1
|
Zheng YN, Zheng YL, Wang XQ, Chen PJ. Role of Exercise on Inflammation Cytokines of Neuropathic Pain in Animal Models. Mol Neurobiol 2024; 61:10288-10301. [PMID: 38714582 DOI: 10.1007/s12035-024-04214-4] [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: 11/20/2022] [Accepted: 04/25/2024] [Indexed: 05/10/2024]
Abstract
Neuropathic pain (NP) resulting from a lesion or disease of the somatosensory system can lead to loss of function and reduced life quality. Neuroinflammation plays a vital role in the development and maintenance of NP. Exercise as an economical, effective, and nonpharmacological treatment, recommended by clinical practice guidelines, has been proven to alleviate chronic NP. Previous studies have shown that exercise decreases NP by modifying inflammation; however, the exact mechanisms of exercise-mediated NP are unclear. Therefore, from the perspective of neuroinflammation, this review mainly discussed the effects of exercise on inflammatory cytokines in different parts of NP conduction pathways, such as the brain, spinal cord, dorsal root ganglion, sciatic nerve, and blood in rat/mice models. Results suggested that exercise training could modulate neuroinflammation, inhibit astrocyte glial cell proliferation and microglial activation, alter the macrophage phenotype, reduce the expression of proinflammatory cytokines, increase anti-inflammatory cytokine levels, and positively modulate the state of the immune system, thereby relieving NP.
Collapse
Affiliation(s)
- Ya-Nan Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
- Rehabilitation Treatment Center, The First Rehabilitation Hospital of Shanghai, Shanghai, 200090, China
| | - Yi-Li Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Xue-Qiang Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Pei-Jie Chen
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China.
| |
Collapse
|
2
|
He WC, Hou SL, Wang KB, Xu N, Li K, Xiong T, Luo J. Treadmill running on neuropathic pain: via modulation of neuroinflammation. Front Mol Neurosci 2024; 17:1345864. [PMID: 38989156 PMCID: PMC11233809 DOI: 10.3389/fnmol.2024.1345864] [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: 11/28/2023] [Accepted: 05/24/2024] [Indexed: 07/12/2024] Open
Abstract
Neuropathic pain is a type of chronic pain caused by an injury or somatosensory nervous system disease. Drugs and exercise could effectively relieve neuropathic pain, but no treatment can completely stop neuropathic pain. The integration of exercise into neuropathic pain management has attracted considerable interest in recent years, and treadmill training is the most used among exercise therapies. Neuropathic pain can be effectively treated if its mechanism is clarified. In recent years, the association between neuroinflammation and neuropathic pain has been explored. Neuroinflammation can trigger proinflammatory cytokines, activate microglia, inhibit descending pain modulatory systems, and promote the overexpression of brain-derived neurotrophic factor, which lead to the generation of neuropathic pain and hypersensitivity. Treadmill exercise can alleviate neuropathic pain mainly by regulating neuroinflammation, including inhibiting the activity of pro-inflammatory factors and over activation of microglia in the dorsal horn, regulating the expression of mu opioid receptor expression in the rostral ventromedial medulla and levels of γ-aminobutyric acid to activate the descending pain modulatory system and the overexpression of brain-derived neurotrophic factor. This article reviews and summarizes research on the effect of treadmill exercise on neuropathic pain and its role in the regulation of neuroinflammation to explore its benefits for neuropathic pain treatment.
Collapse
Affiliation(s)
- Wei-Chun He
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Shuang-Long Hou
- Department of Sport Rehabilitation, Xi’an Physical Education University, Xi’an, China
| | - Kai-Bin Wang
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Ning Xu
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Ke Li
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Ting Xiong
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
| | - Jing Luo
- Department of Rehabilitation Medicine, General Hospital of NingXia Medical University, Yinchuan, China
- Department of Sport Rehabilitation, Xi’an Physical Education University, Xi’an, China
| |
Collapse
|
3
|
Sliwinski C, Heutehaus L, Taberner FJ, Weiss L, Kampanis V, Tolou-Dabbaghian B, Cheng X, Motsch M, Heppenstall PA, Kuner R, Franz S, Lechner SG, Weidner N, Puttagunta R. Contribution of mechanoreceptors to spinal cord injury-induced mechanical allodynia. Pain 2024; 165:1336-1347. [PMID: 38739766 PMCID: PMC11090032 DOI: 10.1097/j.pain.0000000000003139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/29/2023] [Accepted: 10/27/2023] [Indexed: 05/16/2024]
Abstract
ABSTRACT Evidence from previous studies supports the concept that spinal cord injury (SCI)-induced neuropathic pain (NP) has its neural roots in the peripheral nervous system. There is uncertainty about how and to which degree mechanoreceptors contribute. Sensorimotor activation-based interventions (eg, treadmill training) have been shown to reduce NP after experimental SCI, suggesting transmission of pain-alleviating signals through mechanoreceptors. The aim of the present study was to understand the contribution of mechanoreceptors with respect to mechanical allodynia in a moderate mouse contusion SCI model. After genetic ablation of tropomyosin receptor kinase B expressing mechanoreceptors before SCI, mechanical allodynia was reduced. The identical genetic ablation after SCI did not yield any change in pain behavior. Peptidergic nociceptor sprouting into lamina III/IV below injury level as a consequence of SCI was not altered by either mechanoreceptor ablation. However, skin-nerve preparations of contusion SCI mice 7 days after injury yielded hyperexcitability in nociceptors, not in mechanoreceptors, which makes a substantial direct contribution of mechanoreceptors to NP maintenance unlikely. Complementing animal data, quantitative sensory testing in human SCI subjects indicated reduced mechanical pain thresholds, whereas the mechanical detection threshold was not altered. Taken together, early mechanoreceptor ablation modulates pain behavior, most likely through indirect mechanisms. Hyperexcitable nociceptors seem to be the main drivers of SCI-induced NP. Future studies need to focus on injury-derived factors triggering early-onset nociceptor hyperexcitability, which could serve as targets for more effective therapeutic interventions.
Collapse
Affiliation(s)
- Christopher Sliwinski
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Laura Heutehaus
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Lisa Weiss
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Vasileios Kampanis
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Bahardokht Tolou-Dabbaghian
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Xing Cheng
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie Motsch
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Steffen Franz
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan G. Lechner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
4
|
Chen HH, Mohsin M, Ge JY, Feng YT, Wang JG, Ou YS, Jiang ZJ, Hu BY, Liu XJ. Optogenetic Activation of Peripheral Somatosensory Neurons in Transgenic Mice as a Neuropathic Pain Model for Assessing the Therapeutic Efficacy of Analgesics. ACS Pharmacol Transl Sci 2024; 7:236-248. [PMID: 38230281 PMCID: PMC10789130 DOI: 10.1021/acsptsci.3c00254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 01/18/2024]
Abstract
Optogenetics is a novel biotechnology widely used to precisely manipulate a specific peripheral sensory neuron or neural circuit. However, the use of optogenetics to assess the therapeutic efficacy of analgesics is elusive. In this study, we generated a transgenic mouse stain in which all primary somatosensory neurons can be optogenetically activated to mimic neuronal hyperactivation in the neuropathic pain state for the assessment of analgesic effects of drugs. A transgenic mouse was generated using the advillin-Cre line mated with the Ai32 strain, in which channelrhodopsin-2 fused to enhanced yellow fluorescence protein (ChR2-EYFP) was conditionally expressed in all types of primary somatosensory neurons (advillincre/ChR2+/+). Immunofluorescence and transdermal photostimulation on the hindpaws were used to verify the transgenic mice. Optical stimulation to evoke pain-like paw withdrawal latency was used to assess the analgesic effects of a series of drugs. Injury- and pain-related molecular biomarkers were investigated with immunohistofluorescence. We found that the expression of ChR2-EYFP was observed in many primary afferents of paw skin and sciatic nerves and in primary sensory neurons and laminae I and II of the spinal dorsal horns in advillincre/ChR2+/+ mice. Transdermal blue light stimulation of the transgenic mouse hindpaw evoked nocifensive paw withdrawal behavior. Treatment with gabapentin, some channel blockers, and local anesthetics, but not opioids or COX-1/2 inhibitors, prolonged the paw withdrawal latency in the transgenic mice. The analgesic effect of gabapentin was also verified by the decreased expression of injury- and pain-related molecular biomarkers. These optogenetic mice provide a promising model for assessing the therapeutic efficacy of analgesics in neuropathic pain.
Collapse
Affiliation(s)
- Hao-Hao Chen
- School
of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
- Pain
and Related Diseases Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Muhammad Mohsin
- Pain
and Related Diseases Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Jia-Yi Ge
- School
of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yu-Ting Feng
- School
of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jing-Ge Wang
- School
of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yu-Sen Ou
- Pain
and Related Diseases Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Zuo-Jie Jiang
- Pain
and Related Diseases Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Bo-Ya Hu
- Pain
and Related Diseases Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Xing-Jun Liu
- School
of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China
- Pain
and Related Diseases Research Laboratory, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| |
Collapse
|
5
|
Toloui A, Ramawad HA, Gharin P, Vaccaro AR, Zarei H, Hosseini M, Yousefifard M, Rahimi-Movaghar V. The Role of Exercise in the Alleviation of Neuropathic Pain Following Traumatic Spinal Cord Injuries: A Systematic Review and Meta-analysis. Neurospine 2023; 20:1073-1087. [PMID: 37798999 PMCID: PMC10562228 DOI: 10.14245/ns.2346588.294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 10/07/2023] Open
Abstract
OBJECTIVE The objective of this systematic review and meta-analysis was to assess the efficacy of exercise in neuropathic pain following traumatic spinal cord injuries. METHODS The search was conducted in MEDLINE, Embase, Scopus, and Web of Science by the end of 2022. Two independent researchers included the articles based on the inclusion and exclusion criteria. A standardized mean difference was calculated for each data and they were pooled to calculate an overall effect size. To assess the heterogeneity between studies, I2 and chi-square tests were utilized. In the case of heterogeneity, meta-regression was performed to identify the potential source. RESULTS Fifteen preclinical studies were included. Meta-analysis demonstrated that exercise significantly improves mechanical allodynia (standardized mean difference [SMD], -1.59; 95% confidence interval [CI], -2.16 to -1.02; p < 0.001; I2 = 90.37%), thermal hyperalgesia (SMD, 1.95; 95% CI, 0.96-2.94; p < 0.001), and cold allodynia (SMD, -2.92; 95% CI, -4.4 to -1.43; p < 0.001). The improvement in mechanical allodynia is significantly more in animals with a compression model of SCI (meta-regression coefficient, -1.33; 95% CI, -1.84 to -0.57; p < 0.001) and in mild SCI (p < 0.001). Additionally, the improvement was more prominent if the training was started 7 to 8 days postinjury (coefficient, -2.54; 95% CI, -3.85 to -1.23; p < 0.001) and was continued every day (coefficient, -1.99; 95% CI, -3.07 to -0.9; p < 0.001). Likewise, voluntary exercise demonstrated a significantly more effect size (coefficient, -1.45; 95% CI, -2.67 to -0.23; p = 0.02). CONCLUSION Exercise is effective in the amelioration of neuropathic pain. This effect in mechanical allodynia is more prominent if voluntary, continuous training is initiated in the subacute phase of mild SCI.
Collapse
Affiliation(s)
- Amirmohammad Toloui
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamzah Adel Ramawad
- Department of Emergency Medicine, NYC Health + Hospitals, Coney Island, New York, NY, USA
| | - Pantea Gharin
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alexander R. Vaccaro
- Department of Orthopedics and Neurosurgery, Rothman Institute, Thomas Jefferson University, Philadelphia, PA, USA
| | - Hamed Zarei
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Injuries Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Nees TA, Wang N, Adamek P, Zeitzschel N, Verkest C, La Porta C, Schaefer I, Virnich J, Balkaya S, Prato V, Morelli C, Begay V, Lee YJ, Tappe-Theodor A, Lewin GR, Heppenstall PA, Taberner FJ, Lechner SG. Role of TMEM100 in mechanically insensitive nociceptor un-silencing. Nat Commun 2023; 14:1899. [PMID: 37019973 PMCID: PMC10076432 DOI: 10.1038/s41467-023-37602-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Mechanically silent nociceptors are sensory afferents that are insensitive to noxious mechanical stimuli under normal conditions but become sensitized to such stimuli during inflammation. Using RNA-sequencing and quantitative RT-PCR we demonstrate that inflammation upregulates the expression of the transmembrane protein TMEM100 in silent nociceptors and electrophysiology revealed that over-expression of TMEM100 is required and sufficient to un-silence silent nociceptors in mice. Moreover, we show that mice lacking TMEM100 do not develop secondary mechanical hypersensitivity-i.e., pain hypersensitivity that spreads beyond the site of inflammation-during knee joint inflammation and that AAV-mediated overexpression of TMEM100 in articular afferents in the absence of inflammation is sufficient to induce mechanical hypersensitivity in remote skin regions without causing knee joint pain. Thus, our work identifies TMEM100 as a key regulator of silent nociceptor un-silencing and reveals a physiological role for this hitherto enigmatic afferent subclass in triggering spatially remote secondary mechanical hypersensitivity during inflammation.
