synaptic inhibition, resulting from elevated [Cl-]i after the reduction of KCC2 by activated microglia. Furthermore, our results suggested that factors from degenerating nerve terminals may diffuse into the sural zone, whereby they induced the development of allodynia in the sural nerve area, while other factors in the sural zone may mediate persistent allodynia through the same pathway.
Collapse
Affiliation(s)
- Yoshinori Kosaka
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan
| | - Tsukasa Yafuso
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan
| | - Chigusa Shimizu-Okabe
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan
| | - Jeongtae Kim
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan; Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Shiori Kobayashi
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan
| | - Nobuhiko Okura
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan
| | - Hironobu Ando
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan
| | - Akihito Okabe
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan; Department of Nutritional Science, Faculty of Health and Welfare, Seinan Jo Gakuin University, Fukuoka 803-0835, Japan
| | - Chitoshi Takayama
- Department of Molecular Anatomy, Graduate School of Medicine, University of the Ryukyus, 207 Uehara 207, Nishihara, Okinawa 9030215, Japan.
| |
Collapse
|
24
|
Tang BL. The Expanding Therapeutic Potential of Neuronal KCC2. Cells 2020; 9:E240. [PMID: 31963584 PMCID: PMC7016893 DOI: 10.3390/cells9010240] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 02/06/2023] Open
Abstract
Dysfunctions in GABAergic inhibitory neural transmission occur in neuronal injuries and neurological disorders. The potassium-chloride cotransporter 2 (KCC2, SLC12A5) is a key modulator of inhibitory GABAergic inputs in healthy adult neurons, as its chloride (Cl-) extruding activity underlies the hyperpolarizing reversal potential for GABAA receptor Cl- currents (EGABA). Manipulation of KCC2 levels or activity improve symptoms associated with epilepsy and neuropathy. Recent works have now indicated that pharmacological enhancement of KCC2 function could reactivate dormant relay circuits in an injured mouse's spinal cord, leading to functional recovery and the attenuation of neuronal abnormality and disease phenotype associated with a mouse model of Rett syndrome (RTT). KCC2 interacts with Huntingtin and is downregulated in Huntington's disease (HD), which contributed to GABAergic excitation and memory deficits in the R6/2 mouse HD model. Here, these recent advances are highlighted, which attest to KCC2's growing potential as a therapeutic target for neuropathological conditions resulting from dysfunctional inhibitory input.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; ; Tel.: +65-6516-1040
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
| |
Collapse
|
25
|
Halbhuber L, Achtner C, Luhmann HJ, Sinning A, Kilb W. Coincident Activation of Glutamate Receptors Enhances GABA A Receptor-Induced Ionic Plasticity of the Intracellular Cl --Concentration in Dissociated Neuronal Cultures. Front Cell Neurosci 2019; 13:497. [PMID: 31787883 PMCID: PMC6856009 DOI: 10.3389/fncel.2019.00497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/21/2019] [Indexed: 01/09/2023] Open
Abstract
Massive activation of γ-amino butyric acid A (GABAA) receptors during pathophysiological activity induces an increase in the intracellular Cl--concentration ([Cl-]i), which is sufficient to render GABAergic responses excitatory. However, to what extent physiological levels of GABAergic activity can influence [Cl-]i is not known. Aim of the present study is to reveal whether moderate activation of GABAA receptors mediates functionally relevant [Cl-]i changes and whether these changes can be augmented by coincident glutamatergic activity. To address these questions, we used whole-cell patch-clamp recordings from cultured cortical neurons [at days in vitro (DIV) 6-22] to determine changes in the GABA reversal potential (EGABA) induced by short bursts of GABAergic and/or synchronized glutamatergic stimulation. These experiments revealed that pressure-application of 10 short muscimol pulses at 10 Hz induced voltage-dependent [Cl-]i changes. Under current-clamp conditions this muscimol burst induced a [Cl-]i increase of 3.1 ± 0.4 mM (n = 27), which was significantly enhanced to 4.6 ± 0.5 mM (n = 27) when glutamate was applied synchronously with the muscimol pulses. The muscimol-induced [Cl-]i increase significantly attenuated the inhibitory effect of GABA, as determined by the GABAergic rheobase shift. The synchronous coapplication of glutamate pulses had no additional effect on the attenuation of GABAergic inhibition, despite the larger [Cl-]i transients under these conditions. In summary, these results indicate that moderate GABAergic activity can induce functionally relevant [Cl-]i transients, which were enhanced by coincident glutamate pulses. This ionic plasticity of [Cl-]i may contribute to short-term plasticity of the GABAergic system.
