51
|
Lind AL, Emami Khoonsari P, Sjödin M, Katila L, Wetterhall M, Gordh T, Kultima K. Spinal Cord Stimulation Alters Protein Levels in the Cerebrospinal Fluid of Neuropathic Pain Patients: A Proteomic Mass Spectrometric Analysis. Neuromodulation 2017; 19:549-62. [PMID: 27513633 DOI: 10.1111/ner.12473] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Electrical neuromodulation by spinal cord stimulation (SCS) is a well-established method for treatment of neuropathic pain. However, the mechanism behind the pain relieving effect in patients remains largely unknown. In this study, we target the human cerebrospinal fluid (CSF) proteome, a little investigated aspect of SCS mechanism of action. METHODS Two different proteomic mass spectrometry protocols were used to analyze the CSF of 14 SCS responsive neuropathic pain patients. Each patient acted as his or her own control and protein content was compared when the stimulator was turned off for 48 hours, and after the stimulator had been used as normal for three weeks. RESULTS Eighty-six proteins were statistically significantly altered in the CSF of neuropathic pain patients using SCS, when comparing the stimulator off condition to the stimulator on condition. The top 12 of the altered proteins are involved in neuroprotection (clusterin, gelsolin, mimecan, angiotensinogen, secretogranin-1, amyloid beta A4 protein), synaptic plasticity/learning/memory (gelsolin, apolipoprotein C1, apolipoprotein E, contactin-1, neural cell adhesion molecule L1-like protein), nociceptive signaling (neurosecretory protein VGF), and immune regulation (dickkopf-related protein 3). CONCLUSION Previously unknown effects of SCS on levels of proteins involved in neuroprotection, nociceptive signaling, immune regulation, and synaptic plasticity are demonstrated. These findings, in the CSF of neuropathic pain patients, expand the picture of SCS effects on the neurochemical environment of the human spinal cord. An improved understanding of SCS mechanism may lead to new tracks of investigation and improved treatment strategies for neuropathic pain.
Collapse
Affiliation(s)
- Anne-Li Lind
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Payam Emami Khoonsari
- Department of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | - Marcus Sjödin
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala//GE Healthcare, Sweden
| | - Lenka Katila
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Magnus Wetterhall
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala//GE Healthcare, Sweden
| | - Torsten Gordh
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| |
Collapse
|
52
|
Walters ET. How is chronic pain related to sympathetic dysfunction and autonomic dysreflexia following spinal cord injury? Auton Neurosci 2017; 209:79-89. [PMID: 28161248 DOI: 10.1016/j.autneu.2017.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/29/2022]
Abstract
Autonomic dysreflexia (AD) and neuropathic pain occur after severe injury to higher levels of the spinal cord. Mechanisms underlying these problems have rarely been integrated in proposed models of spinal cord injury (SCI). Several parallels suggest significant overlap of these mechanisms, although the relationships between sympathetic function (dysregulated in AD) and nociceptive function (dysregulated in neuropathic pain) are complex. One general mechanism likely to be shared is central sensitization - enhanced responsiveness and synaptic reorganization of spinal circuits that mediate sympathetic reflexes or that process and relay pain-related information to the brain. Another is enhanced sensory input to spinal circuits caused by extensive alterations in primary sensory neurons. Both AD and SCI-induced neuropathic pain are associated with spinal sprouting of peptidergic nociceptors that might increase synaptic input to the circuits involved in AD and SCI pain. In addition, numerous nociceptors become hyperexcitable, hypersensitive to chemicals associated with injury and inflammation, and spontaneously active, greatly amplifying sensory input to sensitized spinal circuits. As discussed with the aid of a preliminary functional model, these effects are likely to have mutually reinforcing relationships with each other, and with consequences of SCI-induced interruption of descending excitatory and inhibitory influences on spinal circuits, with SCI-induced inflammation in the spinal cord and in DRGs, and with activity in sympathetic fibers within DRGs that promotes local inflammation and spontaneous activity in sensory neurons. This model suggests that interventions selectively targeting hyperactivity in C-nociceptors might be useful for treating chronic pain and AD after high SCI.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA.
| |
Collapse
|
53
|
Kramer JLK, Minhas NK, Jutzeler CR, Erskine ELKS, Liu LJW, Ramer MS. Neuropathic pain following traumatic spinal cord injury: Models, measurement, and mechanisms. J Neurosci Res 2016; 95:1295-1306. [PMID: 27617844 DOI: 10.1002/jnr.23881] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/19/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023]
Abstract
Neuropathic pain following spinal cord injury (SCI) is notoriously difficult to treat and is a high priority for many in the SCI population. Resolving this issue requires animal models fidelic to the clinical situation in terms of injury mechanism and pain phenotype. This Review discusses the means by which neuropathic pain has been induced and measured in experimental SCI and compares these with human outcomes, showing that there is a substantial disconnection between experimental investigations and clinical findings in a number of features. Clinical injury level is predominantly cervical, whereas injury in the laboratory is modeled mainly at the thoracic cord. Neuropathic pain is primarily spontaneous or tonic in people with SCI (with a relatively smaller incidence of allodynia), but measures of evoked responses (to thermal and mechanical stimuli) are almost exclusively used in animals. There is even the question of whether pain per se has been under investigation in most experimental SCI studies rather than simply enhanced reflex activity with no affective component. This Review also summarizes some of the problems related to clinical assessment of neuropathic pain and how advanced imaging techniques may circumvent a lack of patient/clinician objectivity and discusses possible etiologies of neuropathic pain following SCI based on evidence from both clinical studies and animal models, with examples of cellular and molecular changes drawn from the entire neuraxis from primary afferent terminals to cortical sensory and affective centers. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- John L K Kramer
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Nikita K Minhas
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Catherine R Jutzeler
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Erin L K S Erskine
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa J W Liu
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Matt S Ramer
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
54
|
Qiu Y, Chen WY, Wang ZY, Liu F, Wei M, Ma C, Huang YG. Simvastatin Attenuates Neuropathic Pain by Inhibiting the RhoA/LIMK/Cofilin Pathway. Neurochem Res 2016; 41:2457-2469. [DOI: https:/doi.org/10.1007/s11064-016-1958-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
|
55
|
A data science approach to candidate gene selection of pain regarded as a process of learning and neural plasticity. Pain 2016; 157:2747-2757. [DOI: 10.1097/j.pain.0000000000000694] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
56
|
Simvastatin Attenuates Neuropathic Pain by Inhibiting the RhoA/LIMK/Cofilin Pathway. Neurochem Res 2016; 41:2457-69. [PMID: 27216618 DOI: 10.1007/s11064-016-1958-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/26/2016] [Accepted: 05/12/2016] [Indexed: 12/21/2022]
Abstract
Neuropathic pain occurs due to deleterious changes in the nervous system caused by a lesion or dysfunction. Currently, neuropathic pain management is unsatisfactory and remains a challenge in clinical practice. Studies have suggested that actin cytoskeleton remodeling may be associated with neural plasticity and may involve a nociceptive mechanism. Here, we found that the RhoA/LIM kinase (LIMK)/cofilin pathway, which regulates actin dynamics, was activated after chronic constriction injury (CCI) of the sciatic nerve. Treatments that reduced RhoA/LIMK/cofilin pathway activity, including simvastatin, the Rho kinase inhibitor Y-27632, and the synthetic peptide Tat-S3, attenuated actin filament disruption in the dorsal root ganglion and CCI-induced neuropathic pain. Over-activation of the cytoskeleton caused by RhoA/LIMK/cofilin pathway activation may produce a scaffold for the trafficking of nociceptive signaling factors, leading to chronic neuropathic pain. Here, we found that simvastatin significantly decreased the ratio of membrane/cytosolic RhoA, which was significantly increased after CCI, by inhibiting the RhoA/LIMK/cofilin pathway. This effect was highly dependent on the function of the cytoskeleton as a scaffold for signal trafficking. We conclude that simvastatin attenuated neuropathic pain in rats subjected to CCI by inhibiting actin-mediated intracellular trafficking to suppress RhoA/LIMK/cofilin pathway activity.