Collapse
Affiliation(s)
- Timo A Nees
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department for Orthopeadics, Heidelberg University Hospital, Heidelberg, Germany
| | - Na Wang
- Institute of Pathophysiology, Yan'an University, Yan'an, China
| | - Pavel Adamek
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nadja Zeitzschel
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clement Verkest
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carmen La Porta
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Irina Schaefer
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Julie Virnich
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Selin Balkaya
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vincenzo Prato
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Chiara Morelli
- SISSA: Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy
| | - Valerie Begay
- Department of Neuroscience, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Young Jae Lee
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, Republic of Korea
| | | | - Gary R Lewin
- Department of Neuroscience, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Paul A Heppenstall
- SISSA: Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy
| | - Francisco J Taberner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Instituto de Neurosciencias de Alicante, Universidad Miguel Hernández - CSIC, Alicante, Spain
| | - Stefan G Lechner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
7
|
Moderate-Intensity Treadmill Exercise Promotes mTOR-Dependent Motor Cortical Neurotrophic Factor Expression and Functional Recovery in a Murine Model of Crush Spinal Cord Injury (SCI). Mol Neurobiol 2023; 60:960-978. [PMID: 36385234 DOI: 10.1007/s12035-022-03117-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022]
Abstract
Treadmill exercise is widely considered an effective strategy for restoration of skilled motor function after spinal cord injury (SCI). However, the specific exercise intensity that optimizes recovery and the underlying mechanistic basis of this recovery remain unclear. To that end, we sought to investigate the effect of different treadmill exercise intensities on cortical mTOR activity, a key regulator of functional recovery following CNS trauma, in an animal model of C5 crush spinal cord injury (SCI). Following injury, animals were subjected to treadmill exercise for 4 consecutive weeks at three different intensities (low intensity [LEI]; moderate intensity [MEI]; and high intensity [HEI]). Motor function recovery was assessed by horizontal ladder test, cylinder rearing test, and electrophysiology, while neurotrophic factors and cortical mechanistic target of rapamycin (mTOR) pathway-related proteins were assessed by Western blotting. The activation of the cortical mTOR pathway and axonal sprouting was evaluated by immunofluorescence and the changes of plasticity in motor cortex neurons were assessed by Golgi staining. In keeping with previous studies, we found that 4 weeks of treadmill training resulted in improved skilled motor function, enhanced nerve conduction capability, increased neuroplasticity, and axonal sprouting. Importantly, we also demonstrated that when compared with the LEI group, MEI and HEI groups demonstrated elevated expression of brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), phosphorylated ribosomal S6 protein (p-S6), and protein kinase B (p-Akt), consistent with an intensity-dependent activation of the mTOR pathway and neurotrophic factor expression in the motor cortex. We also observed impaired exercise endurance and higher mortality during training in the HEI group than in the LEI and MEI groups. Collectively, our findings suggest that treadmill exercise following SCI is an effective means of promoting recovery and highlight the importance of the cortical mTOR pathway and neurotrophic factors as mediators of this effect. Importantly, our findings also demonstrate that excessive exercise can be detrimental, suggesting that moderation may be the optimal strategy. These findings provide an important foundation for further investigation of treadmill training as a modality for recovery following spinal cord injury and of the underlying mechanisms.
Collapse
|
8
|
Jergova S, Dugan EA, Sagen J. Attenuation of SCI-Induced Hypersensitivity by Intensive Locomotor Training and Recombinant GABAergic Cells. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010084. [PMID: 36671656 PMCID: PMC9854592 DOI: 10.3390/bioengineering10010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
The underlying mechanisms of spinal cord injury (SCI)-induced chronic pain involve dysfunctional GABAergic signaling and enhanced NMDA signaling. Our previous studies showed that SCI hypersensitivity in rats can be attenuated by recombinant rat GABAergic cells releasing NMDA blocker serine-histogranin (SHG) and by intensive locomotor training (ILT). The current study combines these approaches and evaluates their analgesic effects on a model of SCI pain in rats. Cells were grafted into the spinal cord at 4 weeks post-SCI to target the chronic pain, and ILT was initiated 5 weeks post-SCI. The hypersensitivity was evaluated weekly, which was followed by histological and biochemical assays. Prolonged effects of the treatment were evaluated in subgroups of animals after we discontinued ILT. The results show attenuation of tactile, heat and cold hypersensitivity in all of the treated animals and reduced levels of proinflammatory cytokines IL1β and TNFα in the spinal tissue and CSF. Animals with recombinant grafts and ILT showed the preservation of analgesic effects even during sedentary periods when the ILT was discontinued. Retraining helped to re-establish the effect of long-term training in all of the groups, with the greatest impact being in animals with recombinant grafts. These findings suggest that intermittent training in combination with cell therapy might be an efficient approach to manage chronic pain in SCI patients.
Collapse
|
9
|
Franz S, Heutehaus L, Tappe-Theodor A, Weidner N, Treede RD, Schuh-Hofer S. Noxious radiant heat evokes bi-component nociceptive withdrawal reflexes in spinal cord injured humans-A clinical tool to study neuroplastic changes of spinal neural circuits. Front Hum Neurosci 2023; 17:1141690. [PMID: 37200949 PMCID: PMC10185789 DOI: 10.3389/fnhum.2023.1141690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/04/2023] [Indexed: 05/20/2023] Open
Abstract
Investigating nocifensive withdrawal reflexes as potential surrogate marker for the spinal excitation level may widen the understanding of maladaptive nociceptive processing after spinal cord injury (SCI). The aim of this prospective, explorative cross-sectional observational study was to investigate the response behavior of individuals with SCI to noxious radiant heat (laser) stimuli and to assess its relation to spasticity and neuropathic pain, two clinical consequences of spinal hyperexcitability/spinal disinhibition. Laser stimuli were applied at the sole and dorsum of the foot and below the fibula head. Corresponding reflexes were electromyography (EMG) recorded ipsilateral. Motor responses to laser stimuli were analyzed and related to clinical readouts (severity of injury/spasticity/pain), using established clinical assessment tools. Twenty-seven participants, 15 with SCI (age 18-63; 6.5 years post-injury; AIS-A through D) and 12 non-disabled controls, [non-disabled controls (NDC); age 19-63] were included. The percentage of individuals with SCI responding to stimuli (70-77%; p < 0.001), their response rates (16-21%; p < 0.05) and their reflex magnitude (p < 0.05) were significantly higher compared to NDC. SCI-related reflexes clustered in two time-windows, indicating involvement of both A-delta- and C-fibers. Spasticity was associated with facilitated reflexes in SCI (Kendall-tau-b p ≤ 0.05) and inversely associated with the occurrence/severity of neuropathic pain (Fisher's exact p < 0.05; Eta-coefficient p < 0.05). However, neuropathic pain was not related to reflex behavior. Altogether, we found a bi-component motor hyperresponsiveness of SCI to noxious heat, which correlated with spasticity, but not neuropathic pain. Laser-evoked withdrawal reflexes may become a suitable outcome parameter to explore maladaptive spinal circuitries in SCI and to assess the effect of targeted treatment strategies. Registration: https://drks.de/search/de/trial/DRKS00006779.
Collapse
Affiliation(s)
- Steffen Franz
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
- Steffen Franz,
| | - Laura Heutehaus
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Anke Tappe-Theodor
- Department of Molecular Pharmacology, Medical Faculty Heidelberg, Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Rolf-Detlef Treede
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
| | - Sigrid Schuh-Hofer
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Ruprecht-Karls-University of Heidelberg, Mannheim, Germany
- Department of Neurology and Epileptology, University of Tübingen, Tübingen, Germany
- *Correspondence: Sigrid Schuh-Hofer,
| |
Collapse
|
10
|
Saruta K, Fukutoku T, Kumagai G, Nagaoki T, Tsukuda M, Nitobe Y, Wada K, Asari T, Fujita T, Sasaki I, Nikaido Y, Shimoyama S, Ueno S, Ishibashi Y. Intraperitoneal Administration of Etizolam Improves Locomotor Function in Mice After Spinal Cord Injury. Neurotrauma Rep 2023; 4:82-96. [PMID: 36874147 PMCID: PMC9983139 DOI: 10.1089/neur.2022.0071] [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] [Indexed: 02/24/2023] Open
Abstract
Neuroinflammation occurs in the acute phase of spinal cord injury (SCI) and inhibits neural regeneration. In mouse models, etizolam (ETZ) is a strong anxiolytic with unclear effects on SCI. This study investigated the effects of short-term administration of ETZ on neuroinflammation and behavior in mice after SCI. We administrated an ETZ (0.5 mg/kg) daily intraperitoneal injection from the day after SCI for 7 days. Mice were randomly divided into three groups (sham group: only laminectomy, saline group, and ETZ group). Inflammatory cytokine concentrations in the injured spinal cord epicenter were measured using an enzyme-linked immunosorbent assay on day 7 after SCI to evaluate spinal cord inflammation in the acute phase. Behavior analysis was performed the day before surgery and on days 7, 14, 28, and 42 after surgery. The behavioral analysis included anxiety-like behavior using the open field test, locomotor function using the Basso Mouse Scale, and sensory function using the mechanical and heat test. Inflammatory cytokine concentrations were significantly lower in the ETZ group than in the saline group in the acute phase after spinal surgery. After SCI, anxiety-like behaviors and sensory functions were comparable between the ETZ and saline groups. ETZ administration reduced neuroinflammation in the spinal cord and improved locomotor function. Gamma-amino butyric acid type A receptor stimulants may be effective therapeutic agents for patients with SCI.
Collapse
Affiliation(s)
- Kenya Saruta
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tatsuhiro Fukutoku
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Gentaro Kumagai
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Toshihide Nagaoki
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Manami Tsukuda
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yohshiro Nitobe
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kanichiro Wada
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Toru Asari
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Taku Fujita
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Isamu Sasaki
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshikazu Nikaido
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.,Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shuji Shimoyama
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shinya Ueno
- Department of Neurophysiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasuyuki Ishibashi
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
11
|
The Impact of Activity-Based Interventions on Neuropathic Pain in Experimental Spinal Cord Injury. Cells 2022; 11:cells11193087. [PMID: 36231048 PMCID: PMC9563089 DOI: 10.3390/cells11193087] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Physical activity-based rehabilitative interventions represent the main treatment concept for people suffering from spinal cord injury (SCI). The role such interventions play in the relief of neuropathic pain (NP) states is emerging, along with underlying mechanisms resulting in SCI-induced NP (SCI-NP). Animal models have been used to investigate the benefits of activity-based interventions (ABI), such as treadmill training, wheel running, walking, swimming, and bipedal standing. These activity-based paradigms have been shown to modulate inflammatory-related alterations as well as induce functional and structural changes in the spinal cord gray matter circuitry correlated with pain behaviors. Thus far, the research available provides an incomplete picture of the cellular and molecular pathways involved in this beneficial effect. Continued research is essential for understanding how such interventions benefit SCI patients suffering from NP and allow the development of individualized rehabilitative therapies. This article reviews preclinical studies on this specific topic, goes over mechanisms involved in SCI-NP in relation to ABI, and then discusses the effectiveness of different activity-based paradigms as they relate to different forms, intensity, initiation times, and duration of ABI. This article also summarizes the mechanisms of respective interventions to ameliorate NP after SCI and provides suggestions for future research directions.
Collapse
|
12
|
Eller OC, Willits AB, Young EE, Baumbauer KM. Pharmacological and non-pharmacological therapeutic interventions for the treatment of spinal cord injury-induced pain. FRONTIERS IN PAIN RESEARCH 2022; 3:991736. [PMID: 36093389 PMCID: PMC9448954 DOI: 10.3389/fpain.2022.991736] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022] Open
Abstract
Spinal cord injury (SCI) is a complex neurophysiological disorder, which can result in many long-term complications including changes in mobility, bowel and bladder function, cardiovascular function, and metabolism. In addition, most individuals with SCI experience some form of chronic pain, with one-third of these individuals rating their pain as severe and unrelenting. SCI-induced chronic pain is considered to be "high impact" and broadly affects a number of outcome measures, including daily activity, physical and cognitive function, mood, sleep, and overall quality of life. The majority of SCI pain patients suffer from pain that emanates from regions located below the level of injury. This pain is often rated as the most severe and the underlying mechanisms involve injury-induced plasticity along the entire neuraxis and within the peripheral nervous system. Unfortunately, current therapies for SCI-induced chronic pain lack universal efficacy. Pharmacological treatments, such as opioids, anticonvulsants, and antidepressants, have been shown to have limited success in promoting pain relief. In addition, these treatments are accompanied by many adverse events and safety issues that compound existing functional deficits in the spinally injured, such as gastrointestinal motility and respiration. Non-pharmacological treatments are safer alternatives that can be specifically tailored to the individual and used in tandem with pharmacological therapies if needed. This review describes existing non-pharmacological therapies that have been used to treat SCI-induced pain in both preclinical models and clinical populations. These include physical (i.e., exercise, acupuncture, and hyper- or hypothermia treatments), psychological (i.e., meditation and cognitive behavioral therapy), and dietary interventions (i.e., ketogenic and anti-inflammatory diet). Findings on the effectiveness of these interventions in reducing SCI-induced pain and improving quality of life are discussed. Overall, although studies suggest non-pharmacological treatments could be beneficial in reducing SCI-induced chronic pain, further research is needed. Additionally, because chronic pain, including SCI pain, is complex and has both emotional and physiological components, treatment should be multidisciplinary in nature and ideally tailored specifically to the patient.