Collapse
Affiliation(s)
- Lisa Halbhuber
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Cécilia Achtner
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Anne Sinning
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center Mainz, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
26
|
Akhter ET, Griffith RW, English AW, Alvarez FJ. Removal of the Potassium Chloride Co-Transporter from the Somatodendritic Membrane of Axotomized Motoneurons Is Independent of BDNF/TrkB Signaling But Is Controlled by Neuromuscular Innervation. eNeuro 2019; 6:ENEURO.0172-19.2019. [PMID: 31541001 PMCID: PMC6795555 DOI: 10.1523/eneuro.0172-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/21/2019] [Accepted: 09/15/2019] [Indexed: 01/09/2023] Open
Abstract
The potassium-chloride cotransporter (KCC2) maintains the low intracellular chloride found in mature central neurons and controls the strength and direction of GABA/glycine synapses. We found that following axotomy as a consequence of peripheral nerve injuries (PNIs), KCC2 protein is lost throughout the somatodendritic membrane of axotomized spinal cord motoneurons after downregulation of kcc2 mRNA expression. This large loss likely depolarizes the reversal potential of GABA/glycine synapses, resulting in GABAergic-driven spontaneous activity in spinal motoneurons similar to previous reports in brainstem motoneurons. We hypothesized that the mechanism inducing KCC2 downregulation in spinal motoneurons following peripheral axotomy might be mediated by microglia or motoneuron release of BDNF and TrkB activation as has been reported on spinal cord dorsal horn neurons after nerve injury, motoneurons after spinal cord injury (SCI), and in many other central neurons throughout development or a variety of pathologies. To test this hypothesis, we used genetic approaches to interfere with microglia activation or delete bdnf from specifically microglia or motoneurons, as well as pharmacology (ANA-12) and pharmacogenetics (F616A mice) to block TrkB activation. We show that KCC2 dysregulation in axotomized motoneurons is independent of microglia, BDNF, and TrkB. KCC2 is instead dependent on neuromuscular innervation; KCC2 levels are restored only when motoneurons reinnervate muscle. Thus, downregulation of KCC2 occurs specifically while injured motoneurons are regenerating and might be controlled by target-derived signals. GABAergic and glycinergic synapses might therefore depolarize motoneurons disconnected from their targets and contribute to augment motoneuron activity known to promote motor axon regeneration.
Collapse
Affiliation(s)
- Erica Tracey Akhter
- Departments of Physiology, Emory University, Atlanta, GA 30322
- Cell Biology, Emory University, Atlanta, GA 30322
| | | | | | | |
Collapse
|
27
|
Tibbett JA, Field‐Fote EC, Thomas CK, Widerström‐Noga EG. Spasticity and Pain after Spinal Cord Injury: Impact on Daily Life and the Influence of Psychological Factors. PM R 2019; 12:119-129. [DOI: 10.1002/pmrj.12218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/30/2019] [Indexed: 01/31/2023]
Affiliation(s)
- Jacqueline A. Tibbett
- The Miami Project to Cure Paralysis University of Miami Miami FL
- Department of Physiology and BiophysicsUniversity of Miami Miami FL
| | - Edelle C. Field‐Fote
- Shepherd Center, Crawford Research Institute Atlanta GA
- Division of Physical TherapyEmory University School of Medicine Atlanta GA
| | - Christine K. Thomas
- The Miami Project to Cure Paralysis University of Miami Miami FL
- Department of Physiology and BiophysicsUniversity of Miami Miami FL
- Department of Neurological SurgeryUniversity of Miami Miami FL
| | - Eva G. Widerström‐Noga
- The Miami Project to Cure Paralysis University of Miami Miami FL
- Department of Neurological SurgeryUniversity of Miami Miami FL
| |
Collapse
|
28
|
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
|
29
|
Cordshagen A, Busch W, Winklhofer M, Nothwang HG, Hartmann AM. Phosphoregulation of the intracellular termini of K +-Cl - cotransporter 2 (KCC2) enables flexible control of its activity. J Biol Chem 2018; 293:16984-16993. [PMID: 30201606 DOI: 10.1074/jbc.ra118.004349] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/01/2018] [Indexed: 12/22/2022] Open
Abstract
The pivotal role of K+-Cl- cotransporter 2 (KCC2) in inhibitory neurotransmission and severe human diseases fosters interest in understanding posttranslational regulatory mechanisms such as (de)phosphorylation. Here, the regulatory role of the five bona fide phosphosites Ser31, Thr34, Ser932, Thr999, and Thr1008 was investigated by the use of alanine and aspartate mutants. Tl+-based flux analyses in HEK-293 cells demonstrated increased transport activity for S932D (mimicking phosphorylation) and T1008A (mimicking dephosphorylation), albeit to a different extent. Increased activity was due to changes in intrinsic activity, as it was not caused by increased cell-surface abundance. Substitutions of Ser31, Thr34, or Thr999 had no effect. Additionally, we show that the indirect actions of the known KCC2 activators staurosporine and N-ethylmaleimide (NEM) involved multiple phosphosites. S31D, T34A, S932A/D, T999A, or T1008A/D abrogated staurosporine mediated stimulation, and S31A, T34D, or S932D abolished NEM-mediated stimulation. This demonstrates for the first time differential effects of staurosporine and NEM on KCC2. In addition, the staurosporine-mediated effects involved both KCC2 phosphorylation and dephosphorylation with Ser932 and Thr1008 being bona fide target sites. In summary, our data reveal a complex phosphoregulation of KCC2 that provides the transporter with a toolbox for graded activity and integration of different signaling pathways.