Collapse
|
57
|
Hansen CN, Norden DM, Faw TD, Deibert R, Wohleb ES, Sheridan JF, Godbout JP, Basso DM. Lumbar Myeloid Cell Trafficking into Locomotor Networks after Thoracic Spinal Cord Injury. Exp Neurol 2016; 282:86-98. [PMID: 27191729 DOI: 10.1016/j.expneurol.2016.05.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/28/2016] [Accepted: 05/13/2016] [Indexed: 01/05/2023]
Abstract
Spinal cord injury (SCI) promotes inflammation along the neuroaxis that jeopardizes plasticity, intrinsic repair and recovery. While inflammation at the injury site is well-established, less is known within remote spinal networks. The presence of bone marrow-derived immune (myeloid) cells in these areas may further impede functional recovery. Previously, high levels of the gelatinase, matrix metalloproteinase-9 (MMP-9) occurred within the lumbar enlargement after thoracic SCI and impeded activity-dependent recovery. Since SCI-induced MMP-9 potentially increases vascular permeability, myeloid cell infiltration may drive inflammatory toxicity in locomotor networks. Therefore, we examined neurovascular reactivity and myeloid cell infiltration in the lumbar cord after thoracic SCI. We show evidence of region-specific recruitment of myeloid cells into the lumbar but not cervical region. Myeloid infiltration occurred with concomitant increases in chemoattractants (CCL2) and cell adhesion molecules (ICAM-1) around lumbar vasculature 24h and 7days post injury. Bone marrow GFP chimeric mice established robust infiltration of bone marrow-derived myeloid cells into the lumbar gray matter 24h after SCI. This cell infiltration occurred when the blood-spinal cord barrier was intact, suggesting active recruitment across the endothelium. Myeloid cells persisted as ramified macrophages at 7days post injury in parallel with increased inhibitory GAD67 labeling. Importantly, macrophage infiltration required MMP-9.
Collapse
Affiliation(s)
- Christopher N Hansen
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Diana M Norden
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Timothy D Faw
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Rochelle Deibert
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Eric S Wohleb
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Division of Biosciences, , The Ohio State University, Columbus, OH 43210, USA.
| | - John F Sheridan
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Division of Biosciences, , The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan P Godbout
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA
| | - D Michele Basso
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
58
|
Wu J, Zhang Y, Yang P, Enkhjargal B, Manaenko A, Tang J, Pearce WJ, Hartman R, Obenaus A, Chen G, Zhang JH. Recombinant Osteopontin Stabilizes Smooth Muscle Cell Phenotype via Integrin Receptor/Integrin-Linked Kinase/Rac-1 Pathway After Subarachnoid Hemorrhage in Rats. Stroke 2016; 47:1319-27. [PMID: 27006454 DOI: 10.1161/strokeaha.115.011552] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 02/22/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND PURPOSE Recombinant osteopontin (rOPN) has been reported to be neuroprotective in stroke animal models. The purpose of this study is to investigate a potential role and mechanism of nasal administration of rOPN on preserving the vascular smooth muscle phenotype in early brain injury after subarachnoid hemorrhage (SAH). METHODS One hundred and ninety-two male adult Sprague-Dawley rats were used. The SAH model was induced by endovascular perforation. Integrin-linked kinase small interfering RNA was intracerebroventricularly injected 48 hours before SAH. The integrin receptor antagonist fibronectin-derived peptide Gly-Arg-Gly-Asp-Ser-Pro (GRGDSP), focal adhesion kinase inhibitor Fib-14, and Rac-1 inhibitor NSC23766 were administered 1 hour before SAH induction. rOPN was administered via the intracerebroventricular and nasal route after SAH. SAH grade, neurological scores, brain water content, brain swelling, hematoxylin and eosin staining, India ink angiography, Western blots, and immunofluorescence were used to study the mechanisms of rOPN on the vascular smooth muscle phenotypic transformation. RESULTS The marker proteins of vascular smooth muscle phenotypic transformation α-smooth muscle actin decreased and embryonic smooth muscle myosin heavy chain (SMemb) increased significantly at 24 and 72 hours in the cerebral arteries after SAH. rOPN prevented the changes of α-smooth muscle actin and SMemb and significantly alleviated neurobehavioral dysfunction, increased the cross-sectional area and the lumen diameter of the cerebral arteries, reduced the brain water content and brain swelling, and improved the wall thickness of cerebral arteries. These effects of rOPN were abolished by GRGDSP, integrin-linked kinase small interfering RNA, and NSC23766. Intranasal application of rOPN at 3 hours after SAH also reduced neurological deficits. CONCLUSIONS rOPN prevented the vascular smooth muscle phenotypic transformation and improved the neurological outcome, which was possibly mediated by the integrin receptor/integrin-linked kinase/Rac-1 pathway.