Collapse
Affiliation(s)
- Olivia C. Eller
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Adam B. Willits
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Erin E. Young
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Kyle M. Baumbauer
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
13
|
Cheng X, Yu Z, Hu W, Chen J, Chen W, Wang L, Li X, Zhang W, Chen J, Zou X, Chen W, Wan Y. Voluntary exercise ameliorates neuropathic pain by suppressing calcitonin gene-related peptide and ionized calcium-binding adapter molecule 1 overexpression in the lumbar dorsal horns in response to injury to the cervical spinal cord. Exp Neurol 2022; 354:114105. [PMID: 35525308 DOI: 10.1016/j.expneurol.2022.114105] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Neuropathic pain (NP) is a frequent finding in patients diagnosed with spinal cord injuries (SCIs). To improve our understanding of the maladaptive changes taking place in the lumbar spinal cord that can lead to the development of NP and to find alternative options to treat this condition, we aimed to investigate the effects of voluntary exercise on NP after SCI and to elucidate its potential mechanisms. METHODS A rat model of post-SCI NP induced by compression of the posterior or lateral cervical spinal cord was used to evaluate the effects of voluntary exercise by measuring the bilateral withdrawal of the hind paws using the Von Frey filament and Hargreaves tests. The place escape/avoid paradigm was used to evaluate supraspinal pain processing and somatosensory evoked potentials (SEPs) were used to examine disturbances in proprioception. Locomotor function was evaluated using Basso, Beattie, and Bresnahan (BBB) scoring. Pathologic findings in hematoxylin and eosin-stained tissue and magnetic resonance imaging were used to evaluate the morphological changes after SCI. The lesion size within the cervical spinal cord was evaluated by staining with Eriochrome cyanine R. Quantitative polymerase chain reaction and immunohistochemistry were used to assess the expression of calcitonin gene-related peptide (CGRP) and ionized calcium-binding adapter molecule 1 (Iba-1) in the lumbar dorsal horns. RESULTS All injured rats developed mechanical hypersensitivity, hyposensitivity, and thermal hyperalgesia in the contralateral hind paws at 1 week post-injury. Rats that underwent lateral compression injury developed NP in the ipsilateral hind paws 1 week later than rats with a posterior compression injury. Our findings revealed that voluntary exercise ameliorated mechanical allodynia and thermal hyperalgesia, and significantly improved proprioception as measured by SEP, but had no impact on mechanical hypoalgesia or motor recovery and provided no significant neuroprotection after recovery from an acute SCI. SCI-induced NP was accompanied by increased expression of CGRP and Iba-1 in the lumbar dorsal horn. These responses were reduced in rats that underwent voluntary exercise. CONCLUSIONS Voluntary exercise ameliorates NP that develops in rats after compression injury. Increased expression of CGRP and Iba-1 in the lumbar dorsal horns of rats exhibiting symptoms of NP suggests that microglial activation might play a crucial role in its development. Collectively, voluntary exercise may be a promising therapeutic modality to treat NP that develops clinically in response to SCI.
Collapse
Affiliation(s)
- Xing Cheng
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Zhengran Yu
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Wenjie Hu
- Department of Rehabilitation Medicine, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jiacheng Chen
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China; Graduate School of Biomedical Engineering, Institute of Engineering, University of New South Wales, Sydney 1001, Australia
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Le Wang
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Xiang Li
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Wenwu Zhang
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Jiewen Chen
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Xuenong Zou
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Wenli Chen
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.
| | - Yong Wan
- Department of Spine Surgery, Orthopedic Research Institute, The First Affiliated Hospital of Sun Yat-sen University; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China.
| |
Collapse
|
14
|
Dietz V, Knox K, Moore S, Roberts N, Corona KK, Dulin JN. Dorsal horn neuronal sparing predicts the development of at-level mechanical allodynia following cervical spinal cord injury in mice. Exp Neurol 2022; 352:114048. [PMID: 35304102 DOI: 10.1016/j.expneurol.2022.114048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/03/2022] [Accepted: 03/13/2022] [Indexed: 11/04/2022]
Abstract
Spinal cord injury (SCI) frequently results in immediate and sustained neurological dysfunction, including intractable neuropathic pain in approximately 60-80% of individuals. SCI induces immediate mechanical damage to spinal cord tissue followed by a period of secondary injury in which tissue damage is further propagated, contributing to the development of anatomically unique lesions. Variability in lesion size and location influences the degree of motor and sensory dysfunction incurred by an individual. We predicted that variability in lesion parameters may also explain why some, but not all, experimental animals develop mechanical sensitivity after SCI. To characterize the relationship of lesion anatomy to mechanical allodynia, we utilized a mouse cervical hemicontusion model of SCI that has been shown to lead to the development and persistence of mechanical allodynia in the ipsilateral forelimb after injury. At four weeks post-SCI, the numbers and locations of surviving neurons were quantified along with total lesion volume and nociceptive fiber sprouting. We found that the subset of animals exhibiting mechanical allodynia had significantly increased neuronal sparing in the ipsilateral dorsal horn around the lesion epicenter compared to animals that did not exhibit mechanical allodynia. Additionally, we failed to observe significant differences between groups in nociceptive fiber density in the dorsal horn around the lesion epicenter. Notably, we found that impactor probe displacement upon administration of the SCI surgery was significantly lower in sensitive animals compared with not-sensitive animals. Together, our data indicate that lesion severity negatively correlates with the manifestation of at-level mechanical hypersensitivity and suggests that sparing of dorsal horn neurons may be required for the development of neuropathic pain.
Collapse
Affiliation(s)
- Valerie Dietz
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Katelyn Knox
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Sherilynne Moore
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Nolan Roberts
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | | | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
15
|
Naro A, Billeri L, Balletta T, Lauria P, Onesta MP, Calabrò RS. Finding the Way to Improve Motor Recovery of Patients with Spinal Cord Lesions: A Case-Control Pilot Study on a Novel Neuromodulation Approach. Brain Sci 2022; 12:119. [PMID: 35053862 PMCID: PMC8773706 DOI: 10.3390/brainsci12010119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
Robot-assisted rehabilitation (RAR) and non-invasive brain stimulation (NIBS) are interventions that, both individually and combined, can significantly enhance motor performance after spinal cord injury (SCI). We sought to determine whether repetitive transcranial magnetic stimulation (rTMS) combined with active transvertebral direct current stimulation (tvDCS) (namely, NIBS) in association with RAR (RAR + NIBS) improves lower extremity motor function more than RAR alone in subjects with motor incomplete SCI (iSCI). Fifteen adults with iSCI received one daily session of RAR+NIBS in the early afternoon, six sessions weekly, for eight consecutive weeks. Outcome measures included the 6 min walk test (6MWT), the 10 m walk test (10MWT), the timed up and go (TUG) to test mobility and balance, the Walking Index for Spinal Cord Injury (WISCI II), the Functional Independence Measure-Locomotion (FIM-L), the manual muscle testing for lower extremity motor score (LEMS), the modified Ashworth scale for lower limbs (MAS), and the visual analog scale (VAS) for pain. The data of these subjects were compared with those of 20 individuals matched for clinical and demographic features who previously received the same amount or RAR without NIBS (RAR - NIBS). All patients completed the trial, and none reported any side effects either during or following the training. The 10MWT improved in both groups, but the increase was significantly greater following RAR + NIBS than RAR - NIBS. The same occurred for the FIM-L, LEMS, and WISCI II. No significant differences were appreciable concerning the 6MWT and TUG. Conversely, RAR - NIBS outperformed RAR + NIBS regarding the MAS and VAS. Pairing tvDCS with rTMS during RAR can improve lower extremity motor function more than RAR alone can do. Future research with a larger sample size is recommended to determine longer-term effects on motor function and activities of daily living.
Collapse
Affiliation(s)
- Antonino Naro
- IRCCS Centro Neurolesi Bonino Pulejo Piemonte, Via Palermo, SS 113, Ctr. Casazza, 98124 Messina, Italy; (A.N.); (L.B.); (T.B.); (P.L.)
| | - Luana Billeri
- IRCCS Centro Neurolesi Bonino Pulejo Piemonte, Via Palermo, SS 113, Ctr. Casazza, 98124 Messina, Italy; (A.N.); (L.B.); (T.B.); (P.L.)
| | - Tina Balletta
- IRCCS Centro Neurolesi Bonino Pulejo Piemonte, Via Palermo, SS 113, Ctr. Casazza, 98124 Messina, Italy; (A.N.); (L.B.); (T.B.); (P.L.)
| | - Paola Lauria
- IRCCS Centro Neurolesi Bonino Pulejo Piemonte, Via Palermo, SS 113, Ctr. Casazza, 98124 Messina, Italy; (A.N.); (L.B.); (T.B.); (P.L.)
| | | | - Rocco Salvatore Calabrò
- IRCCS Centro Neurolesi Bonino Pulejo Piemonte, Via Palermo, SS 113, Ctr. Casazza, 98124 Messina, Italy; (A.N.); (L.B.); (T.B.); (P.L.)
| |
Collapse
|
16
|
Verdú E, Homs J, Boadas-Vaello P. Physiological Changes and Pathological Pain Associated with Sedentary Lifestyle-Induced Body Systems Fat Accumulation and Their Modulation by Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:13333. [PMID: 34948944 PMCID: PMC8705491 DOI: 10.3390/ijerph182413333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
A sedentary lifestyle is associated with overweight/obesity, which involves excessive fat body accumulation, triggering structural and functional changes in tissues, organs, and body systems. Research shows that this fat accumulation is responsible for several comorbidities, including cardiovascular, gastrointestinal, and metabolic dysfunctions, as well as pathological pain behaviors. These health concerns are related to the crosstalk between adipose tissue and body systems, leading to pathophysiological changes to the latter. To deal with these health issues, it has been suggested that physical exercise may reverse part of these obesity-related pathologies by modulating the cross talk between the adipose tissue and body systems. In this context, this review was carried out to provide knowledge about (i) the structural and functional changes in tissues, organs, and body systems from accumulation of fat in obesity, emphasizing the crosstalk between fat and body tissues; (ii) the crosstalk between fat and body tissues triggering pain; and (iii) the effects of physical exercise on body tissues and organs in obese and non-obese subjects, and their impact on pathological pain. This information may help one to better understand this crosstalk and the factors involved, and it could be useful in designing more specific training interventions (according to the nature of the comorbidity).
Collapse
Affiliation(s)
- Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| | - Judit Homs
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
- Department of Physical Therapy, EUSES-University of Girona, 17190 Salt, Spain
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| |
Collapse
|
17
|
Walker JR, Detloff MR. Plasticity in Cervical Motor Circuits following Spinal Cord Injury and Rehabilitation. BIOLOGY 2021; 10:biology10100976. [PMID: 34681075 PMCID: PMC8533179 DOI: 10.3390/biology10100976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Spinal cord injury results in a decreased quality of life and impacts hundreds of thousands of people in the US alone. This review discusses the underlying cellular mechanisms of injury and the concurrent therapeutic hurdles that impede recovery. It then describes the phenomena of neural plasticity—the nervous system’s ability to change. The primary focus of the review is on the impact of cervical spinal cord injury on control of the upper limbs. The neural plasticity that occurs without intervention is discussed, which shows new connections growing around the injury site and the involvement of compensatory movements. Rehabilitation-driven neural plasticity is shown to have the ability to guide connections to create more normal functions. Various novel stimulation and recording technologies are outlined for their role in further improving rehabilitative outcomes and gains in independence. Finally, the importance of sensory input, an often-overlooked aspect of motor control, is shown in driving neural plasticity. Overall, this review seeks to delineate the historical and contemporary research into neural plasticity following injury and rehabilitation to guide future studies. Abstract Neuroplasticity is a robust mechanism by which the central nervous system attempts to adapt to a structural or chemical disruption of functional connections between neurons. Mechanical damage from spinal cord injury potentiates via neuroinflammation and can cause aberrant changes in neural circuitry known as maladaptive plasticity. Together, these alterations greatly diminish function and quality of life. This review discusses contemporary efforts to harness neuroplasticity through rehabilitation and neuromodulation to restore function with a focus on motor recovery following cervical spinal cord injury. Background information on the general mechanisms of plasticity and long-term potentiation of the nervous system, most well studied in the learning and memory fields, will be reviewed. Spontaneous plasticity of the nervous system, both maladaptive and during natural recovery following spinal cord injury is outlined to provide a baseline from which rehabilitation builds. Previous research has focused on the impact of descending motor commands in driving spinal plasticity. However, this review focuses on the influence of physical therapy and primary afferent input and interneuron modulation in driving plasticity within the spinal cord. Finally, future directions into previously untargeted primary afferent populations are presented.
Collapse
|
18
|
Spinal cord injury in mice impacts central and peripheral pathology in a severity-dependent manner. Pain 2021; 163:1172-1185. [PMID: 34490852 DOI: 10.1097/j.pain.0000000000002471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Chronic pain is a common medical complication experienced by those living with spinal cord injury (SCI) and leads to worsened quality of life. The pathophysiology of SCI pain is poorly understood, hampering the development of safe and efficacious therapeutics. We therefore sought to develop a clinically relevant model of SCI with a strong pain phenotype and characterize the central and peripheral pathology after injury. A contusion (50 kdyn) injury, with and without sustained compression (60 seconds) of the spinal cord, was carried out on female C57BL/6J mice. Mice with compression of the spinal cord exhibited significantly greater heat and mechanical hypersensitivity starting at 7 days post-injury, concomitant with reduced locomotor function, compared to those without compression. Immunohistochemical analysis of spinal cord tissue revealed significantly less myelin sparing and increased macrophage activation in mice with compression compared to those without. As measured by flow cytometry, immune cell infiltration and activation were significantly greater in the spinal cord (phagocytic myeloid cells and microglia) and dorsal root ganglia (Ly6C+ monocytes) following compression injury. We also decided to investigate the gastrointestinal microbiome, as it has been shown to be altered in SCI patients and has recently been shown to play a role in immune system maturation and pain. We found increased dysbiosis of the gastrointestinal microbiome in an injury severity-dependent manner. The use of this contusion-compression model of SCI may help advance the preclinical assessment of acute and chronic SCI pain and lead to a better understanding of mechanisms contributing to this pain.