Collapse
Affiliation(s)
- Antje Cordshagen
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences
| | - Wiebke Busch
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences
| | - Michael Winklhofer
- Institute for Biology and Environmental Sciences IBU, and.,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Hans Gerd Nothwang
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences.,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| | - Anna-Maria Hartmann
- From the Neurogenetics group, Center of Excellence Hearing4all, School of Medicine and Health Sciences, .,Research Center for Neurosensory Sciences, Carl von Ossietzky University Oldenburg, 26111 Oldenburg, Germany
| |
Collapse
|
30
|
Greenwald JD, Shafritz KM. An Integrative Neuroscience Framework for the Treatment of Chronic Pain: From Cellular Alterations to Behavior. Front Integr Neurosci 2018; 12:18. [PMID: 29875641 PMCID: PMC5974053 DOI: 10.3389/fnint.2018.00018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/04/2018] [Indexed: 12/21/2022] Open
Abstract
Chronic pain can result from many pain syndromes including complex regional pain syndrome (CRPS), phantom limb pain and chronic low back pain, among others. On a molecular level, chronic pain syndromes arise from hypersensitization within the dorsal horn of the spinal cord, a process known as central sensitization. Central sensitization involves an upregulation of ionotropic and metabotropic glutamate receptors (mGluRs) similar to that of long-term potentiation (LTP). Regions of the brain in which LTP occurs, such as the amygdala and hippocampus, are implicated in fear- and memory-related brain circuity. Chronic pain dramatically influences patient quality of life. Individuals with chronic pain may develop pain-related anxiety and pain-related fear. The syndrome also alters functional connectivity in the default-mode network (DMN) and salience network. On a cellular/molecular level, central sensitization may be reversed through degradative glutamate receptor pathways. This, however, rarely happens. Instead, cortical brain regions may serve in a top-down regulatory capacity for the maintenance or alleviation of pain. Specifically, the medial prefrontal cortex (mPFC), which plays a critical role in fear-related brain circuits, the DMN, and salience network may be the driving forces in this process. On a cellular level, the mPFC may form new neural circuits through LTP that may cause extinction of pre-existing pain pathways found within fear-related brain circuits, the DMN, and salience network. In order to promote new LTP connections between the mPFC and other key brain structures, such as the amygdala and insula, we propose a holistic rehabilitation program including cognitive behavioral therapy (CBT) and revolving around: (1) cognitive reappraisals; (2) mindfulness meditation; and (3) functional rehabilitation. Unlike current medical interventions focusing upon pain-relieving medications, we do not believe that chronic pain treatment should focus on reversing the effects of central sensitization. Instead, we propose here that it is critical to focus on non-invasive efforts to promote new neural circuits originating from the mPFC.
Collapse
Affiliation(s)
- Jess D. Greenwald
- Department of Psychology, Hofstra University, Hempstead, NY, United States
| | - Keith M. Shafritz
- Department of Psychology, Hofstra University, Hempstead, NY, United States
- Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
31
|
Schulte JT, Wierenga CJ, Bruining H. Chloride transporters and GABA polarity in developmental, neurological and psychiatric conditions. Neurosci Biobehav Rev 2018; 90:260-271. [PMID: 29729285 DOI: 10.1016/j.neubiorev.2018.05.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
Neuronal chloride regulation is a determinant factor for the dynamic tuning of GABAergic inhibition during and beyond brain development. This regulation is mainly dependent on the two co-transporters K+/Cl- co-transporter KCC2 and Na+/K+/Cl- co-transporter NKCC1, whose activity can decrease or increase neuronal chloride concentrations respectively. Altered expression and/or activity of either of these co-transporters has been associated with a wide variety of brain disorders including developmental disorders, epilepsy, schizophrenia and stroke. Here, we review current knowledge on chloride transporter expression and activity regulation and highlight the intriguing potential for existing and future interventions to support chloride homeostasis across a wide range of mental disorders and neurological conditions.