Collapse
Affiliation(s)
- Jiang Wu
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Yang Zhang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Peng Yang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Budbazar Enkhjargal
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Anatol Manaenko
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Jiping Tang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - William J Pearce
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Richard Hartman
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Andre Obenaus
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Gang Chen
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA.
| | - John H Zhang
- From the Department of Neurosurgery (J.W., G.C.), the First Affiliated Hospital of Soochow University, Suzhou, China; and Department of Physiology (J.W., Y.Z., P.Y., B.E., A.M., J.T., W.J.P., R.H., A.O., J.H.Z.), School of Behavioral Science (R.H.), Department of Pediatrics (A.O.), and Department of Anesthesiology (J.H.Z.), Loma Linda University, CA.
| |
Collapse
|
59
|
Li HS, Ke J, Zhao GZ, Wu LA, Kou JP, Liu HC. Effects of Danggui-Shaoyao-San on the Influence of Spatial Learning and Memory Induced by Experimental Tooth Movement. Chin Med J (Engl) 2016; 128:1948-55. [PMID: 26168837 PMCID: PMC4717939 DOI: 10.4103/0366-6999.160562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The pain caused by orthodontic treatment has been considered as tough problems in orthodontic practice. There is substantial literature on pain which has exactly effected on learning and memory; orthodontic tooth movement affected the emotional status has been showed positive outcomes. Danggui-Shaoyao-San (DSS) is a Traditional Chinese Medicine prescription that has been used for pain treatment and analgesic effect for orthodontic pain via inhibiting the activations of neuron and glia. We raised the hypothesis that DSS could restore the impaired abilities of spatial learning and memory via regulating neuron or glia expression in the hippocampus. METHODS A total of 36 rats were randomly divided into three groups: (1) Sham group (n = 12), rats underwent all the operation procedure except for the placement of orthodontic forces and received saline treatment; (2) experimental tooth movement (ETM) group (n = 12), rats received saline treatment and ETM; (3) DSS + ETM (DETM) group (n = 12), rats received DSS treatment and ETM. All DETM group animals were administered with DSS at a dose of 150 mg/kg. Morris water maze test was evaluated; immunofluorescent histochemistry was used to identify astrocytes activation, and immunofluorescent dendritic spine analysis was used to identify the dendritic spines morphological characteristics expression levels in hippocampus. RESULTS Maze training sessions during the 5 successive days revealed that ETM significantly deficits in progressive learning in rats, DSS that was given from day 5 prior to ETM enhanced progressive learning. The ETM group rats took longer to cross target quadrant during the probe trial and got less times to cross-platform than DETM group. The spine density in hippocampus in ETM group was significantly decreased compared to the sham group. In addition, thin and mature spine density were decreased too. However, the DSS administration could reverse the dendritic shrinkage and increase the spine density compared to the ETM group. Astrocytes activation showed the opposite trend in hippocampus dentate gyrus (DG). CONCLUSIONS Treatment with DSS could restore the impaired abilities on ETM-induced decrease of learning and memory behavior. The decreased spines density in the hippocampus and astrocytes activation in DG of hippocampus in the ETM group rats may be related with the decline of the ability of learning and memory. The ability to change the synaptic plasticity in hippocampus after DSS administration may be correlated with the alleviation of impairment of learn and memory after ETM treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Hong-Chen Liu
- Institute of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
60
|
Zhao P, Hill M, Liu S, Chen L, Bangalore L, Waxman SG, Tan AM. Dendritic spine remodeling following early and late Rac1 inhibition after spinal cord injury: evidence for a pain biomarker. J Neurophysiol 2016; 115:2893-910. [PMID: 26936986 DOI: 10.1152/jn.01057.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/01/2016] [Indexed: 12/15/2022] Open
Abstract
Neuropathic pain is a significant complication following spinal cord injury (SCI) with few effective treatments. Drug development for neuropathic pain often fails because preclinical studies do not always translate well to clinical conditions. Identification of biological characteristics predictive of disease state or drug responsiveness could facilitate more effective clinical translation. Emerging evidence indicates a strong correlation between dendritic spine dysgenesis and neuropathic pain. Because dendritic spines are located on dorsal horn neurons within the spinal cord nociceptive system, dendritic spine remodeling provides a unique opportunity to understand sensory dysfunction after SCI. In this study, we provide support for the postulate that dendritic spine profiles can serve as biomarkers for neuropathic pain. We show that dendritic spine profiles after SCI change to a dysgenic state that is characteristic of neuropathic pain in a Rac1-dependent manner. Suppression of the dysgenic state through inhibition of Rac1 activity is accompanied by attenuation of neuropathic pain. Both dendritic spine dysgenesis and neuropathic pain return when inhibition of Rac1 activity is lifted. These findings suggest the utility of dendritic spines as structural biomarkers for neuropathic pain.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Myriam Hill
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Shujun Liu
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Lubin Chen
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Lakshmi Bangalore
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Andrew M Tan
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
61
|
Wang Y, Lu YF, Li CL, Sun W, Li Z, Wang RR, He T, Yang F, Yang Y, Wang XL, Guan SM, Chen J. Involvement of Rac1 signalling pathway in the development and maintenance of acute inflammatory pain induced by bee venom injection. Br J Pharmacol 2016; 173:937-50. [PMID: 26700000 DOI: 10.1111/bph.13413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The Rho GTPase, Rac1, is involved in the pathogenesis of neuropathic pain induced by malformation of dendritic spines in the spinal dorsal horn (sDH) neurons. In the present study, the contribution of spinal Rac1 to peripheral inflammatory pain was studied. EXPERIMENTAL APPROACH Effects of s.c. bee venom (BV) injection on cellular localization of Rac1 in the rat sDH was determined with double labelling immunofluorescence. Activation of Rac1 and its downstream effector p21-activated kinase (PAK), ERKs and p38 MAPK in inflammatory pain states was evaluated with a pull-down assay and Western blotting. The preventive and therapeutic analgesic effects of intrathecal administration of NSC23766, a selective inhibitor of Rac1, on BV-induced spontaneous nociception and pain hypersensitivity were investigated. KEY RESULTS Rac1 labelling was mainly localized within neurons in both the superficial and deep layers of the sDH in rats of naïve, vehicle-treated and inflamed (BV injected) groups. GTP-Rac1-PAK and ERKs/p38 were activated following s.c. BV injection. Post-treatment with intrathecal NSC23766 significantly inhibited GTP-Rac1 activity and phosphorylation of Rac1-PAK, ERKs and p38 MAPK in the sDH. Both pre-treatment and post-treatment with intrathecal NSC23766 dose-dependently attenuated the paw flinches, primary thermal and mechanical hyperalgesia and the mirror-image thermal hyperalgesia induced by BV injection, but without affecting the baseline pain sensitivity and motor coordination. CONCLUSIONS AND IMPLICATIONS The spinal GTP-Rac1-PAK-ERK/p38MAPK signalling pathway is involved in both the development and maintenance of peripheral inflammatory pain and can be used as a potential molecular target for developing a novel therapeutic strategy for clinical pain.