Collapse
|
19
|
Treadmill training based on the overload principle promotes locomotor recovery in a mouse model of chronic spinal cord injury. Exp Neurol 2021; 345:113834. [PMID: 34370998 DOI: 10.1016/j.expneurol.2021.113834] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/16/2021] [Accepted: 08/02/2021] [Indexed: 11/21/2022]
Abstract
Rehabilitative treatment, including treadmill training, is considered an important strategy for restoring motor function after spinal cord injury (SCI). However, many unexplained problems persist regarding the appropriate rehabilitative method and the mechanism underlying the beneficial effects of rehabilitation. Moreover, only a few preclinical studies have been performed on rehabilitative interventions for chronic SCI, although most patients have chronic injuries. In fact, several preclinical studies reported that rehabilitative training was less effective when applied during the chronic phase than when applied sooner. While numerous studies have examined the effects of treadmill training during the subacute phase, the training conditions vary considerably among preclinical reports. Therefore, establishing a standard training protocol is essential for achieving beneficial rehabilitation effects at the chronic stage. Since the difficulty of applying an appropriate training load hinders training at constant speeds, it is important to adjust the training intensity in accordance with the exercise tolerance of an individual animal to provide further functional recovery benefits. Here, we created a novel quadrupedal treadmill training protocol based on the overload principle for mice with incomplete thoracic SCI. We subjected SCI model mice to rehabilitative training according to the protocol for two consecutive weeks starting at 42 days after injury. We examined the treadmill speeds at which the mice were able to run based on the severity of paresis and investigated the impact of the protocol on functional recovery. Assessment of running speed changes during the treadmill training period revealed faster treadmill speeds for mice with mild paresis than for those with severe paresis. The training parameters, including the speed and distance traveled, were positively correlated with the changes in motor function. These results suggest that the most suitable running speed during treadmill training differs according to the level of motor dysfunction and that running longer distances has a positive impact on motor functional recovery. Based on this established protocol, we compared functional and histological results between the chronic SCI groups with and without rehabilitation. The gait analyses showed significantly better functional improvement in the rehabilitation group than in the nonrehabilitation group. Histological analyses revealed that the BDNF- and VGLUT1-positive areas of lumbar enlargement were significantly increased in the rehabilitation group. These findings implied that rehabilitation promoted not only motor performance but also motor control, including forelimb-hindlimb coordination, even in chronic SCI, resulting in functional improvement by treadmill training alone. Therefore, rehabilitative training based on the overload principle appears to be one of the appropriate treatment options for incomplete thoracic SCI, and evidence of its efficacy exists in actual clinical settings.
Collapse
|
20
|
Harman KA, DeVeau KM, Squair JW, West CR, Krassioukov AV, Magnuson DSK. Effects of early exercise training on the severity of autonomic dysreflexia following incomplete spinal cord injury in rodents. Physiol Rep 2021; 9:e14969. [PMID: 34337884 PMCID: PMC8327165 DOI: 10.14814/phy2.14969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/04/2022] Open
Abstract
Hemodynamic instability and cardiovascular (CV) dysfunction are hallmarks of patients living with cervical and high thoracic spinal cord injuries (SCI). Individuals experience bouts of autonomic dysreflexia (AD) and persistent hypotension which hamper the activities of daily living. Despite the widespread use of exercise training to improve health and CV function after SCI, little is known about how different training modalities impact hemodynamic stability and severity of AD in a model of incomplete SCI. In this study, we used implantable telemetry devices to assess animals with T2 contusions following 3.5 weeks of exercise training initiated 8 days post-injury: passive hindlimb cycling (T2-CYC, n = 5) or active forelimb swimming (T2-SW, n = 6). Uninjured and non-exercised SCI control groups were also included (CON, n = 6; T2-CON, n = 7; T10-CON, n = 6). Five weeks post-injury, both T2-CON and T2-CYC presented with resting hypotension compared to uninjured CON and T10-CON groups; no differences were noted in resting blood pressure in T2-SW versus CON and T10-CON. Furthermore, pressor responses to colorectal distention (AD) were larger in all T2-injured groups compared to T10-CON, and were not attenuated by either form of exercise training. Interestingly, when T2-injured animals were re-stratified based on terminal BBB scores (regardless of training group), animals with limited hindlimb recovery (T2-LOW, n = 7) had more severe AD responses. Our results suggest that the spontaneous recovery of locomotor and autonomic function after severe but incomplete T2 SCI also influences the severity of AD, and that short periods (3.5 weeks) of passive hindlimb cycling or active forelimb swimming are ineffective in this model.
Collapse
Affiliation(s)
- Kathryn A. Harman
- Department of Health & Sport SciencesUniversity of LouisvilleLouisvilleKYUSA
- Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Kathryn M. DeVeau
- Department of Anatomy and Cell BiologyGeorge Washington UniversityWashingtonD.C.USA
| | - Jordan W. Squair
- International Collaboration on Repair DiscoveriesUniversity of British ColumbiaVancouverBCCanada
| | - Christopher R. West
- International Collaboration on Repair DiscoveriesUniversity of British ColumbiaVancouverBCCanada
| | - Andrei V. Krassioukov
- International Collaboration on Repair DiscoveriesUniversity of British ColumbiaVancouverBCCanada
- GF Strong Rehabilitation CentreVancouver Health AuthorityVancouverCanada
| | - David S. K. Magnuson
- Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKYUSA
- Department of Neurological SurgeryUniversity of LouisvilleLouisvilleKYUSA
| |
Collapse
|
21
|
Li X, Wang Q, Ding J, Wang S, Dong C, Wu Q. Exercise training modulates glutamic acid decarboxylase-65/67 expression through TrkB signaling to ameliorate neuropathic pain in rats with spinal cord injury. Mol Pain 2021; 16:1744806920924511. [PMID: 32418502 PMCID: PMC7235678 DOI: 10.1177/1744806920924511] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuropathic pain is one of the most frequently stated complications after spinal cord injury. In post-spinal cord injury, the decrease of gamma aminobutyric acid synthesis within the distal spinal cord is one of the main causes of neuropathic pain. The predominant research question of this study was whether exercise training may promote the expression of glutamic acid decarboxylase-65 and glutamic acid decarboxylase-67, which are key enzymes of gamma aminobutyric acid synthesis, within the distal spinal cord through tropomyosin-related kinase B signaling, as its synthesis assists to relieve neuropathic pain after spinal cord injury. Animal experiment was conducted, and all rats were allocated into five groups: Sham group, SCI/PBS group, SCI-TT/PBS group, SCI/tropomyosin-related kinase B-IgG group, and SCI-TT/tropomyosin-related kinase B-IgG group, and then T10 contusion SCI model was performed as well as the tropomyosin-related kinase B-IgG was used to block the tropomyosin-related kinase B activation. Mechanical withdrawal thresholds and thermal withdrawal latencies were used for assessing pain-related behaviors. Western blot analysis was used to detect the expression of brain-derived neurotrophic factor, tropomyosin-related kinase B, CREB, p-REB, glutamic acid decarboxylase-65, and glutamic acid decarboxylase-67 within the distal spinal cord. Immunohistochemistry was used to analyze the distribution of CREB, p-CREB, glutamic acid decarboxylase-65, and glutamic acid decarboxylase-67 within the distal spinal cord dorsal horn. The results showed that exercise training could significantly mitigate the mechanical allodynia and thermal hyperalgesia in post-spinal cord injury and increase the synthesis of brain-derived neurotrophic factor, tropomyosin-related kinase B, CREB, p-CREB, glutamic acid decarboxylase-65, and glutamic acid decarboxylase-67 within the distal spinal cord. After the tropomyosin-related kinase B signaling was blocked, the analgesic effect of exercise training was inhibited, and in the SCI-TT/tropomyosin-related kinase B-IgG group, the synthesis of CREB, p-CREB, glutamic acid decarboxylase-65, and glutamic acid decarboxylase-67 within the distal spinal cord were also significantly reduced compared with the SCI-TT/PBS group. This study shows that exercise training may increase the glutamic acid decarboxylase-65 and glutamic acid decarboxylase-67 expression within the spinal cord dorsal horn through the tropomyosin-related kinase B signaling, and this mechanism may play a vital role in relieving the neuropathic pain of rats caused by incomplete SCI.
Collapse
Affiliation(s)
- Xiangzhe Li
- Rehabilitation Medical Center, the Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China
| | - Qinghua Wang
- Laboratory Animal Center, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jie Ding
- Departments of Respiratory Care, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Sheng Wang
- Rehabilitation Medical Center, the Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Qinfeng Wu
- Rehabilitation Medical Center, the Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, People's Republic of China
| |
Collapse
|
22
|
Eller OC, Yang X, Fuentes IM, Pierce AN, Jones BM, Brake AD, Wang R, Dussor G, Christianson JA. Voluntary Wheel Running Partially Attenuates Early Life Stress-Induced Neuroimmune Measures in the Dura and Evoked Migraine-Like Behaviors in Female Mice. Front Physiol 2021; 12:665732. [PMID: 34122137 PMCID: PMC8194283 DOI: 10.3389/fphys.2021.665732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
Migraine is a complex neurological disorder that affects three times more women than men and can be triggered by endogenous and exogenous factors. Stress is a common migraine trigger and exposure to early life stress increases the likelihood of developing chronic pain disorders later in life. Here, we used our neonatal maternal separation (NMS) model of early life stress to investigate whether female NMS mice have an increased susceptibility to evoked migraine-like behaviors and the potential therapeutic effect of voluntary wheel running. NMS was performed for 3 h/day during the first 3 weeks of life and initial observations were made at 12 weeks of age after voluntary wheel running (Exercise, -Ex) or sedentary behavior (-Sed) for 4 weeks. Mast cell degranulation rates were significantly higher in dura mater from NMS-Sed mice, compared to either naïve-Sed or NMS-Ex mice. Protease activated receptor 2 (PAR2) protein levels in the dura were significantly increased in NMS mice and a significant interaction of NMS and exercise was observed for transient receptor potential ankyrin 1 (TRPA1) protein levels in the dura. Behavioral assessments were performed on adult (>8 weeks of age) naïve and NMS mice that received free access to a running wheel beginning at 4 weeks of age. Facial grimace, paw mechanical withdrawal threshold, and light aversion were measured following direct application of inflammatory soup (IS) onto the dura or intraperitoneal (IP) nitroglycerin (NTG) injection. Dural IS resulted in a significant decrease in forepaw withdrawal threshold in all groups of mice, while exercise significantly increased grimace score across all groups. NTG significantly increased grimace score, particularly in exercised mice. A significant effect of NMS and a significant interaction effect of exercise and NMS were observed on hindpaw sensitivity following NTG injection. Significant light aversion was observed in NMS mice, regardless of exercise, following NTG. Finally, exercise significantly reduced calcitonin gene-related peptide (CGRP) protein level in the dura of NMS and naïve mice. Taken together, these findings suggest that while voluntary wheel running improved some measures in NMS mice that have been associated with increased migraine susceptibility, behavioral outcomes were not impacted or even worsened by exercise.
Collapse
Affiliation(s)
- Olivia C. Eller
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Xiaofang Yang
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Isabella M. Fuentes
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Angela N. Pierce
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Physiology, Kansas City University of Medicine and Biosciences, Joplin, MO, United States
| | - Brittni M. Jones
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Aaron D. Brake
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ruipeng Wang
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, United States
| | - Julie A. Christianson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
23
|
Gan Z, Li H, Naser PV, Oswald MJ, Kuner R. Suppression of neuropathic pain and comorbidities by recurrent cycles of repetitive transcranial direct current motor cortex stimulation in mice. Sci Rep 2021; 11:9735. [PMID: 33958647 PMCID: PMC8102487 DOI: 10.1038/s41598-021-89122-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/12/2021] [Indexed: 11/16/2022] Open
Abstract
Transcranial, minimally-invasive stimulation of the primary motor cortex (M1) has recently emerged to show promise in treating clinically refractory neuropathic pain. However, there is a major need for improving efficacy, reducing variability and understanding mechanisms. Rodent models hold promise in helping to overcome these obstacles. However, there still remains a major divide between clinical and preclinical studies with respect to stimulation programs, analysis of pain as a multidimensional sensory-affective-motivational state and lack of focus on chronic phases of established pain. Here, we employed direct transcranial M1 stimulation (M1 tDCS) either as a single 5-day block or recurring blocks of repetitive stimulation over early or chronic phases of peripherally-induced neuropathic pain in mice. We report that repeated blocks of stimulation reverse established neuropathic mechanical allodynia more strongly than a single 5-day regime and also suppress cold allodynia, aversive behavior and anxiety without adversely affecting motor function over a long period. Activity mapping revealed highly selective alterations in the posterior insula, periaqueductal gray subdivisions and superficial spinal laminae in reversal of mechanical allodynia. Our preclinical data reveal multimodal analgesia and improvement in quality of life by multiple blocks of M1 tDCS and uncover underlying brain networks, thus helping promote clinical translation.