Collapse
Affiliation(s)
- Joran T Schulte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands
| | - Corette J Wierenga
- Division of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Hilgo Bruining
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands.
| |
Collapse
|
32
|
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
|
33
|
Ludwig A, Rivera C, Uvarov P. A noninvasive optical approach for assessing chloride extrusion activity of the K-Cl cotransporter KCC2 in neuronal cells. BMC Neurosci 2017; 18:23. [PMID: 28143398 PMCID: PMC5286847 DOI: 10.1186/s12868-017-0336-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 01/12/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cation-chloride cotransporters (CCCs) are indispensable for maintaining chloride homeostasis in multiple cell types, but K-Cl cotransporter KCC2 is the only CCC member with an exclusively neuronal expression in mammals. KCC2 is critical for rendering fast hyperpolarizing responses of ionotropic γ-aminobutyric acid and glycine receptors in adult neurons, for neuronal migration in the developing central nervous system, and for the formation and maintenance of small dendritic protrusions-dendritic spines. Deficit in KCC2 expression and/or activity is associated with epilepsy and neuropathic pain, and effective strategies are required to search for novel drugs augmenting KCC2 function. RESULTS We revised current methods to develop a noninvasive optical approach for assessing KCC2 transport activity using a previously characterized genetically encoded chloride sensor. Our protocol directly assesses dynamics of KCC2-mediated chloride efflux and allows measuring genuine KCC2 activity with good spatial and temporal resolution. As a proof of concept, we used this approach to compare transport activities of the two known KCC2 splice isoforms, KCC2a and KCC2b, in mouse neuronal Neuro-2a cells. CONCLUSIONS Our noninvasive optical protocol proved to be efficient for assessment of furosemide-sensitive chloride fluxes. Transport activities of the N-terminal splice isoforms KCC2a and KCC2b obtained by the novel approach matched to those reported previously using standard methods for measuring chloride fluxes.
Collapse
Affiliation(s)
- Anastasia Ludwig
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), INSERM U1024, CNRS 8197, Paris, France
| | - Claudio Rivera
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- INSERM U901, Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France
- UMR S901, Aix-Marseille Université, Marseille, France
| | - Pavel Uvarov
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biosciences and Neuroscience Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Wu H, Shao A, Zhao M, Chen S, Yu J, Zhou J, Liang F, Shi L, Dixon BJ, Wang Z, Ling C, Hong Y, Zhang J. Melatonin attenuates neuronal apoptosis through up-regulation of K(+) -Cl(-) cotransporter KCC2 expression following traumatic brain injury in rats. J Pineal Res 2016; 61:241-50. [PMID: 27159133 DOI: 10.1111/jpi.12344] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 05/05/2016] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) initiates a complex cascade of neurochemical and signaling changes that leads to neuronal apoptosis, which contributes to poor outcomes for patients with TBI. The neuron-specific K(+) -Cl(-) cotransporter-2 (KCC2), the principal Cl(-) extruder in adult neurons, plays an important role in Cl(-) homeostasis and neuronal function. This present study was designed to investigate the expression pattern of KCC2 following TBI and to evaluate whether or not melatonin is able to prevent neuronal apoptosis by modulating KCC2 expression in a Sprague Dawley rat controlled cortical impact model of TBI. The time course study showed decreased mRNA and protein expression of KCC2 in the ipsilateral peri-core parietal cortex after TBI. Double immunofluorescence staining demonstrated that KCC2 is located in the plasma membrane of neurons. In addition, melatonin (10 mg/kg) was injected intraperitoneally at 5 minutes and repeated at 1, 2, 3, and 4 hours after brain trauma, and brain samples were extracted 24 hours after TBI. Compared to the vehicle group, melatonin treatment altered the down-regulation of KCC2 expression in both mRNA and protein levels after TBI. Also, melatonin treatment increased the protein levels of brain-derived neurotrophic factor (BDNF) and phosphorylated extracellular signal-regulated kinase (p-ERK). Simultaneously, melatonin administration ameliorated cortical neuronal apoptosis, reduced brain edema, and attenuated neurological deficits after TBI. In conclusion, our findings suggested that melatonin restores KCC2 expression, inhibits neuronal apoptosis and attenuates secondary brain injury after TBI, partially through activation of BDNF/ERK pathway.