Collapse
Affiliation(s)
- Yan Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Yun-Fei Lu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Chun-Li Li
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Wei Sun
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Zhen Li
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Rui-Rui Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Ting He
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Fan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China
| | - Su-Min Guan
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Key Laboratory of Brain Stress and Behavior, PLA, Xi'an, 710038, China.,Beijing Institute for Brain Disorders, Beijing, 100069, China
| |
Collapse
|
62
|
Zhang K, Zhang J, Zhou Y, Chen C, Li W, Ma L, Zhang L, Zhao J, Gan W, Zhang L, Tang P. Remodeling the Dendritic Spines in the Hindlimb Representation of the Sensory Cortex after Spinal Cord Hemisection in Mice. PLoS One 2015; 10:e0132077. [PMID: 26132157 PMCID: PMC4489092 DOI: 10.1371/journal.pone.0132077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 06/09/2015] [Indexed: 01/01/2023] Open
Abstract
Spinal cord injury (SCI) can induce remodeling of multiple levels of the cerebral cortex system especially in the sensory cortex. The aim of this study was to assess, in vivo and bilaterally, the remodeling of dendritic spines in the hindlimb representation of the sensory cortex after spinal cord hemisection. Thy1-YFP transgenic mice were randomly divided into the control group and the SCI group, and the spinal vertebral plates (T11–T12) of all mice were excised. Next, the left hemisphere of the spinal cord (T12) was hemisected in the SCI group. The hindlimb representations of the sensory cortex in both groups were imaged bilaterally on the day before (0d), and three days (3d), two weeks (2w), and one month (1m) after the SCI. The rates of stable, newly formed, and eliminated spines were calculated by comparing images of individual dendritic spine in the same areas at different time points. In comparison to the control group, the rate of newly formed spines in the contralateral sensory cortex of the SCI group increased at three days and two weeks after injury. The rates of eliminated spines in the bilateral sensory cortices increased and the rate of stable spines in the bilateral cortices declined at two weeks and one month. From three days to two weeks, the stable rates of bilaterally stable spines in the SCI group decreased. In comparison to the control group and contralateral cortex in the SCI group, the re-emerging rate of eliminated spines in ipsilateral cortex of the SCI group decreased significantly. The stable rates of newly formed spines in bilateral cortices of the SCI group decreased from two weeks to one month. We found that the remodeling in the hindlimb representation of the sensory cortex after spinal cord hemisection occurred bilaterally. This remodeling included eliminating spines and forming new spines, as well as changing the reorganized regions of the brain cortex after the SCI over time. Soon after the SCI, the cortex was remodeled by increasing spine formation in the contralateral cortex. Then it was remodeled prominently by eliminating spines of bilateral cortices. Spinal cord hemisection also caused traditional stable spines to become unstable and led the eliminated spines even more hard to recur especially in the ipsilateral cortex of the SCI group. In addition, it also made the new formed spines unstable.
Collapse
Affiliation(s)
- Kexue Zhang
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Jinhui Zhang
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Yanmei Zhou
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, People's Republic of China
| | - Chao Chen
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Wei Li
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, People's Republic of China
| | - Lei Ma
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, People's Republic of China
| | - Licheng Zhang
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Jingxin Zhao
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Wenbiao Gan
- Department of Physiology and Neuroscience, New York University School of Medicine, New York, New York, 10016, United States of America
| | - Lihai Zhang
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, People's Republic of China
| | - Peifu Tang
- Department of Orthopaedics, General Hospital of Chinese PLA, Beijing, 100853, People's Republic of China
| |
Collapse
|
63
|
Argüello EJ, Silva RJ, Huerta MK, Avila RS. Computational modeling of peripheral pain: a commentary. Biomed Eng Online 2015; 14:56. [PMID: 26062616 PMCID: PMC4464699 DOI: 10.1186/s12938-015-0049-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/08/2015] [Indexed: 01/18/2023] Open
Abstract
This commentary is intended to find possible explanations for the low impact of computational modeling on pain research. We discuss the main strategies that have been used in building computational models for the study of pain. The analysis suggests that traditional models lack biological plausibility at some levels, they do not provide clinically relevant results, and they cannot capture the stochastic character of neural dynamics. On this basis, we provide some suggestions that may be useful in building computational models of pain with a wider range of applications.
Collapse
Affiliation(s)
- Erick J Argüello
- Laboratorio "C" at Universidad Simón Bolívar, Caracas, Venezuela.
| | - Ricardo J Silva
- Secretaría de Educación Superior, Ciencia, Tecnología e Innovación (SENESCYT), Guayaquil, Ecuador. .,Programa Promeinfo, Universidad de Guayaquil, Guayaquil, Ecuador.
| | - Mónica K Huerta
- Secretaría de Educación Superior, Ciencia, Tecnología e Innovación (SENESCYT), Guayaquil, Ecuador. .,Universidad Politécnica Salesiana (UPS), Cuenca, Ecuador. .,Grupo de Redes y Telemática Aplicada at Universidad Simón Bolívar, Caracas, Venezuela.
| | - René S Avila
- Universidad Politécnica Salesiana (UPS), Cuenca, Ecuador.
| |
Collapse
|
64
|
Tejada-Simon MV. Modulation of actin dynamics by Rac1 to target cognitive function. J Neurochem 2015; 133:767-79. [PMID: 25818528 DOI: 10.1111/jnc.13100] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/11/2015] [Accepted: 03/14/2015] [Indexed: 12/14/2022]
Abstract
The small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in cells. Formation of extensions at the surface of the cell is required for migration and even for cell invasion and metastases. Because an elevated level and hyperactivation of this protein has been associated with metastasis in cancer, direct regulators of Rac1 are currently envisioned as a potential strategy to treat certain cancers. Less research, however, has been done regarding the role of this small GTP-binding protein in brain development, where it has an important role in dendritic spine morphogenesis through the regulation of actin. Alteration of dendritic development and spinogenesis has been often associated with mental disorders. Rac1 is associated with and required for learning and the formation of memories in the brain. Rac1 appears to be dysregulated in certain neurodevelopmental disorders that present all these three alterations: mental retardation, atypical synaptic plasticity and aberrant spine morphology. Thus, to develop novel therapies for rescuing cognitive impairment, a reasonable approach might be to target this protein, Rac1, which plays a pivotal role in directing signals that regulate actin dynamics, which in turn might have an effect in spine cytoarchitecture and synaptic function. It is possible that novel drugs that regulate Rac1 activation and function could modulate actin cytoskeleton and spine dynamics, representing potential candidates to repair intellectual disability in disorders associated with spine abnormalities. Herein, we present a list of the current Rac1 inhibitors that might fulfill this role together with a summary of the latest findings concerning their function as they relate to neuronal studies. While the small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in different type of cells, it appears to be also required for learning and the formation of memories in the brain. Abnormal regulation of this protein has been associated with cognitive disabilities, atypical synaptic plasticity and abnormal morphology of dendritic spines in certain neurodevelopmental disorders. Thus, modulation of Rac1 activity using novel inhibitors might be a strategy to reestablish cognitive function.