Collapse
Affiliation(s)
- Zheng Gan
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Han Li
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Paul Vincent Naser
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Manfred Josef Oswald
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Rohini Kuner
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
24
|
Leitzelar BN, Koltyn KF. Exercise and Neuropathic Pain: A General Overview of Preclinical and Clinical Research. SPORTS MEDICINE-OPEN 2021; 7:21. [PMID: 33751253 PMCID: PMC7984211 DOI: 10.1186/s40798-021-00307-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
Neuropathic pain is a disease of the somatosensory system that is characterized by tingling, burning, and/or shooting pain. Medication is often the primary treatment, but it can be costly, thus there is an interest in understanding alternative low-cost treatments such as exercise. The following review includes an overview of the preclinical and clinical literature examining the influence of exercise on neuropathic pain. Preclinical studies support the hypothesis that exercise reduces hyperalgesia and allodynia in animal models of neuropathic pain. In human research, observational studies suggest that those who are more physically active have lower risk of developing neuropathic pain compared to those who are less active. Exercise studies suggest aerobic exercise training (e.g., 16 weeks); a combination of aerobic and resistance exercise training (e.g., 10–12 weeks); or high-intensity interval training (e.g., 15 weeks) reduces aspects of neuropathic pain such as worst pain over the past month, pain over the past 24 h, pain scores, or pain interference. However, not all measures of pain improve following exercise training (e.g., current pain, heat pain threshold). Potential mechanisms and future directions are also discussed to aid in the goal of understanding the role of exercise in the management of neuropathic pain. Future research using standardized methods to further understanding of the dose of exercise needed to manage neuropathic pain is warranted.
Collapse
Affiliation(s)
- Brianna N Leitzelar
- Department of Kinesiology, University of Wisconsin-Madison, 1300 University Ave., Madison, WI, 53706, USA
| | - Kelli F Koltyn
- Department of Kinesiology, University of Wisconsin-Madison, 1300 University Ave., Madison, WI, 53706, USA.
| |
Collapse
|
25
|
Dugan EA, Schachner B, Jergova S, Sagen J. Intensive Locomotor Training Provides Sustained Alleviation of Chronic Spinal Cord Injury-Associated Neuropathic Pain: A Two-Year Pre-Clinical Study. J Neurotrauma 2021; 38:789-802. [PMID: 33218293 DOI: 10.1089/neu.2020.7378] [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: 12/20/2022] Open
Abstract
Neuropathic pain often accompanies the functional deficits associated with spinal cord injury (SCI) and further reduces a patient's quality of life. Clinical and pre-clinical research is beginning to highlight the beneficial role that rehabilitative therapies such as locomotor training can have not only on functional recovery but also on chronic pain management. Our group has previously developed an intensive locomotor training (ILT) treadmill protocol on rats that reduced SCI neuropathic pain symptoms for at least 3 months. We have extended these findings in the current study to evaluate the ability of regular ILT regimen over a 2 year period post-SCI to maintain neuropathic pain reduction. To assess this, the rat clip compression SCI model (T7/8) was used and treadmill training was initiated starting 4 weeks after SCI and continuing through the duration of the study. Results showed continued suppression of SCI neuropathic pain responses (reduced mechanical, heat, and cold hypersensitivity throughout the entire time course of the study). In contrast, non-exercised rats showed consistent and sustained neuropathic pain responses during this period. In addition, prolonged survival and improved locomotor outcomes were observed in rats undergoing ILT as the study longevity progressed. Potential contributory mechanisms underlying beneficial effects of ILT include reduced inflammation and restoration of anti-nociceptive inhibitory processes as indicated by neurochemical assays in spinal tissue of remaining rats at 2 years post-SCI. The benefits of chronic ILT suggest that long-term physical exercise therapy can produce powerful and prolonged management of neuropathic pain, partly through sustained reduction of spinal pathological processes.
Collapse
Affiliation(s)
- Elizabeth A Dugan
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Benjamin Schachner
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Stanislava Jergova
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Jacqueline Sagen
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| |
Collapse
|
26
|
Bannerman CA, Douchant K, Sheth PM, Ghasemlou N. The gut-brain axis and beyond: Microbiome control of spinal cord injury pain in humans and rodents. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 9:100059. [PMID: 33426367 PMCID: PMC7779861 DOI: 10.1016/j.ynpai.2020.100059] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/26/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a devastating injury to the central nervous system in which 60 to 80% of patients experience chronic pain. Unfortunately, this pain is notoriously difficult to treat, with few effective options currently available. Patients are also commonly faced with various compounding injuries and medical challenges, often requiring frequent hospitalization and antibiotic treatment. Change in the gut microbiome from the "normal" state to one of imbalance, referred to as gut dysbiosis, has been found in both patients and rodent models following SCI. Similarities exist in the bacterial changes observed after SCI and other diseases with chronic pain as an outcome. These changes cause a shift in the regulation of inflammation, causing immune cell activation and secretion of inflammatory mediators that likely contribute to the generation/maintenance of SCI pain. Therefore, correcting gut dysbiosis may be used as a tool towards providing patients with effective pain management and improved quality of life.
Collapse
Affiliation(s)
- Courtney A. Bannerman
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Katya Douchant
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- Gastrointestinal Disease Research Unit, Kingston Health Sciences Center, Kingston, Ontario, Canada
| | - Prameet M. Sheth
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada
- Division of Microbiology, Kingston Health Sciences Centre, Kingston, Ontario, Canada
- Gastrointestinal Disease Research Unit, Kingston Health Sciences Center, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- Department of Anesthesiology and Perioperative Medicine, Kingston Health Sciences Centre, Kingston, Ontario, Canada
- Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario, Canada
| |
Collapse
|
27
|
Huang Y, Lu Y, Zhao X, Zhang J, Zhang F, Chen Y, Bi L, Gu J, Jiang Z, Wu X, Li Q, Liu Y, Shen J, Liu X. Cytokine activin C ameliorates chronic neuropathic pain in peripheral nerve injury rodents by modulating the TRPV1 channel. Br J Pharmacol 2020; 177:5642-5657. [PMID: 33095918 PMCID: PMC7707095 DOI: 10.1111/bph.15284] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/06/2020] [Accepted: 09/25/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE The cytokine activin C is mainly expressed in small-diameter dorsal root ganglion (DRG) neurons and suppresses inflammatory pain. However, the effects of activin C in neuropathic pain remain elusive. EXPERIMENTAL APPROACH Male rats and wild-type and TRPV1 knockout mice with peripheral nerve injury - sciatic nerve axotomy and spinal nerve ligation in rats; chronic constriction injury (CCI) in mice - provided models of chronic neuropathic pain. Ipsilateral lumbar (L)4-5 DRGs were assayed for activin C expression. Chronic neuropathic pain animals were treated with intrathecal or locally pre-administered activin C or the vehicle. Nociceptive behaviours and pain-related markers in L4-5 DRGs and spinal cord were evaluated. TRPV1 channel modulation by activin C was measured. KEY RESULTS Following peripheral nerve injury, expression of activin βC subunit mRNA and activin C protein was markedly up-regulated in L4-5 DRGs of animals with axotomy, SNL or CCI. [Correction added on 26 November 2020, after first online publication: The preceding sentence has been corrected in this current version.] Intrathecal activin C dose-dependently inhibited neuropathic pain in spinal nerve ligated rats. Local pre-administration of activin C decreased neuropathic pain, macrophage infiltration into ipsilateral L4-5 DRGs and microglial reaction in L4-5 spinal cords of mice with CCI. In rat DRG neurons, activin C enhanced capsaicin-induced TRPV1 currents. Pre-treatment with activin C reduced capsaicin-evoked acute hyperalgesia and normalized capsaicin-evoked persistent hypothermia in mice. Finally, the analgesic effect of activin C was abolished in TRPV1 knockout mice with CCI. CONCLUSION AND IMPLICATIONS Activin C inhibits neuropathic pain by modulating TRPV1 channels, revealing potential analgesic applications in chronic neuropathic pain therapy.
Collapse
Affiliation(s)
- Ya‐Kun Huang
- School of PharmacyNantong UniversityNantongChina
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Yu‐Gang Lu
- Department of Anesthesiology, Shanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Xin Zhao
- Department of GeriatricsRenji Hospital, School of Medicine, Shanghai Jiaotong UniversityShanghaiChina
| | - Jing‐Bing Zhang
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | | | - Yong Chen
- School of PharmacyNantong UniversityNantongChina
| | - Ling‐Bo Bi
- School of PharmacyNantong UniversityNantongChina
| | - Jia‐Hui Gu
- School of PharmacyNantong UniversityNantongChina
| | - Zuo‐Jie Jiang
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Xiao‐Man Wu
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Qing‐Yi Li
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Yanli Liu
- College of Pharmaceutical ScienceSoochow UniversitySuzhouChina
| | - Jian‐Xin Shen
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| | - Xing‐Jun Liu
- School of PharmacyNantong UniversityNantongChina
- Pain and Related Diseases Research LaboratoryShantou University Medical CollegeShantouChina
| |
Collapse
|
28
|
Cheng X, Xiao F, Xie R, Hu H, Wan Y. Alternate thermal stimulation ameliorates thermal sensitivity and modulates calbindin-D 28K expression in lamina I and II and dorsal root ganglia in a mouse spinal cord contusion injury model. FASEB J 2020; 35:e21173. [PMID: 33225523 DOI: 10.1096/fj.202001775r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/30/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
Neuropathic pain (NP) is a common complication that negatively affects the lives of patients with spinal cord injury (SCI). The disruption in the balance of excitatory and inhibitory neurons in the spinal cord dorsal horn contributes to the development of SCI and induces NP. The calcium-binding protein (CaBP) calbindin-D 28K (CaBP-28K) is highly expressed in excitatory interneurons, and the CaBP parvalbumin (PV) is present in inhibitory neurons in the dorsal horn. To better define the changes in the CaBPs contributing to the development of SCI-induced NP, we examined the changes in CaBP-28K and PV staining density in the lumbar (L4-6) lamina I and II, and their relationship with NP after mild spinal cord contusion injury in mice. We additionally examined the effects of alternate thermal stimulation (ATS). Compared with sham mice, injured animals developed mechanical allodynia in response to light mechanical stimuli and exhibited mechanical hyporesponsiveness to noxious mechanical stimuli. The decreased response latency to heat stimuli and increased response latency to cold stimuli at 7 days post injury suggested that the injured mice developed heat hyperalgesia and cold hypoalgesia, respectively. Temperature preference tests showed significant warm allodynia after injury. Animals that underwent ATS (15-18 and 35-40°C; +5 minutes/stimulation/day; 5 days/week) displayed significant amelioration of heat hyperalgesia, cold hypoalgesia, and warm allodynia after 2 weeks of ATS. In contrast, mechanical sensitivity was not influenced by ATS. Analysis of the CaBP-28K positive signal in L4-6 lamina I and II indicated an increase in staining density after SCI, which was associated with an increase in the number of CaBP-28K-stained L4-6 dorsal root ganglion (DRG) neurons. ATS decreased the CaBP-28K staining density in L4-6 spinal cord and DRG in injured animals, and was significantly and strongly correlated with ATS alleviation of pain behavior. The expression of PV showed no changes in lamina I and II after ATS in SCI animals. Thus, ATS partially decreases the pain behavior after SCI by modulating the changes in CaBP-associated excitatory-inhibitory neurons.
Collapse
Affiliation(s)
- Xing Cheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Spinal Cord Injury Center, Heidelberg University, Heidelberg, Germany
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Rong Xie
- Department of Thyroid Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Haijun Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Yong Wan
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, People's Republic of China
| |
Collapse
|
29
|
Kang J, Cho SS, Kim HY, Lee BH, Cho HJ, Gwak YS. Regional Hyperexcitability and Chronic Neuropathic Pain Following Spinal Cord Injury. Cell Mol Neurobiol 2020; 40:861-878. [PMID: 31955281 PMCID: PMC11448802 DOI: 10.1007/s10571-020-00785-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 01/02/2020] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) causes maladaptive changes to nociceptive synaptic circuits within the injured spinal cord. Changes also occur at remote regions including the brain stem, limbic system, cortex, and dorsal root ganglia. These maladaptive nociceptive synaptic circuits frequently cause neuronal hyperexcitability in the entire nervous system and enhance nociceptive transmission, resulting in chronic central neuropathic pain following SCI. The underlying mechanism of chronic neuropathic pain depends on the neuroanatomical structures and electrochemical communication between pre- and postsynaptic neuronal membranes, and propagation of synaptic transmission in the ascending pain pathways. In the nervous system, neurons are the only cell type that transmits nociceptive signals from peripheral receptors to supraspinal systems due to their neuroanatomical and electrophysiological properties. However, the entire range of nociceptive signaling is not mediated by any single neuron. Current literature describes regional studies of electrophysiological or neurochemical mechanisms for enhanced nociceptive transmission post-SCI, but few studies report the electrophysiological, neurochemical, and neuroanatomical changes across the entire nervous system following a regional SCI. We, along with others, have continuously described the enhanced nociceptive transmission in the spinal dorsal horn, brain stem, thalamus, and cortex in SCI-induced chronic central neuropathic pain condition, respectively. Thus, this review summarizes the current understanding of SCI-induced neuronal hyperexcitability and maladaptive nociceptive transmission in the entire nervous system that contributes to chronic central neuropathic pain.