Collapse
Affiliation(s)
- Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mingfei Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jun Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingyi Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Feng Liang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Brandon J Dixon
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Zhen Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chenhan Ling
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuan Hong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Korvela M, Lind AL, Wetterhall M, Gordh T, Andersson M, Pettersson J. Quantification of 10 elements in human cerebrospinal fluid from chronic pain patients with and without spinal cord stimulation. J Trace Elem Med Biol 2016; 37:1-7. [PMID: 27473826 DOI: 10.1016/j.jtemb.2016.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/23/2016] [Accepted: 06/01/2016] [Indexed: 12/14/2022]
Abstract
Neuropathic pain affects 1-10% of the general population and is caused by a lesion or disease of the somatosensory nervous system. Spinal cord stimulation (SCS), a method where implanted electrodes stimulate the spinal cord, has been successfully used to treat drug-resistant neuropathic pain, but the mechanism of action is largely unknown. Studies show that SCS changes the protein levels in CSF (cerebrospinal fluid) of pain patients. Several neurological conditions have been shown to alter the elemental composition of CSF. Therefore changes in the levels of ions and trace elements in the CSF may correspond to SCS use. This study used ICP-MS (Inductively coupled plasma mass spectrometry) and ICP-AES (Inductively coupled plasma atomic emission spectroscopy) to quantify 10 elements in CSF from chronic neuropathic pain patients using SCS. The element concentrations in CSF from patients with SCS treatment on/off, were measured. No effect on the element concentrations in CSF from treatment with SCS could be detected. Also, the elemental concentrations in pooled CSF from patients without chronic neuropathic pain was determined and compared to the patients using SCS. The concentration of the elements Ca, Sr, Na, K, P, Mg and Ti were, significantly higher in patients compared to the CSF-control.
Collapse
Affiliation(s)
- Marcus Korvela
- Department of Chemistry-BMC, Uppsala University, Box 599, 751 24 Uppsala, Sweden
| | - Anne-Li Lind
- Department of Chemistry-BMC, Uppsala University, Box 599, 751 24 Uppsala, Sweden; Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, 751 85 Uppsala, Sweden
| | | | - Torsten Gordh
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, 751 85 Uppsala, Sweden
| | - Marit Andersson
- Department of Chemistry-BMC, Uppsala University, Box 599, 751 24 Uppsala, Sweden
| | - Jean Pettersson
- Department of Chemistry-BMC, Uppsala University, Box 599, 751 24 Uppsala, Sweden.
| |
Collapse
|
36
|
Doyon N, Vinay L, Prescott SA, De Koninck Y. Chloride Regulation: A Dynamic Equilibrium Crucial for Synaptic Inhibition. Neuron 2016; 89:1157-1172. [PMID: 26985723 DOI: 10.1016/j.neuron.2016.02.030] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 12/24/2015] [Accepted: 02/18/2016] [Indexed: 01/02/2023]
Abstract
Fast synaptic inhibition relies on tight regulation of intracellular Cl(-). Chloride dysregulation is implicated in several neurological and psychiatric disorders. Beyond mere disinhibition, the consequences of Cl(-) dysregulation are multifaceted and best understood in terms of a dynamical system involving complex interactions between multiple processes operating on many spatiotemporal scales. This dynamical perspective helps explain many unintuitive manifestations of Cl(-) dysregulation. Here we discuss how taking into account dynamical regulation of intracellular Cl(-) is important for understanding how synaptic inhibition fails, how to best detect that failure, why Cl(-) regulation is energetically so expensive, and the overall consequences for therapeutics.
Collapse
Affiliation(s)
- Nicolas Doyon
- Institut Universitaire en Santé Mentale de Québec, Québec, QC G1J 2G3, Canada; Department of Mathematics and Statistics, Université Laval, Québec, QC G1V 0A6, Canada
| | - Laurent Vinay
- Team P3M, Institut de Neurosciences de la Timone, UMR 7289, CNRS and Aix Marseille Université, F-13385 Marseille, France
| | - Steven A Prescott
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yves De Koninck
- Institut Universitaire en Santé Mentale de Québec, Québec, QC G1J 2G3, Canada; Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
37
|
Wu H, Che X, Tang J, Ma F, Pan K, Zhao M, Shao A, Wu Q, Zhang J, Hong Y. The K(+)-Cl(-) Cotransporter KCC2 and Chloride Homeostasis: Potential Therapeutic Target in Acute Central Nervous System Injury. Mol Neurobiol 2016; 53:2141-51. [PMID: 25941074 DOI: 10.1007/s12035-015-9162-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/26/2015] [Indexed: 12/11/2022]
Abstract
The K(+)-Cl(-) cotransporter-2 (KCC2) is a well-known member of the electroneutral cation-chloride cotransporters with a restricted expression pattern to neurons. This transmembrane protein mediates the efflux of Cl(-) out of neurons and exerts a critical role in inhibitory γ-aminobutyric acidergic (GABAergic) and glycinergic neurotransmission. Moreover, KCC2 participates in the regulation of various physiological processes of neurons, including cell migration, dendritic outgrowth, spine morphology, and dendritic synaptogenesis. It is important to note that down-regulation of KCC2 is associated with the pathogenesis of multiple neurological diseases, which is of particular relevance to acute central nervous system (CNS) injury. In this review, we aim to survey the pathogenic significance of KCC2 down-regulation under the condition of acute CNS injuries. We propose that further elucidation of the molecular mechanisms regarding KCC2 down-regulation after acute CNS injuries is necessary because of potential promising avenues for prevention and treatment of acute CNS injury.