Collapse
Affiliation(s)
- Maria V Tejada-Simon
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA.,Department of Biology, University of Houston, Houston, Texas, USA.,Department of Psychology, University of Houston, Houston, Texas, USA.,Biology of Behavior Institute (BoBI), University of Houston, Houston, Texas, USA
| |
Collapse
|
65
|
A role for Kalirin-7 in nociceptive sensitization via activity-dependent modulation of spinal synapses. Nat Commun 2015; 6:6820. [PMID: 25865668 PMCID: PMC4403379 DOI: 10.1038/ncomms7820] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/03/2015] [Indexed: 12/20/2022] Open
Abstract
Synaptic plasticity is the cornerstone of processes underlying persistent nociceptive activity-induced changes in normal nociceptive sensitivity. Kalirin-7 is a multifunctional guanine-nucleotide-exchange factor (GEF) for Rho GTPases that is characterized by its localization at excitatory synapses, interactions with glutamate receptors and its ability to dynamically modulate the neuronal cytoskeleton. Here we show that spinally expressed Kalirin-7 is required for persistent nociceptive activity-dependent synaptic long-term potentiation as well as activity-dependent remodelling of synaptic spines in the spinal dorsal horn, thereby orchestrating functional and structural plasticity during the course of inflammatory pain.
Collapse
|
66
|
|
67
|
|
68
|
Tan AM. Dendritic spine dysgenesis in neuropathic pain. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:385-408. [PMID: 25744680 DOI: 10.1016/bs.pmbts.2014.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The failure of neuropathic pain to abate even years after trauma suggests that adverse changes to synaptic function must exist in a chronic pathological state in nociceptive pathways. The chronicity of neuropathic pain therefore underscores the importance of understanding the contribution of dendritic spines--micron-sized postsynaptic structures that represent modifiable sites of synaptic contact. Historically, dendritic spines have been of great interest to the learning and memory field. More recent evidence points to the exciting implication that abnormal dendritic spine structure following disease or injury may represent a "molecular memory" for maintaining chronic pain. Dendritic spine dysgenesis in dorsal horn neurons contributes to nociceptive hyperexcitability associated with neuropathic pain, as demonstrated in multiple pain models, i.e., spinal cord injury, peripheral nerve injury, diabetic neuropathy, and thermal burn injury. Because of the relationship between dendritic spine structure and neuronal function, a thorough investigation of dendritic spine behavior in the spinal cord is a unique opportunity to better understand the mechanisms of sensory dysfunction after injury or disease. At a conceptual level, a spinal memory mechanism that engages dendritic spine remodeling would also contribute to a broad range of intractable neurological conditions. Molecules involved in regulating dendritic spine plasticity may offer novel targets for the development of effective and durable therapies for neurological disease.
Collapse
Affiliation(s)
- Andrew Michael Tan
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA; Hopkins School, New Haven, Connecticut, USA.
| |
Collapse
|
69
|
Price TJ, Inyang KE. Commonalities between pain and memory mechanisms and their meaning for understanding chronic pain. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:409-34. [PMID: 25744681 DOI: 10.1016/bs.pmbts.2014.11.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Pain sensing neurons in the periphery (called nociceptors) and the central neurons that receive their projections show remarkable plasticity following injury. This plasticity results in amplification of pain signaling that is now understood to be crucial for the recovery and survival of organisms following injury. These same plasticity mechanisms may drive a transition to a nonadaptive chronic pain state if they fail to resolve following the termination of the healing process. Remarkable advances have been achieved in the past two decades in understanding the molecular mechanisms that underlie pain plasticity following injury. The mechanisms bear a striking resemblance to molecular mechanisms involved in learning and memory processes in other brain regions, including the hippocampus and cerebral cortex. Here those mechanisms, their commonalities and subtle differences, will be highlighted and their role in causing chronic pain will be discussed. Arising from these data is the striking argument that chronic pain is a disease of the nervous system, which distinguishes this phenomena from acute pain that is frequently a symptom alerting the organism to injury. This argument has important implications for the development of disease modifying therapeutics.
Collapse
Affiliation(s)
- Theodore J Price
- Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA.
| | - Kufreobong E Inyang
- Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
70
|
Bandaru SP, Liu S, Waxman SG, Tan AM. Dendritic spine dysgenesis contributes to hyperreflexia after spinal cord injury. J Neurophysiol 2014; 113:1598-615. [PMID: 25505110 DOI: 10.1152/jn.00566.2014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hyperreflexia and spasticity are chronic complications in spinal cord injury (SCI), with limited options for safe and effective treatment. A central mechanism in spasticity is hyperexcitability of the spinal stretch reflex, which presents symptomatically as a velocity-dependent increase in tonic stretch reflexes and exaggerated tendon jerks. In this study we tested the hypothesis that dendritic spine remodeling within motor reflex pathways in the spinal cord contributes to H-reflex dysfunction indicative of spasticity after contusion SCI. Six weeks after SCI in adult Sprague-Dawley rats, we observed changes in dendritic spine morphology on α-motor neurons below the level of injury, including increased density, altered spine shape, and redistribution along dendritic branches. These abnormal spine morphologies accompanied the loss of H-reflex rate-dependent depression (RDD) and increased ratio of H-reflex to M-wave responses (H/M ratio). Above the level of injury, spine density decreased compared with below-injury spine profiles and spine distributions were similar to those for uninjured controls. As expected, there was no H-reflex hyperexcitability above the level of injury in forelimb H-reflex testing. Treatment with NSC23766, a Rac1-specific inhibitor, decreased the presence of abnormal dendritic spine profiles below the level of injury, restored RDD of the H-reflex, and decreased H/M ratios in SCI animals. These findings provide evidence for a novel mechanistic relationship between abnormal dendritic spine remodeling in the spinal cord motor system and reflex dysfunction in SCI.