Collapse
Affiliation(s)
- Jonghoon Kang
- Department of Biology, Valdosta State University, Valdosta, GA, 31698, USA
| | - Steve S Cho
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hee Young Kim
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, 42158, South Korea
| | - Bong Hyo Lee
- Department of Acupuncture, Moxibustion and Acupoint, College of Korean Medicine, Daegu Haany University, Daegu, 42158, South Korea
| | - Hee Jung Cho
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, 41944, South Korea.
| | - Young S Gwak
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, 42158, South Korea.
| |
Collapse
|
30
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
31
|
Liu Q, Liu Y, Bian J, Li Q, Zhang Y. The preemptive analgesia of pre-electroacupuncture in rats with formalin-induced acute inflammatory pain. Mol Pain 2020; 15:1744806919866529. [PMID: 31322476 PMCID: PMC6685110 DOI: 10.1177/1744806919866529] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Electroacupuncture has been elicited to effectively alleviate the pain sensation. Preemptive analgesic effect of pre-electroacupuncture has also been suggested in recent studies, while the underlying analgesic mechanism of pre-electroacupuncture requires further investigation. This study aimed to explore the preemptive analgesia of pre-electroacupuncture in formalin-induced acute inflammatory pain model. Methods Forty rats were randomly divided into control, model, pre-electroacupuncture, and post-electroacupuncture group. Inflammatory pain model was induced via injecting 50 µl 5% formalin into the plantar surface of right hind paw, while the equal volume of saline injection in the control group. Rats in the pre-electroacupuncture group were treated with electroacupuncture at ipsilateral Zusanli (ST36) and Weizhong (BL40) acupoints (2 Hz, 1 mA) for 30 min before formalin injection, while received the same electroacupuncture treatment immediately after formalin injection in the post-electroacupuncture group. Flinching number and licking time were recorded during 60 min after formalin injection. Immunofluorescence and Western blot were used to detect the expression of ionized calcium binding adapter molecule 1 (Iba1) and c-fos in spinal cord. Moreover, enzyme-linked immunosorbent assay was applied to measure the secretion of IL-6, IFN-γ, IL-4, substance P, and calcitonin gene-related peptide in spinal cord. Results Paw flinching and licking were obviously induced by formalin injection. Iba1, c-fos, proinflammatory cytokines (IL-6 and IFN-γ), and pain neurotransmitters (substance P and calcitonin gene-related peptide) were dramatically increased in the L4-5 spinal cord after formalin injection, while anti-inflammatory cytokine IL-4 was decreased. Pre-electroacupuncture and post-electroacupuncture administration significantly attenuated formalin-induced nociceptive effects, spinal microglia and neurons activation, proinflammatory cytokines and pain neurotransmitters upregulation, and upregulated the anti-inflammatory cytokine. Furthermore, these effects of pre-electroacupuncture were more significant than that of post-electroacupuncture. Conclusions This study illustrates the potential therapeutic effect of pre-electroacupuncture against acute inflammatory pain and reveals the mechanism underlying pre-electroacupuncture mediated analgesia, thus providing a novel preemptive analgesic treatment.
Collapse
Affiliation(s)
- Qing Liu
- 1 Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yan Liu
- 1 Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jiang Bian
- 1 Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qun Li
- 1 Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ying Zhang
- 1 Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
32
|
Effects of Robot-Assisted Gait Training in Individuals with Spinal Cord Injury: A Meta-analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2102785. [PMID: 32280681 PMCID: PMC7115057 DOI: 10.1155/2020/2102785] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/21/2020] [Indexed: 12/20/2022]
Abstract
Background To investigate the effects of robot-assisted gait training (RAGT) on spasticity and pain in people with spinal cord injury (SCI). Material and methods. Four electronic databases (PubMed, Scopus, Medline, and Cochrane Central Register of Controlled Trials) were searched for studies published up to November 2019. Only human trials and of English language were included. The searched studies were reviewed and extracted independently by two authors. Randomized controlled trials (RCTs) and non-RCTs were pooled separately for analyses. Primary outcome measures included spasticity assessed by Ashworth scale (AS) or modified Ashworth scale (MAS) and pain assessed by VAS. Secondary outcome measures included lower extremity motor score (LEMS) and walking ability (i.e., 6-minute walk test, 10-meter walk test). Results A total of 225 studies were identified. Eighteen studies (7 RCTs and 11 non-RCTs) including 301 subjects met inclusion criteria. The outcome measure of spasticity significantly improved in favor of RAGT group in non-RCTs (AS: 95%CI = −0.202 to -0.068, p ≤ 0.001; MAS: 95%CI = −2.886 to -1.412, p ≤ 0.001). The results on pain did not show significant change after RAGT in either RCTs or non-RCTs. LEMS and walking ability significantly increased in favor of RAGT. Conclusions RAGT can improve spasticity and walking ability in people with SCI. The probable reason for no significant change in pain after RAGT is floor effect. RAGT is beneficial for normalizing muscle tone and for improving lower extremity function in people with SCI without causing extra pain.
Collapse
|
33
|
Mutually beneficial effects of intensive exercise and GABAergic neural progenitor cell transplants in reducing neuropathic pain and spinal pathology in rats with spinal cord injury. Exp Neurol 2020; 327:113208. [PMID: 31962127 DOI: 10.1016/j.expneurol.2020.113208] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 01/13/2023]
Abstract
Spinal cord injury (SCI) produces both locomotor deficits and sensory dysfunction that greatly reduce the overall quality of life. Mechanisms underlying chronic pain include increased neuro-inflammation and changes in spinal processing of sensory signals, with reduced inhibitory GABAergic signaling a likely key player. Our previous research demonstrated that spinal transplantation of GABAergic neural progenitor cells (NPCs) reduced neuropathic pain while intensive locomotor training (ILT) could reduce development of pain and partially reverse already established pain behaviors. Therefore, we evaluate the potential mutually beneficial anti-hypersensitivity effects of NPC transplants cells in combination with early or delayed ILT. NPC transplants were done at 4 weeks post-SCI. ILT, using a progressive ramping treadmill protocol, was initiated either 5 days post-SCI (early: pain prevention group) or at 5 weeks post-SCI (delayed: to reverse established pain) in male Sprague Dawley rats. Results showed that either ILT alone or NPCs alone could partially attenuate SCI neuropathic pain behaviors in both prevention and reversal paradigms. However, the combination of ILT with NPC transplants significantly enhanced neuropathic pain reduction on most of the outcome measures including tests for allodynia, hyperalgesia, and ongoing pain. Immunocytochemical and neurochemical analyses showed decreased pro-inflammatory markers and spinal pathology with individual treatments; these measures were further improved by the combination of either early or delayed ILT and GABAergic cellular transplantation. Lumbar dorsal horn GABAergic neuronal and process density were nearly restored to normal levels by the combination treatment. Together, these interventions may provide a less hostile and more supportive environment for promoting functional restoration in the spinal dorsal horn and attenuation of neuropathic pain following SCI. These findings suggest mutually beneficial effects of ILT and NPC transplants for reducing SCI neuropathic pain.
Collapse
|
34
|
Palandi J, Bobinski F, de Oliveira GM, Ilha J. Neuropathic pain after spinal cord injury and physical exercise in animal models: A systematic review and meta-analysis. Neurosci Biobehav Rev 2019; 108:781-795. [PMID: 31837360 DOI: 10.1016/j.neubiorev.2019.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022]
Abstract
The aim of this systematic review was to summarize the effects of physical exercise on neuropathic pain (NP) in animal models of SCI. The search was conducted in Medline and Science Direct to identify experimental preclinical studies involving animal models of SCI, physical exercise as an intervention and the assessment of NP. Fifteen articles met the eligibility criteria. The review shows that in studies of NP involving animal models of SCI, rodents are the most common species. Thoracic contusion is the most common injury and mechanical and thermal nociception are the most frequently assessed NP components. The benefits of physical exercise vary according to its starting period and total duration. In addition, there is considerable heterogeneity regarding the type and intensity of exercise capable of alleviating NP after SCI. Furthermore, physical exercise has beneficial effects on mechanical, thermal and cold nociception, and spontaneous pain. These results are weakened by the paucity of studies involving these pain outcomes. The review protocol is published for free access on the SyRF platform (http://syrf.org.uk/protocols/).
Collapse
Affiliation(s)
- Juliete Palandi
- Physical Therapy Graduate Program, Department of Physical Therapy, College of Health and Sport Science, Santa Catarina State University (UDESC), Florianópolis, 88080-350, SC, Brazil; Spinal Cord Injury Research Group, Neuromotor System Laboratory, Department of Physical Therapy, College of Health and Sport Science, Santa Catarina State University (UDESC), Florianópolis, 88080-350, SC, Brazil
| | - Franciane Bobinski
- Physical Therapy Graduate Program, Department of Physical Therapy, College of Health and Sport Science, Santa Catarina State University (UDESC), Florianópolis, 88080-350, SC, Brazil; Experimental Neuroscience Laboratory, Graduate Program in Health Sciences, University of Southern of Santa Catarina (UNISUL), Palhoça, 88137-272, SC, Brazil
| | - Gabriela Martins de Oliveira
- Spinal Cord Injury Research Group, Neuromotor System Laboratory, Department of Physical Therapy, College of Health and Sport Science, Santa Catarina State University (UDESC), Florianópolis, 88080-350, SC, Brazil
| | - Jocemar Ilha
- Physical Therapy Graduate Program, Department of Physical Therapy, College of Health and Sport Science, Santa Catarina State University (UDESC), Florianópolis, 88080-350, SC, Brazil; Spinal Cord Injury Research Group, Neuromotor System Laboratory, Department of Physical Therapy, College of Health and Sport Science, Santa Catarina State University (UDESC), Florianópolis, 88080-350, SC, Brazil.
| |
Collapse
|
35
|
Yasko JR, Moss IL, Mains RE. Transcriptional Profiling of Non-injured Nociceptors After Spinal Cord Injury Reveals Diverse Molecular Changes. Front Mol Neurosci 2019; 12:284. [PMID: 32038157 PMCID: PMC6988781 DOI: 10.3389/fnmol.2019.00284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/08/2019] [Indexed: 01/01/2023] Open
Abstract
Traumatic spinal cord injury (SCI) has devastating implications for patients, including a high predisposition for developing chronic pain distal to the site of injury. Chronic pain develops weeks to months after injury, consequently, patients are treated after irreparable changes have occurred. Nociceptors are central to chronic pain; however, the diversity of this cellular population presents challenges to understanding mechanisms and attributing pain modalities to specific cell types. To begin to address how peripheral sensory neurons below the injury level may contribute to the below-level pain reported by SCI patients, we examined SCI-induced changes in gene expression in lumbar dorsal root ganglia (DRG) below the site of injury. SCI was performed at the T10 vertebral level, with injury produced by a vessel clip with a closing pressure of 15 g for 1 min. Alterations in gene expression produce long-term sensory changes, therefore, we were interested in studying SCI-induced transcripts before the onset of chronic pain, which may trigger changes in downstream signaling pathways and ultimately facilitate the transmission of pain. To examine changes in the nociceptor subpopulation in DRG distal to the site of injury, we retrograde labeled sensory neurons projecting to the hairy hindpaw skin with fluorescent dye and collected the corresponding lumbar (L2–L6) DRG 4 days post-injury. Following dissociation, labeled neurons were purified by fluorescence-activated cell sorting (FACS). RNA was extracted from sorted sensory neurons of naïve, sham, or SCI mice and sequenced. Transcript abundances validated that the desired population of nociceptors were isolated. Cross-comparisons to data sets from similar studies confirmed, we were able to isolate our cells of interest and identify a unique pattern of gene expression within a subpopulation of neurons projecting to the hairy hindpaw skin. Differential gene expression analysis showed high expression levels and significant transcript changes 4 days post-injury in SCI cell populations relevant to the onset of chronic pain. Regulatory interrelationships predicted by pathway analysis implicated changes within the synaptogenesis signaling pathway as well as networks related to inflammatory signaling mechanisms, suggesting a role for synaptic plasticity and a correlation with pro-inflammatory signaling in the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Jessica R Yasko
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| | - Isaac L Moss
- Department of Orthopedic Surgery and the Comprehensive Spine Center, University of Connecticut Health Center, Farmington, CT, United States
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
36
|
Chariker JH, Gomes C, Brabazon F, Harman KA, Ohri SS, Magnuson DSK, Whittemore SR, Petruska JC, Rouchka EC. Transcriptome of dorsal root ganglia caudal to a spinal cord injury with modulated behavioral activity. Sci Data 2019; 6:83. [PMID: 31175296 PMCID: PMC6555821 DOI: 10.1038/s41597-019-0088-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/29/2019] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating clinical condition resulting in significant disabilities. Apart from local injury within the spinal cord, SCI patients develop a myriad of complications including multi-organ dysfunction. Some of the dysfunctions may be directly or indirectly related to the sensory neurons of the dorsal root ganglia (DRG), which signal to both the spinal cord and the peripheral organs. After SCI, some classes of DRG neurons exhibit sensitization and undergo axonal sprouting both peripherally and centrally. Such physiological and anatomical re-organization after SCI contributes to both adaptive and maladaptive plasticity processes, which may be modulated by activity and exercise. In this study, we collected comprehensive gene expression data in whole DRG below the levels of the injury to compare the effects of SCI with and without two different forms of exercise in rats.
Collapse
Affiliation(s)
- Julia H Chariker
- Department of Neuroscience Training, University of Louisville, 522 East Gray Street, Louisville, Kentucky, 40202, USA
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, 522 East Gray Street, Louisville, Kentucky, 40202, USA
| | - Cynthia Gomes
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
| | - Fiona Brabazon
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
- Wiley Publishing, Hoboken, NJ, 07030, USA
| | - Kathryn A Harman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Health & Sport Sciences, University of Louisville, 2100 South Floyd Street, Louisville, KY, 40208, USA
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
| | - David S K Magnuson
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
| | - Scott R Whittemore
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
| | - Jeffrey C Petruska
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA.