Collapse
Affiliation(s)
- Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Xiaoru Che
- Department of Cardiology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Junjia Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Feiqiang Ma
- Department of Emergency Medicine, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kun Pan
- Department of Neurosurgery, New York-Presbyterian Hospital, New York, NY, USA
| | - Mingfei Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yuan Hong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
38
|
Kahle KT, Delpire E. Kinase-KCC2 coupling: Cl- rheostasis, disease susceptibility, therapeutic target. J Neurophysiol 2016; 115:8-18. [PMID: 26510764 PMCID: PMC4760510 DOI: 10.1152/jn.00865.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/25/2015] [Indexed: 01/06/2023] Open
Abstract
The intracellular concentration of Cl(-) ([Cl(-)]i) in neurons is a highly regulated variable that is established and modulated by the finely tuned activity of the KCC2 cotransporter. Despite the importance of KCC2 for neurophysiology and its role in multiple neuropsychiatric diseases, our knowledge of the transporter's regulatory mechanisms is incomplete. Recent studies suggest that the phosphorylation state of KCC2 at specific residues in its cytoplasmic COOH terminus, such as Ser940 and Thr906/Thr1007, encodes discrete levels of transporter activity that elicit graded changes in neuronal Cl(-) extrusion to modulate the strength of synaptic inhibition via Cl(-)-permeable GABAA receptors. In this review, we propose that the functional and physical coupling of KCC2 to Cl(-)-sensitive kinase(s), such as the WNK1-SPAK kinase complex, constitutes a molecular "rheostat" that regulates [Cl(-)]i and thereby influences the functional plasticity of GABA. The rapid reversibility of (de)phosphorylation facilitates regulatory precision, and multisite phosphorylation allows for the control of KCC2 activity by different inputs via distinct or partially overlapping upstream signaling cascades that may become more or less important depending on the physiological context. While this adaptation mechanism is highly suited to maintaining homeostasis, its adjustable set points may render it vulnerable to perturbation and dysregulation. Finally, we suggest that pharmacological modulation of this kinase-KCC2 rheostat might be a particularly efficacious strategy to enhance Cl(-) extrusion and therapeutically restore GABA inhibition.
Collapse
Affiliation(s)
- Kristopher T Kahle
- Departments of Neurosurgery and Pediatrics, Yale School of Medicine, New Haven, Connecticut; Yale Neurogenetics Program, Yale School of Medicine, New Haven, Connecticut; and
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
39
|
Kahle KT, Khanna AR, Alper SL, Adragna NC, Lauf PK, Sun D, Delpire E. K-Cl cotransporters, cell volume homeostasis, and neurological disease. Trends Mol Med 2015; 21:513-23. [PMID: 26142773 PMCID: PMC4834970 DOI: 10.1016/j.molmed.2015.05.008] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/10/2015] [Accepted: 05/29/2015] [Indexed: 11/24/2022]
Abstract
K(+)-Cl(-) cotransporters (KCCs) were originally characterized as regulators of red blood cell (RBC) volume. Since then, four distinct KCCs have been cloned, and their importance for volume regulation has been demonstrated in other cell types. Genetic models of certain KCCs, such as KCC3, and their inhibitory WNK-STE20/SPS1-related proline/alanine-rich kinase (SPAK) serine-threonine kinases, have demonstrated the evolutionary necessity of these molecules for nervous system cell volume regulation, structure, and function, and their involvement in neurological disease. The recent characterization of a swelling-activated dephosphorylation mechanism that potently stimulates the KCCs has pinpointed a potentially druggable switch of KCC activity. An improved understanding of WNK/SPAK-mediated KCC cell volume regulation in the nervous system might reveal novel avenues for the treatment of multiple neurological diseases.