Collapse
Affiliation(s)
- Samira P Bandaru
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Shujun Liu
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Andrew M Tan
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
71
|
Tan AM, Waxman SG. Dendritic spine dysgenesis in neuropathic pain. Neurosci Lett 2014; 601:54-60. [PMID: 25445354 DOI: 10.1016/j.neulet.2014.11.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/12/2014] [Accepted: 11/15/2014] [Indexed: 12/20/2022]
Abstract
Neuropathic pain is a significant unmet medical need in patients with variety of injury or disease insults to the nervous system. Neuropathic pain often presents as a painful sensation described as electrical, burning, or tingling. Currently available treatments have limited effectiveness and narrow therapeutic windows for safety. More powerful analgesics, e.g., opioids, carry a high risk for chemical dependence. Thus, a major challenge for pain research is the elucidation of the mechanisms that underlie neuropathic pain and developing targeted strategies to alleviate pathological pain. The mechanistic link between dendritic spine structure and circuit function could explain why neuropathic pain is difficult to treat, since nociceptive processing pathways are adversely "hard-wired" through the reorganization of dendritic spines. Several studies in animal models of neuropathic pain have begun to reveal the functional contribution of dendritic spine dysgenesis in neuropathic pain. Previous reports have demonstrated three primary changes in dendritic spine structure on nociceptive dorsal horn neurons following injury or disease, which accompany chronic intractable pain: (I) increased density of dendritic spines, particularly mature mushroom-spine spines, (II) redistribution of spines toward dendritic branch locations close to the cell body, and (III) enlargement of the spine head diameter, which generally presents as a mushroom-shaped spine. Given the important functional implications of spine distribution, density, and shape for synaptic and neuronal function, the study of dendritic spine abnormality may provide a new perspective for investigating pain, and the identification of specific molecular players that regulate spine morphology may guide the development of more effective and long-lasting therapies.
Collapse
Affiliation(s)
- Andrew M Tan
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Neurology and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Neurology and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
72
|
Walters ET. Neuroinflammatory contributions to pain after SCI: roles for central glial mechanisms and nociceptor-mediated host defense. Exp Neurol 2014; 258:48-61. [PMID: 25017887 DOI: 10.1016/j.expneurol.2014.02.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/23/2014] [Accepted: 02/02/2014] [Indexed: 12/30/2022]
Abstract
Neuropathic pain after spinal cord injury (SCI) is common, often intractable, and can be severely debilitating. A number of mechanisms have been proposed for this pain, which are discussed briefly, along with methods for revealing SCI pain in animal models, such as the recently applied conditioned place preference test. During the last decade, studies of animal models have shown that both central neuroinflammation and behavioral hypersensitivity (indirect reflex measures of pain) persist chronically after SCI. Interventions that reduce neuroinflammation have been found to ameliorate pain-related behavior, such as treatment with agents that inhibit the activation states of microglia and/or astroglia (including IL-10, minocycline, etanercept, propentofylline, ibudilast, licofelone, SP600125, carbenoxolone). Reversal of pain-related behavior has also been shown with disruption by an inhibitor (CR8) and/or genetic deletion of cell cycle-related proteins, deletion of a truncated receptor (trkB.T1) for brain-derived neurotrophic factor (BDNF), or reduction by antisense knockdown or an inhibitor (AMG9810) of the activity of channels (TRPV1 or Nav1.8) important for electrical activity in primary nociceptors. Nociceptor activity is known to drive central neuroinflammation in peripheral injury models, and nociceptors appear to be an integral component of host defense. Thus, emerging results suggest that spinal and systemic effects of SCI can activate nociceptor-mediated host defense responses that interact via neuroinflammatory signaling with complex central consequences of SCI to drive chronic pain. This broader view of SCI-induced neuroinflammation suggests new targets, and additional complications, for efforts to develop effective treatments for neuropathic SCI pain.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, University of Texas Medical School at Houston, TX, USA.
| |
Collapse
|
73
|
Hagenston AM, Simonetti M. Neuronal calcium signaling in chronic pain. Cell Tissue Res 2014; 357:407-26. [PMID: 25012522 DOI: 10.1007/s00441-014-1942-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/03/2014] [Indexed: 01/03/2023]
Abstract
Acute physiological pain, the unpleasant sensory response to a noxious stimulus, is essential for animals and humans to avoid potential injury. Pathological pain that persists after the original insult or injury has subsided, however, not only results in individual suffering but also imposes a significant cost on society. Improving treatments for long-lasting pathological pain requires a comprehensive understanding of the biological mechanisms underlying pain perception and the development of pain chronicity. In this review, we aim to highlight some of the major findings related to the involvement of neuronal calcium signaling in the processes that mediate chronic pain.
Collapse
Affiliation(s)
- Anna M Hagenston
- University of Heidelberg, Neurobiology, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany,
| | | |
Collapse
|
74
|
Wang C, Wang H, Pang J, Li L, Zhang S, Song G, Li N, Cao J, Zhang L. Glial Cell-Derived Neurotrophic Factor Attenuates Neuropathic Pain in a Mouse Model of Chronic Constriction Injury: Possible Involvement of E-cadherin/p120ctn Signaling. J Mol Neurosci 2014; 54:156-63. [DOI: 10.1007/s12031-014-0266-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 02/14/2014] [Indexed: 12/22/2022]
|
75
|
Ommaya AK, Adams KM, Allman RM, Collins EG, Cooper RA, Dixon CE, Fishman PS, Henry JA, Kardon R, Kerns RD, Kupersmith J, Lo A, Macko R, McArdle R, McGlinchey RE, McNeil MR, O'Toole TP, Peckham PH, Tuszynski MH, Waxman SG, Wittenberg GF. Guest editorial: Opportunities in rehabilitation research. ACTA ACUST UNITED AC 2013; 50:vii-xxxii. [PMID: 24203548 DOI: 10.1682/jrrd.2012.09.0167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
76
|
Burn injury-induced mechanical allodynia is maintained by Rac1-regulated dendritic spine dysgenesis. Exp Neurol 2013; 248:509-19. [DOI: 10.1016/j.expneurol.2013.07.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/16/2013] [Accepted: 07/26/2013] [Indexed: 11/22/2022]
|
77
|
Beauparlant J, van den Brand R, Barraud Q, Friedli L, Musienko P, Dietz V, Courtine G. Undirected compensatory plasticity contributes to neuronal dysfunction after severe spinal cord injury. ACTA ACUST UNITED AC 2013; 136:3347-61. [PMID: 24080153 DOI: 10.1093/brain/awt204] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Severe spinal cord injury in humans leads to a progressive neuronal dysfunction in the chronic stage of the injury. This dysfunction is characterized by premature exhaustion of muscle activity during assisted locomotion, which is associated with the emergence of abnormal reflex responses. Here, we hypothesize that undirected compensatory plasticity within neural systems caudal to a severe spinal cord injury contributes to the development of neuronal dysfunction in the chronic stage of the injury. We evaluated alterations in functional, electrophysiological and neuromorphological properties of lumbosacral circuitries in adult rats with a staggered thoracic hemisection injury. In the chronic stage of the injury, rats exhibited significant neuronal dysfunction, which was characterized by co-activation of antagonistic muscles, exhaustion of locomotor muscle activity, and deterioration of electrochemically-enabled gait patterns. As observed in humans, neuronal dysfunction was associated with the emergence of abnormal, long-latency reflex responses in leg muscles. Analyses of circuit, fibre and synapse density in segments caudal to the spinal cord injury revealed an extensive, lamina-specific remodelling of neuronal networks in response to the interruption of supraspinal input. These plastic changes restored a near-normal level of synaptic input within denervated spinal segments in the chronic stage of injury. Syndromic analysis uncovered significant correlations between the development of neuronal dysfunction, emergence of abnormal reflexes, and anatomical remodelling of lumbosacral circuitries. Together, these results suggest that spinal neurons deprived of supraspinal input strive to re-establish their synaptic environment. However, this undirected compensatory plasticity forms aberrant neuronal circuits, which may engage inappropriate combinations of sensorimotor networks during gait execution.