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA.
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA.
| | - Eric C Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, 522 East Gray Street, Louisville, Kentucky, 40202, USA.
- Department of Computer Engineering and Computer Science, Speed School of Engineering, University of Louisville, Duthie Center for Engineering, 2301 South 3rd St., Louisville, Kentucky, 40292, USA.
| |
Collapse
|
37
|
Batista CM, Mariano ED, Onuchic F, Dale CS, dos Santos GB, Cristante AF, Otoch JP, Teixeira MJ, Morgalla M, Lepski G. Characterization of traumatic spinal cord injury model in relation to neuropathic pain in the rat. Somatosens Mot Res 2019; 36:14-23. [DOI: 10.1080/08990220.2018.1563537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Chary Marquez Batista
- Department of Neurology, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Eric Domingos Mariano
- Department of Neurology, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Fernando Onuchic
- Department of Neurology, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | | | - Gustavo Bispo dos Santos
- Department of Orthopedic and Traumatology, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Alexandre Fogaça Cristante
- Department of Orthopedic and Traumatology, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Jose Pinhata Otoch
- Department of Surgery, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | | | - Matthias Morgalla
- Department of Neurosurgery, Eberhard-Karls University, Tuebingen, Germany
| | - Guilherme Lepski
- Department of Neurosurgery, Eberhard-Karls University, Tuebingen, Germany
- Department of Psychiatry, School of Medicine, University de São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Keller AV, Hainline C, Rees K, Krupp S, Prince D, Wood BD, Shum-Siu A, Burke DA, Petruska JC, Magnuson DSK. Nociceptor-dependent locomotor dysfunction after clinically-modeled hindlimb muscle stretching in adult rats with spinal cord injury. Exp Neurol 2019; 318:267-276. [PMID: 30880143 DOI: 10.1016/j.expneurol.2019.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 01/10/2023]
Abstract
In the course of investigating how common clinical treatments and adaptive technologies affect recovery after spinal cord injury (SCI), we discovered that a clinically-modeled hindlimb stretching protocol dramatically, but transiently, reduces locomotor function. Nociceptive sensory input is capable of altering motor output at the spinal level, and nociceptive neurons are sensitized after SCI. Here we tested the hypotheses that stretch-induced locomotor deficits are dependent on nociceptive afferents by depleting TRPV1+ sensory afferents using capsaicin injections in neonatal rats. Following maturation, animals received 25g-cm contusive SCI at T10. After plateau of locomotor recovery at 6 weeks, daily stretching was performed for 3 weeks, followed by 2 weeks without stretch, and again for two additional weeks. Animals were sacrificed 2 h after the last stretching session for histological assessments. Consistent with previous findings, stretch-induced drops in locomotor function were observed in nociceptor-intact animals but were nearly absent in nociceptor-depleted animals. These functional changes were accompanied by corresponding increases in the number of c-Fos+ nuclei throughout the lumbar enlargement. As expected, nociceptor-depleted animals had very little CGRP+ axonal innervation of the dorsal horn. Nociceptor-intact stretched animals had significantly higher levels of CGRP+ as compared to non-stretched SCI rats, suggesting that stretching promoted intraspinal CGRP+ sprouting. These results indicate that stretch-induced locomotor dysfunction in animals with incomplete SCI involves C-fibers, adding a negative post-SCI role to their adaptive roles (e.g., bladder control), and suggesting that the clinical use of muscle stretching to combat contractures and spasticity may be unintentionally detrimental to locomotor function.
Collapse
Affiliation(s)
- Anastasia V Keller
- Department of Physiology, University of Louisville, School of Medicine, HSC A 1115, 500 South Preston Street, Louisville, KY 40292, USA
| | - Casey Hainline
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Kathleen Rees
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Sarah Krupp
- Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, 511 South Floyd, Room 111, Louisville, KY 40202, USA
| | - Daniella Prince
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Brittney D Wood
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Alice Shum-Siu
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Jeffrey C Petruska
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, 511 South Floyd, Room 111, Louisville, KY 40202, USA
| | - David S K Magnuson
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, 511 South Floyd, Room 111, Louisville, KY 40202, USA; Department of Physiology, University of Louisville, School of Medicine, HSC A 1115, 500 South Preston Street, Louisville, KY 40292, USA.
| |
Collapse
|
39
|
Yu P, Zhang W, Liu Y, Sheng C, So KF, Zhou L, Zhu H. The effects and potential mechanisms of locomotor training on improvements of functional recovery after spinal cord injury. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 147:199-217. [DOI: 10.1016/bs.irn.2019.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Chhaya SJ, Quiros-Molina D, Tamashiro-Orrego AD, Houlé JD, Detloff MR. Exercise-Induced Changes to the Macrophage Response in the Dorsal Root Ganglia Prevent Neuropathic Pain after Spinal Cord Injury. J Neurotrauma 2018; 36:877-890. [PMID: 30152715 DOI: 10.1089/neu.2018.5819] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Spinal cord injury (SCI) induces neuropathic pain that is refractory to treatment. Central and peripheral immune responses to SCI play critical roles in pain development. Although immune responses in the dorsal horn have been implicated in SCI-pain, immune mechanisms in the periphery, especially in the dorsal root ganglia (DRG), where nociceptor cell bodies reside, have not been well studied. Exercise is an immunomodulator, and we showed previously that early exercise after SCI reduces pain development. However, the mechanisms of exercise-mediated pain reduction are not understood. Therefore, we examined the 1) underlying immune differences in the spinal cord and DRG between rats with and without pain and 2) immunomodulatory effects of exercise in pain reduction. Rats were subjected to a unilateral contusion at C5 and tested for pain development using von Frey and mechanical conflict-avoidance paradigms. A subgroup of rats was exercised on forced running wheels starting at 5 days post-injury for 4 weeks. We observed greater microglial activation in the C7-C8 dorsal horn of rats with SCI-induced pain compared to rats with normal sensation, and early exercise reduced this activation independently of pain behavior. Further, abnormal pain sensation strongly correlated with an increased number of DRG macrophages. Importantly, exercise-treated rats that maintain normal sensation also have a lower number of macrophages in the DRG. Our data suggest that macrophage presence in the DRG may be an important effector of pain development, and early wheel walking exercise may mediate pain prevention by modulating the injury-induced macrophage response in the DRG. Further supportive evidence demonstrated that rats that developed pain despite exercise intervention still displayed a significantly elevated number of macrophages in the DRG. Collectively, these data suggest that macrophage presence in the DRG may be an amenable cellular target for future therapies.
Collapse
Affiliation(s)
- Soha J Chhaya
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - Daniel Quiros-Molina
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - Alessandra D Tamashiro-Orrego
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - John D Houlé
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - Megan Ryan Detloff
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Tashiro S, Nishimura S, Shinozaki M, Takano M, Konomi T, Tsuji O, Nagoshi N, Toyama Y, Liu M, Okano H, Nakamura M. The Amelioration of Pain-Related Behavior in Mice with Chronic Spinal Cord Injury Treated with Neural Stem/Progenitor Cell Transplantation Combined with Treadmill Training. J Neurotrauma 2018; 35:2561-2571. [PMID: 29790403 DOI: 10.1089/neu.2017.5537] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Progress in regenerative medicine is realizing the possibility of neural regeneration and functional recovery in spinal cord injury (SCI). Recently, rehabilitation has attracted much attention with respect to the synergistic promotion of functional recovery in combination with neural stem/progenitor cell (NS/PC) transplantation, even in the chronic refractory phase of SCI. Nevertheless, sensory disturbance is one of the most prominent sequelae, even though the effects of combination or single therapies have been investigated mostly in the context of motor recovery. To determine how combination therapy with treadmill training (TMT) and NS/PC transplantation affects the manifestation of thermal allodynia and tactile hyperalgesia in chronic phase SCI, four groups of SCI mice were used to assess pain-related behavior and histological changes: combined transplantation and TMT therapy, transplantation only, TMT only, and control groups. Thermal allodynia and coarse touch-pressure hyperalgesia exhibited significant recovery in the combined therapy group in comparison with controls, whereas there were no significant differences with fine touch-pressure hyperalgesia and motor function. Further investigation revealed fewer fibers remaining in the posterior funiculus, which contained the tracts associated with the two modalities showing less recovery; that is, touch-pressure hyperalgesia and motor function. A significant correlation was only observed between these two modalities. Although no remarkable histological recovery was found within the lesion epicenter, changes indicating amelioration of pain were observed in the lumbar enlargement of the combination therapy group. Our results suggest that amelioration of thermal allodynia and tactile hyperalgesia can be brought about by the additive effect of NS/PC transplantation and TMT. The degree of recovery seems dependent on the distribution of damage.
Collapse
Affiliation(s)
- Syoichi Tashiro
- 1 Department of Rehabilitation Medicine, Keio University School of Medicine , Tokyo, Japan
| | - Soraya Nishimura
- 2 Department of Orthopaedic Surgery, Keio University School of Medicine , Tokyo, Japan
| | - Munehisa Shinozaki
- 3 Department of Physiology, Keio University School of Medicine , Tokyo, Japan
| | - Morito Takano
- 2 Department of Orthopaedic Surgery, Keio University School of Medicine , Tokyo, Japan
| | - Tsunehiko Konomi
- 2 Department of Orthopaedic Surgery, Keio University School of Medicine , Tokyo, Japan .,4 Department of Orthopaedic Surgery, Murayama Medical Center , National Hospital Organization, Tokyo, Japan
| | - Osahiko Tsuji
- 2 Department of Orthopaedic Surgery, Keio University School of Medicine , Tokyo, Japan
| | - Narihito Nagoshi
- 2 Department of Orthopaedic Surgery, Keio University School of Medicine , Tokyo, Japan
| | - Yoshiaki Toyama
- 2 Department of Orthopaedic Surgery, Keio University School of Medicine , Tokyo, Japan
| | - Meigen Liu
- 1 Department of Rehabilitation Medicine, Keio University School of Medicine , Tokyo, Japan
| | - Hideyuki Okano
- 3 Department of Physiology, Keio University School of Medicine , Tokyo, Japan
| | - Masaya Nakamura
- 2 Department of Orthopaedic Surgery, Keio University School of Medicine , Tokyo, Japan
| |
Collapse
|
42
|
Shiao R, Lee-Kubli CA. Neuropathic Pain After Spinal Cord Injury: Challenges and Research Perspectives. Neurotherapeutics 2018; 15:635-653. [PMID: 29736857 PMCID: PMC6095789 DOI: 10.1007/s13311-018-0633-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuropathic pain is a debilitating consequence of spinal cord injury (SCI) that remains difficult to treat because underlying mechanisms are not yet fully understood. In part, this is due to limitations of evaluating neuropathic pain in animal models in general, and SCI rodents in particular. Though pain in patients is primarily spontaneous, with relatively few patients experiencing evoked pains, animal models of SCI pain have primarily relied upon evoked withdrawals. Greater use of operant tasks for evaluation of the affective dimension of pain in rodents is needed, but these tests have their own limitations such that additional studies of the relationship between evoked withdrawals and operant outcomes are recommended. In preclinical SCI models, enhanced reflex withdrawal or pain responses can arise from pathological changes that occur at any point along the sensory neuraxis. Use of quantitative sensory testing for identification of optimal treatment approach may yield improved identification of treatment options and clinical trial design. Additionally, a better understanding of the differences between mechanisms contributing to at- versus below-level neuropathic pain and neuropathic pain versus spasticity may shed insights into novel treatment options. Finally, the role of patient characteristics such as age and sex in pathogenesis of neuropathic SCI pain remains to be addressed.
Collapse
Affiliation(s)
- Rani Shiao
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines, La Jolla, California, 92073, USA
| | - Corinne A Lee-Kubli
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines, La Jolla, California, 92073, USA.
| |
Collapse
|
43
|
Sliwinski C, Nees TA, Puttagunta R, Weidner N, Blesch A. Sensorimotor Activity Partially Ameliorates Pain and Reduces Nociceptive Fiber Density in the Chronically Injured Spinal Cord. J Neurotrauma 2018; 35:2222-2238. [PMID: 29706124 DOI: 10.1089/neu.2017.5431] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A large proportion of patients suffering from spinal cord injury (SCI) develop chronic central neuropathic pain. Previously, we and others have shown that sensorimotor training early after SCI can prevent the development of mechanical allodynia. To determine whether training initiated in the subchronic/chronic phase remains effective, correlates of below-level neuropathic pain were analyzed in the hindpaws 5-10 weeks after a moderate T11 contusion SCI (50 kDyn) in adult female C57BL/6 mice. In a comparison of SCI and sham mice 5 weeks post-injury, about 80% of injured animals developed mechanical hypersensitivity to light mechanical stimuli, whereas testing of noxious stimuli revealed hypo-responsiveness. Thermal sensitivity testing showed a decreased response latency after injury. Without intervention, mechanical and thermal hyper-responsiveness were evident until the end of the experiment (10 weeks). In contrast, treadmill training (2 × 15 min/day; 5 × /week) initiated 6 weeks post-injury resulted in partial amelioration of pain behavior and this effect remained stable. Analysis of calcitonin gene-related peptide (CGRP)-labeled fibers in lamina III-IV of the lumbar dorsal horn revealed an increase in labeling density after SCI. This was not due to changes in the number or size distribution of CGRP-labeled lumbar dorsal root ganglion neurons. Treadmill training reduced the CGRP-labeling density in the spinal cord of injured mice, whereas the density of non-peptidergic isolectin-B4 (IB4)+ fibers showed no changes in lamina IIi and a slight reduction of sparse IB4 labeling in laminae III-IV. Thus, sensorimotor activity initiated in the subchronic/chronic phase of SCI remains effective in ameliorating pain behavior and influencing structural changes of the nociceptive system.