Collapse
Affiliation(s)
- Kristopher T Kahle
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02114, USA; Manton Center for Orphan Disease Research, Children's Hospital Boston, 300 Longwood Avenue, Boston, MA 02114, USA.
| | - Arjun R Khanna
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Seth L Alper
- Renal Division and Molecular and Vascular Medicine Division, Beth Israel Deaconess Medical Center; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Norma C Adragna
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Peter K Lauf
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA; Department of Pathology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15217, USA; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA 15213, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
40
|
Yaksh TL, Woller SA, Ramachandran R, Sorkin LS. The search for novel analgesics: targets and mechanisms. F1000PRIME REPORTS 2015; 7:56. [PMID: 26097729 PMCID: PMC4447049 DOI: 10.12703/p7-56] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The management of the pain state is of great therapeutic relevance to virtually every medical specialty. Failure to manage its expression has deleterious consequence to the well-being of the organism. An understanding of the complex biology of the mechanisms underlying the processing of nociceptive information provides an important pathway towards development of novel and robust therapeutics. Importantly, preclinical models have been of considerable use in determining the linkage between mechanism and the associated behaviorally defined pain state. This review seeks to provide an overview of current thinking targeting pain biology, the use of preclinical models and the development of novel pain therapeutics. Issues pertinent to the strengths and weaknesses of current development strategies for analgesics are considered.
Collapse
|
41
|
Tang D, Qian AH, Song DD, Ben QW, Yao WY, Sun J, Li WG, Xu TL, Yuan YZ. Role of the potassium chloride cotransporter isoform 2-mediated spinal chloride homeostasis in a rat model of visceral hypersensitivity. Am J Physiol Gastrointest Liver Physiol 2015; 308:G767-78. [PMID: 25792562 DOI: 10.1152/ajpgi.00313.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 03/06/2015] [Indexed: 01/31/2023]
Abstract
Visceral hypersensitivity represents an important hallmark in the pathophysiology of irritable bowel syndrome (IBS), of which the mechanisms remain elusive. The present study was designed to examine whether cation-chloride cotransporter (CCC)-mediated chloride (Cl(-)) homeostasis of the spinal cord is involved in chronic stress-induced visceral hypersensitivity. Chronic visceral hypersensitivity was induced by exposing male Wistar rats to water avoidance stress (WAS). RT-PCR, Western blotting, and immunohistochemistry were used to assess the expression of CCCs in the spinal cord. Patch-clamp recordings were performed on adult spinal cord slices to evaluate Cl(-) homeostasis and Cl(-) extrusion capacity of lamina I neurons. Visceral sensitivity was estimated by measuring the abdominal withdrawal reflex in response to colorectal distension (CRD). After 10 days of WAS exposure, levels of both total protein and the oligomeric form of the K(+)-Cl(-) cotransporter isoform 2 (KCC2), but not Na(+)-K(+)-2Cl(-) transporter isoform 1 (NKCC1), were significantly decreased in the dorsal horn of the lumbosacral spinal cord. The downregulation of KCC2 resulted in a depolarizing shifted equilibrium potential of GABAergic inhibitory postsynaptic current and impaired Cl(-) extrusion capacity in lamina I neurons of the lumbosacral spinal cord from WAS rats. Acute noxious CRD disrupted spinal KCC2 expression and function 2 h after the final distention in sham rats, but not in WAS rats. Pharmacological blockade of KCC2 activity by intrathecal injection of a KCC2 inhibitor [(dihydroindenyl)oxy] alkanoic acid enhanced visceral nociceptive sensitivity in sham rats, but not in WAS rats. These results suggest that KCC2 downregulation-mediated impairment of spinal cord Cl(-) homeostasis may play an important role in chronic stress-induced visceral hypersensitivity.
Collapse
Affiliation(s)
- Dong Tang
- Department of Gastroenterology, Ruijin Hospital, Shanghai, China; and
| | - Ai-Hua Qian
- Department of Gastroenterology, Ruijin Hospital, Shanghai, China; and
| | - Dan-Dan Song
- Department of Gastroenterology, Ruijin Hospital, Shanghai, China; and
| | - Qi-Wen Ben
- Department of Gastroenterology, Ruijin Hospital, Shanghai, China; and
| | - Wei-Yan Yao
- Department of Gastroenterology, Ruijin Hospital, Shanghai, China; and
| | - Jing Sun
- Department of Gastroenterology, Ruijin Hospital, Shanghai, China; and
| | - Wei-Guang Li
- Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian-Le Xu
- Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao-Zong Yuan
- Department of Gastroenterology, Ruijin Hospital, Shanghai, China; and
| |
Collapse
|
42
|
Synaptic Inhibition and Disinhibition in the Spinal Dorsal Horn. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:359-83. [DOI: 10.1016/bs.pmbts.2014.11.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Grau JW, Huie JR, Lee KH, Hoy KC, Huang YJ, Turtle JD, Strain MM, Baumbauer KM, Miranda RM, Hook MA, Ferguson AR, Garraway SM. Metaplasticity and behavior: how training and inflammation affect plastic potential within the spinal cord and recovery after injury. Front Neural Circuits 2014; 8:100. [PMID: 25249941 PMCID: PMC4157609 DOI: 10.3389/fncir.2014.00100] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/31/2014] [Indexed: 12/30/2022] Open