Collapse
|
78
|
Bali KK, Venkataramani V, Satagopam VP, Gupta P, Schneider R, Kuner R. Transcriptional mechanisms underlying sensitization of peripheral sensory neurons by granulocyte-/granulocyte-macrophage colony stimulating factors. Mol Pain 2013; 9:48. [PMID: 24067145 PMCID: PMC3852053 DOI: 10.1186/1744-8069-9-48] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 08/25/2013] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Cancer-associated pain is a major cause of poor quality of life in cancer patients and is frequently resistant to conventional therapy. Recent studies indicate that some hematopoietic growth factors, namely granulocyte macrophage colony stimulating factor (GMCSF) and granulocyte colony stimulating factor (GCSF), are abundantly released in the tumor microenvironment and play a key role in regulating tumor-nerve interactions and tumor-associated pain by activating receptors on dorsal root ganglion (DRG) neurons. Moreover, these hematopoietic factors have been highly implicated in postsurgical pain, inflammatory pain and osteoarthritic pain. However, the molecular mechanisms via which G-/GMCSF bring about nociceptive sensitization and elicit pain are not known. RESULTS In order to elucidate G-/GMCSF mediated transcriptional changes in the sensory neurons, we performed a comprehensive, genome-wide analysis of changes in the transcriptome of DRG neurons brought about by exposure to GMCSF or GCSF. We present complete information on regulated genes and validated profiling analyses and report novel regulatory networks and interaction maps revealed by detailed bioinformatics analyses. Amongst these, we validate calpain 2, matrix metalloproteinase 9 (MMP9) and a RhoGTPase Rac1 as well as Tumor necrosis factor alpha (TNFα) as transcriptional targets of G-/GMCSF and demonstrate the importance of MMP9 and Rac1 in GMCSF-induced nociceptor sensitization. CONCLUSION With integrative approach of bioinformatics, in vivo pharmacology and behavioral analyses, our results not only indicate that transcriptional control by G-/GMCSF signaling regulates a variety of established pain modulators, but also uncover a large number of novel targets, paving the way for translational analyses in the context of pain disorders.
Collapse
Affiliation(s)
- Kiran Kumar Bali
- Institute for Pharmacology and Molecular Medicine Partnership Unit, Heidelberg University, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | - Varun Venkataramani
- Institute for Pharmacology and Molecular Medicine Partnership Unit, Heidelberg University, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | - Venkata P Satagopam
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, House of Biomedicine, 7 avenue des Hauts-Fourneaux, L-4362 Esch-sur-Alzette, Luxembourg
- European Molecular Biology Laboratory, Meyerhofstrasse. 1, D-69117 Heidelberg, Germany
| | - Pooja Gupta
- Institute for Pharmacology and Molecular Medicine Partnership Unit, Heidelberg University, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, House of Biomedicine, 7 avenue des Hauts-Fourneaux, L-4362 Esch-sur-Alzette, Luxembourg
- European Molecular Biology Laboratory, Meyerhofstrasse. 1, D-69117 Heidelberg, Germany
| | - Rohini Kuner
- Institute for Pharmacology and Molecular Medicine Partnership Unit, Heidelberg University, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| |
Collapse
|
79
|
Reactive oxygen species contribute to neuropathic pain and locomotor dysfunction via activation of CamKII in remote segments following spinal cord contusion injury in rats. Pain 2013; 154:1699-1708. [DOI: 10.1016/j.pain.2013.05.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/06/2013] [Accepted: 05/09/2013] [Indexed: 01/09/2023]
|
80
|
Spinal serum-inducible and glucocorticoid-inducible kinase 1 mediates neuropathic pain via kalirin and downstream PSD-95-dependent NR2B phosphorylation in rats. J Neurosci 2013; 33:5227-40. [PMID: 23516288 DOI: 10.1523/jneurosci.4452-12.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The coupling of the spinal postsynaptic density-95 (PSD-95) with the glutamatergic N-methyl-d-aspartate receptor NR2B subunit and the subsequent NR2B phosphorylation contribute to pain-related plasticity. Increasing evidence reveals that kalirin, a Rho-guanine nucleotide exchange factor, modulates PSD-95-NR2B-dependent neuroplasticity. Our laboratory recently demonstrated that serum-inducible and glucocorticoid-inducible kinase 1 (SGK1) participates in inflammation-associated pain hypersensitivity by modulating spinal glutamatergic neurotransmission. Because kalirin is one of the proteins in PSD that is highly phosphorylated by various kinases, we tested whether kalirin could be a downstream target of spinal SGK1 that participates in neuropathic pain development via regulation of the PSD-95-NR2B coupling-dependent phosphorylation of NR2B. We observed that spinal nerve ligation (SNL, L5) in male Sprague Dawley rats resulted in behavioral allodynia, which was associated with phosphorylated SGK1 (pSGK1), kalirin, and phosphorylated NR2B (pNR2B) expression and an increase in pSGK1-kalirin-PSD-95-pNR2B coprecipitation in the ipsilateral dorsal horn (L4-L5). SNL-enhanced kalirin immunofluorescence was coincident with pSGK1, PSD-95, and pNR2B immunoreactivity. Small-interfering RNA (siRNA) that targeted spinal kalirin mRNA expression (10 μg, 10 μl; i.t.) reduced SNL-induced allodynia, kalirin and pNR2B expression, as well as kalirin-PSD-95 and PSD-95-pNR2B coupling and costaining without affecting SGK1 phosphorylation. Daily administration of GSK-650394 (an SGK1 antagonist; 100 nm, 10 μl, i.t.) not only exhibited effects similar to the kalirin mRNA-targeting siRNA but also attenuated pSGK1-kalirin costaining and SGK1-kalirin coupling. We suggest that nerve injury could induce spinal SGK1 phosphorylation that subsequently interacts with and upregulates kalirin to participate in neuropathic pain development via PSD-95-NR2B coupling-dependent NR2B phosphorylation.