Collapse
Affiliation(s)
| | - Timo A Nees
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany .,2 Center for Orthopedic and Trauma Surgery, Heidelberg University Hospital , Heidelberg, Germany
| | - Radhika Puttagunta
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany
| | - Norbert Weidner
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany
| | - Armin Blesch
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany .,3 Department of Neurological Surgery and Goodman Campbell Brain and Spine, Stark Neurosciences Research Institute, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
44
|
Faw TD, Lerch JK, Thaxton TT, Deibert RJ, Fisher LC, Basso DM. Unique Sensory and Motor Behavior in Thy1-GFP-M Mice before and after Spinal Cord Injury. J Neurotrauma 2018; 35:2167-2182. [PMID: 29385890 DOI: 10.1089/neu.2017.5395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sensorimotor recovery after spinal cord injury (SCI) is of utmost importance to injured individuals and will rely on improved understanding of SCI pathology and recovery. Novel transgenic mouse lines facilitate discovery, but must be understood to be effective. The purpose of this study was to characterize the sensory and motor behavior of a common transgenic mouse line (Thy1-GFP-M) before and after SCI. Thy1-GFP-M positive (TG+) mice and their transgene negative littermates (TG-) were acquired from two sources (in-house colony, n = 32, Jackson Laboratories, n = 4). C57BL/6J wild-type (WT) mice (Jackson Laboratories, n = 10) were strain controls. Moderate-severe T9 contusion (SCI) or transection (TX) occurred in TG+ (SCI, n = 25, TX, n = 5), TG- (SCI, n = 5), and WT (SCI, n = 10) mice. To determine responsiveness to rehabilitation, a cohort of TG+ mice with SCI (n = 4) had flat treadmill (TM) training 42-49 days post-injury (dpi). To characterize recovery, we performed Basso Mouse Scale, Grid Walk, von Frey Hair, and Plantar Heat Testing before and out to day 42 post-SCI. Open field locomotion was significantly better in the Thy1 SCI groups (TG+ and TG-) compared with WT by 7 dpi (p < 0.01) and was maintained through 42 dpi (p < 0.01). These unexpected locomotor gains were not apparent during grid walking, indicating severe impairment of precise motor control. Thy1 derived mice were hypersensitive to mechanical stimuli at baseline (p < 0.05). After SCI, mechanical hyposensitivity emerged in Thy1 derived groups (p < 0.001), while thermal hyperalgesia occurred in all groups (p < 0.001). Importantly, consistent findings across TG+ and TG- groups suggest that the effects are mediated by the genetic background rather than transgene manipulation itself. Surprisingly, TM training restored mechanical and thermal sensation to baseline levels in TG+ mice with SCI. This behavioral profile and responsiveness to chronic training will be important to consider when choosing models to study the mechanisms underlying sensorimotor recovery after SCI.
Collapse
Affiliation(s)
- Timothy D Faw
- 1 Neuroscience Graduate Program, The Ohio State University , Columbus, Ohio.,2 School of Health and Rehabilitation Sciences, The Ohio State University , Columbus, Ohio.,3 Center for Brain and Spinal Cord Repair, The Ohio State University , Columbus, Ohio
| | - Jessica K Lerch
- 3 Center for Brain and Spinal Cord Repair, The Ohio State University , Columbus, Ohio.,4 Department of Neuroscience, The Ohio State University , Columbus, Ohio
| | - Tyler T Thaxton
- 2 School of Health and Rehabilitation Sciences, The Ohio State University , Columbus, Ohio.,3 Center for Brain and Spinal Cord Repair, The Ohio State University , Columbus, Ohio
| | - Rochelle J Deibert
- 2 School of Health and Rehabilitation Sciences, The Ohio State University , Columbus, Ohio.,3 Center for Brain and Spinal Cord Repair, The Ohio State University , Columbus, Ohio
| | - Lesley C Fisher
- 2 School of Health and Rehabilitation Sciences, The Ohio State University , Columbus, Ohio.,3 Center for Brain and Spinal Cord Repair, The Ohio State University , Columbus, Ohio
| | - D Michele Basso
- 2 School of Health and Rehabilitation Sciences, The Ohio State University , Columbus, Ohio.,3 Center for Brain and Spinal Cord Repair, The Ohio State University , Columbus, Ohio
| |
Collapse
|
45
|
Li H, Kong W, Chambers CR, Yu D, Ganea D, Tuma RF, Ward SJ. The non-psychoactive phytocannabinoid cannabidiol (CBD) attenuates pro-inflammatory mediators, T cell infiltration, and thermal sensitivity following spinal cord injury in mice. Cell Immunol 2018; 329:1-9. [PMID: 29784129 DOI: 10.1016/j.cellimm.2018.02.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/27/2018] [Accepted: 02/28/2018] [Indexed: 01/27/2023]
Abstract
We evaluated the effects of the non-psychoactive cannabinoid cannabidiol (CBD) on the inflammatory response and recovery of function following spinal cord injury (SCI). Female C57Bl/6 mice were exposed to spinal cord contusion injury (T9-10) and received vehicle or CBD (1.5 mg/kg IP) injections for 10 weeks following injury. The effect of SCI and CBD treatment on inflammation was assessed via microarray, qRT-PCR and flow cytometry. Locomotor and bladder function and changes in thermal and mechanical hind paw sensitivity were also evaluated. There was a significant decrease in pro-inflammatory cytokines and chemokines associated with T-cell differentiation and invasion in the SCI-CBD group as well as a decrease in T cell invasion into the injured cord. A higher percentage of SCI mice in the vehicle-treated group (SCI-VEH) went on to develop moderate to severe (0-65.9% baseline thermal threshold) thermal sensitivity as compared with CBD-treated (SCI-CBD) mice. CBD did not affect recovery of locomotor or bladder function following SCI. Taken together, CBD treatment attenuated the development of thermal sensitivity following spinal cord injury and this effect may be related to protection against pathological T-cell invasion.
Collapse
Affiliation(s)
- Hongbo Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States.
| | - Weimin Kong
- Microbiology and Immunology Department, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States.
| | - Christina R Chambers
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States.
| | - Doina Ganea
- Microbiology and Immunology Department, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States.
| | - Ronald F Tuma
- Center for Substance Abuse Research, Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia PA 19140, United States.
| | - Sara Jane Ward
- Center for Substance Abuse Research, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States.
| |
Collapse
|
46
|
Boadas-Vaello P, Homs J, Portero-Tresserra M, Álvarez-Pérez B, Deulofeu M, Verdú E. Graded photochemical spinal cord injury results in chronic hyperalgesia and depression-like behaviour but no anxiety exacerbation in female BALB/c mice. Neurosci Lett 2018; 664:98-106. [DOI: 10.1016/j.neulet.2017.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/29/2017] [Accepted: 11/06/2017] [Indexed: 11/27/2022]
|
47
|
Schneider LE, Henley KY, Turner OA, Pat B, Niedzielko TL, Floyd CL. Application of the Rat Grimace Scale as a Marker of Supraspinal Pain Sensation after Cervical Spinal Cord Injury. J Neurotrauma 2017; 34:2982-2993. [PMID: 27998207 DOI: 10.1089/neu.2016.4665] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Experimental models of neuropathic pain (NP) typically rely on withdrawal responses to assess the presence of pain. Reflexive withdrawal responses to a stimulus are used to evaluate evoked pain and, as such, do not include the assessment of spontaneous NP nor evaluation of the affective and emotional consequences of pain in animal models. Additionally, withdrawal responses can be mediated by spinal cord reflexes and may not accurately indicate supraspinal pain sensation. This is especially true in models of traumatic spinal cord injury (SCI), wherein spastic syndrome, a motor disorder characterized by exaggeration of the stretch reflex that is secondary to hyperexcitability of the spinal reflex, can cause paroxysmal withdrawals not associated with NP sensation. Consequently, the aim of this study was to utilize an assessment of supraspinal pain sensation, the Rat Grimace Scale (RGS), to measure both spontaneous and evoked NP after a contusion SCI at cervical level 5 in adult male rats. Spontaneous and evoked pain were assessed using the RGS to score facial action units before and after the application of a stimulus, respectively. Rodents exhibited significantly higher RGS scores at week 5 post-injury as compared to baseline and laminectomy controls before the application of the stimulus, suggesting the presence of spontaneous NP. Additionally, there was a significant increase in RGS scores after the application of the acetone. These data suggest that the RGS can be used to assess spontaneous NP and determine the presence of evoked supraspinal pain sensation after experimental cervical SCI.
Collapse
Affiliation(s)
- Lonnie E Schneider
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| | - Kathryn Y Henley
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| | - Omari A Turner
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| | - Betty Pat
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| | - Tracy L Niedzielko
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| | - Candace L Floyd
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
48
|
Selective Inhibition of Trigeminovascular Neurons by Fremanezumab: A Humanized Monoclonal Anti-CGRP Antibody. J Neurosci 2017. [PMID: 28642283 DOI: 10.1523/jneurosci.0576-17.2017] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A large body of evidence supports an important role for calcitonin gene-related peptide (CGRP) in migraine pathophysiology. This evidence gave rise to a global effort to develop a new generation of therapeutics that inhibit the interaction of CGRP with its receptor in migraineurs. Recently, a new class of such drugs, humanized anti-CGRP monoclonal antibodies (CGRP-mAbs), were found to be effective in reducing the frequency of migraine. The purpose of this study was to better understand how the CGRP-mAb fremanezumab (TEV-48125) modulates meningeal sensory pathways. To answer this question, we used single-unit recording to determine the effects of fremanezumab (30 mg/kg, IV) and its isotype control Ab on spontaneous and evoked activity in naive and cortical spreading depression (CSD)-sensitized trigeminovascular neurons in the spinal trigeminal nucleus of anesthetized male and female rats. The study demonstrates that, in both sexes, fremanezumab inhibited naive high-threshold (HT) neurons, but not wide-dynamic range trigeminovascular neurons, and that the inhibitory effects on the neurons were limited to their activation from the intracranial dura but not facial skin or cornea. In addition, when given sufficient time, fremanezumab prevents the activation and sensitization of HT neurons by CSD. Mechanistically, these findings suggest that HT neurons play a critical role in the initiation of the perception of headache and the development of cutaneous allodynia and central sensitization. Clinically, the findings may help to explain the therapeutic benefit of CGRP-mAb in reducing headaches of intracranial origin such as migraine with aura and why this therapeutic approach may not be effective for every migraine patient.SIGNIFICANCE STATEMENT Calcitonin gene-related peptide (CGRP) monoclonal antibodies (CGRP-mAbs) are capable of preventing migraine. However, their mechanism of action is unknown. In the current study, we show that, if given enough time, a CGRP-mAb can prevent the activation and sensitization of high-threshold (central) trigeminovascular neurons by cortical spreading depression, but not their activation from the skin or cornea, suggesting a potential explanation for selectivity to migraine headache, but not other pains, and a predominantly peripheral site of action.
Collapse
|
49
|
Huie JR, Morioka K, Haefeli J, Ferguson AR. What Is Being Trained? How Divergent Forms of Plasticity Compete To Shape Locomotor Recovery after Spinal Cord Injury. J Neurotrauma 2017; 34:1831-1840. [PMID: 27875927 DOI: 10.1089/neu.2016.4562] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating syndrome that produces dysfunction in motor and sensory systems, manifesting as chronic paralysis, sensory changes, and pain disorders. The multi-faceted and heterogeneous nature of SCI has made effective rehabilitative strategies challenging. Work over the last 40 years has aimed to overcome these obstacles by harnessing the intrinsic plasticity of the spinal cord to improve functional locomotor recovery. Intensive training after SCI facilitates lower extremity function and has shown promise as a tool for retraining the spinal cord by engaging innate locomotor circuitry in the lumbar cord. As new training paradigms evolve, the importance of appropriate afferent input has emerged as a requirement for adaptive plasticity. The integration of kinematic, sensory, and loading force information must be closely monitored and carefully manipulated to optimize training outcomes. Inappropriate peripheral input may produce lasting maladaptive sensory and motor effects, such as central pain and spasticity. Thus, it is important to closely consider the type of afferent input the injured spinal cord receives. Here we review preclinical and clinical input parameters fostering adaptive plasticity, as well as those producing maladaptive plasticity that may undermine neurorehabilitative efforts. We differentiate between passive (hindlimb unloading [HU], limb immobilization) and active (peripheral nociception) forms of aberrant input. Furthermore, we discuss the timing of initiating exposure to afferent input after SCI for promoting functional locomotor recovery. We conclude by presenting a candidate rapid synaptic mechanism for maladaptive plasticity after SCI, offering a pharmacological target for restoring the capacity for adaptive spinal plasticity in real time.
Collapse
Affiliation(s)
- J Russell Huie
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California
| | - Kazuhito Morioka
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California
| | - Jenny Haefeli
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California
| | - Adam R Ferguson
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California.,2 San Francisco Veterans Affairs Medical Center , San Francisco, California
| |
Collapse
|
50
|
|