Abstract
Research has shown that spinal circuits have the capacity to adapt in response to training, nociceptive stimulation and peripheral inflammation. These changes in neural function are mediated by physiological and neurochemical systems analogous to those that support plasticity within the hippocampus (e.g., long-term potentiation and the NMDA receptor). As observed in the hippocampus, engaging spinal circuits can have a lasting impact on plastic potential, enabling or inhibiting the capacity to learn. These effects are related to the concept of metaplasticity. Behavioral paradigms are described that induce metaplastic effects within the spinal cord. Uncontrollable/unpredictable stimulation, and peripheral inflammation, induce a form of maladaptive plasticity that inhibits spinal learning. Conversely, exposure to controllable or predictable stimulation engages a form of adaptive plasticity that counters these maladaptive effects and enables learning. Adaptive plasticity is tied to an up-regulation of brain derived neurotrophic factor (BDNF). Maladaptive plasticity is linked to processes that involve kappa opioids, the metabotropic glutamate (mGlu) receptor, glia, and the cytokine tumor necrosis factor (TNF). Uncontrollable nociceptive stimulation also impairs recovery after a spinal contusion injury and fosters the development of pain (allodynia). These adverse effects are related to an up-regulation of TNF and a down-regulation of BDNF and its receptor (TrkB). In the absence of injury, brain systems quell the sensitization of spinal circuits through descending serotonergic fibers and the serotonin 1A (5HT 1A) receptor. This protective effect is blocked by surgical anesthesia. Disconnected from the brain, intracellular Cl- concentrations increase (due to a down-regulation of the cotransporter KCC2), which causes GABA to have an excitatory effect. It is suggested that BDNF has a restorative effect because it up-regulates KCC2 and re-establishes GABA-mediated inhibition.
Collapse
Affiliation(s)
- James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - J Russell Huie
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | - Kuan H Lee
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Kevin C Hoy
- Department of Neurosciences, MetroHealth Medical Center and Case Western Reserve University Cleveland, OH, USA
| | - Yung-Jen Huang
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Joel D Turtle
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Misty M Strain
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | | | - Rajesh M Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center Bryan, TX, USA
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center Bryan, TX, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine Atlanta, GA, USA
| |
Collapse
|
44
|
Alessi DR, Zhang J, Khanna A, Hochdörfer T, Shang Y, Kahle KT. The WNK-SPAK/OSR1 pathway: master regulator of cation-chloride cotransporters. Sci Signal 2014; 7:re3. [PMID: 25028718 DOI: 10.1126/scisignal.2005365] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The WNK-SPAK/OSR1 kinase complex is composed of the kinases WNK (with no lysine) and SPAK (SPS1-related proline/alanine-rich kinase) or the SPAK homolog OSR1 (oxidative stress-responsive kinase 1). The WNK family senses changes in intracellular Cl(-) concentration, extracellular osmolarity, and cell volume and transduces this information to sodium (Na(+)), potassium (K(+)), and chloride (Cl(-)) cotransporters [collectively referred to as CCCs (cation-chloride cotransporters)] and ion channels to maintain cellular and organismal homeostasis and affect cellular morphology and behavior. Several genes encoding proteins in this pathway are mutated in human disease, and the cotransporters are targets of commonly used drugs. WNKs stimulate the kinases SPAK and OSR1, which directly phosphorylate and stimulate Cl(-)-importing, Na(+)-driven CCCs or inhibit the Cl(-)-extruding, K(+)-driven CCCs. These coordinated and reciprocal actions on the CCCs are triggered by an interaction between RFXV/I motifs within the WNKs and CCCs and a conserved carboxyl-terminal docking domain in SPAK and OSR1. This interaction site represents a potentially druggable node that could be more effective than targeting the cotransporters directly. In the kidney, WNK-SPAK/OSR1 inhibition decreases epithelial NaCl reabsorption and K(+) secretion to lower blood pressure while maintaining serum K(+). In neurons, WNK-SPAK/OSR1 inhibition could facilitate Cl(-) extrusion and promote γ-aminobutyric acidergic (GABAergic) inhibition. Such drugs could have efficacy as K(+)-sparing blood pressure-lowering agents in essential hypertension, nonaddictive analgesics in neuropathic pain, and promoters of GABAergic inhibition in diseases associated with neuronal hyperactivity, such as epilepsy, spasticity, neuropathic pain, schizophrenia, and autism.
Collapse
Affiliation(s)
- Dario R Alessi
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Jinwei Zhang
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Arjun Khanna
- Department of Neurosurgery, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Thomas Hochdörfer
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Yuze Shang
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02115, USA. Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|