Collapse
|
81
|
Brain-derived neurotrophic factor from bone marrow-derived cells promotes post-injury repair of peripheral nerve. PLoS One 2012; 7:e44592. [PMID: 23028564 PMCID: PMC3446933 DOI: 10.1371/journal.pone.0044592] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 08/03/2012] [Indexed: 12/22/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) stimulates peripheral nerve regeneration. However, the origin of BNDF and its precise effect on nerve repair have not been clarified. In this study, we examined the role of BDNF from bone marrow-derived cells (BMDCs) in post-injury nerve repair. Control and heterozygote BDNF knockout mice (BDNF+/−) received a left sciatic nerve crush using a cerebral blood clip. Especially, for the evaluation of BDNF from BMDCs, studies with bone marrow transplantation (BMT) were performed before the injury. We evaluated nerve function using a rotarod test, sciatic function index (SFI), and motor nerve conduction velocity (MNCV) simultaneously with histological nerve analyses by immunohistochemistry before and after the nerve injury until 8 weeks. BDNF production was examined by immunohistochemistry and mRNA analyses. After the nerve crush, the controls showed severe nerve dysfunction evaluated at 1 week. However, nerve function was gradually restored and reached normal levels by 8 weeks. By immunohistochemistry, BDNF expression was very faint before injury, but was dramatically increased after injury at 1 week in the distal segment from the crush site. BDNF expression was mainly co-localized with CD45 in BMDCs, which was further confirmed by the appearance of GFP-positive cells in the BMT study. Variant analysis of BDNF mRNA also confirmed this finding. BDNF+/− mice showed a loss of function with delayed histological recovery and BDNF+/+→BDNF+/− BMT mice showed complete recovery both functionally and histologically. These results suggested that the attenuated recovery of the BDNF+/− mice was rescued by the transplantation of BMCs and that BDNF from BMDCs has an essential role in nerve repair.
Collapse
|
82
|
Abstract
Diabetic neuropathic pain imposes a huge burden on individuals and society, and represents a major public health problem. Despite aggressive efforts, diabetic neuropathic pain is generally refractory to available clinical treatments. A structure-function link between maladaptive dendritic spine plasticity and pain has been demonstrated previously in CNS and PNS injury models of neuropathic pain. Here, we reasoned that if dendritic spine remodeling contributes to diabetic neuropathic pain, then (1) the presence of malformed spines should coincide with the development of pain, and (2) disrupting maladaptive spine structure should reduce chronic pain. To determine whether dendritic spine remodeling contributes to neuropathic pain in streptozotocin (STZ)-induced diabetic rats, we analyzed dendritic spine morphology and electrophysiological and behavioral signs of neuropathic pain. Our results show changes in dendritic spine shape, distribution, and shape on wide-dynamic-range (WDR) neurons within lamina IV-V of the dorsal horn in diabetes. These diabetes-induced changes were accompanied by WDR neuron hyperexcitability and decreased pain thresholds at 4 weeks. Treatment with NSC23766 (N(6)-[2-[[4-(diethylamino)-1-methylbutyl]amino]-6-methyl-4-pyrimidinyl]-2-methyl-4,6-quinolinediamine trihydrochloride), a Rac1-specific inhibitor known to interfere with spine plasticity, decreased the presence of malformed spines in diabetes, attenuated neuronal hyperresponsiveness to peripheral stimuli, reduced spontaneous firing activity from WDR neurons, and improved nociceptive mechanical pain thresholds. At 1 week after STZ injection, animals with hyperglycemia with no evidence of pain had few or no changes in spine morphology. These results demonstrate that diabetes-induced maladaptive dendritic spine remodeling has a mechanistic role in neuropathic pain. Molecular pathways that control spine morphogenesis and plasticity may be promising future targets for treatment.
Collapse
|
83
|
Gwak YS, Hulsebosch CE. Neuronal hyperexcitability: a substrate for central neuropathic pain after spinal cord injury. Curr Pain Headache Rep 2012; 15:215-22. [PMID: 21387163 DOI: 10.1007/s11916-011-0186-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neuronal hyperexcitability produces enhanced pain transmission in the spinal dorsal horn after spinal cord injury (SCI). Spontaneous and evoked neuronal excitability normally are well controlled by neural circuits. However, SCI produces maladaptive synaptic circuits in the spinal dorsal horn that result in neuronal hyperexcitability. After SCI, activated primary afferent neurons produce enhanced release of glutamate, neuropeptides, adenosine triphosphate, and proinflammatory cytokines, which are known to be major components for pain transmission in the spinal dorsal horn. Enhanced neurochemical events contribute to neuronal hyperexcitability, and neuroanatomical changes also contribute to maladaptive synaptic circuits and neuronal hyperexcitability. These neurochemical and neuroanatomical changes produce enhanced cellular signaling cascades that ensure persistently enhanced pain transmission. This review describes altered neurochemical and neuroanatomical contributions on neuronal hyperexcitability in the spinal dorsal horn, which serve as substrates for central neuropathic pain after SCI.
Collapse
Affiliation(s)
- Young Seob Gwak
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA
| | | |
Collapse
|
84
|
The corneal pain system. Part I: the missing piece of the dry eye puzzle. Ocul Surf 2012; 10:2-14. [PMID: 22330055 DOI: 10.1016/j.jtos.2012.01.002] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 01/23/2023]
Abstract
The traditional model of dry eye disease based on tear deficiency has presented us with many unanswered questions. Recent studies support the notion that dry eye-like symptoms represent non-specific corneal pain and provide new insights into the mechanisms that sustain the integrity of the optical tear layer. Thus, this enigmatic disease can be viewed with a new perspective, which involves the dysfunctional corneal pain system as a central pathogenetic feature of a series of disorders collectively known today as dry eye.
Collapse
|
85
|
Wieseler J, Ellis A, Maier SF, Watkins LR, Falci S. Unilateral T13 and L1 dorsal root avulsion: methods for a novel model of central neuropathic pain. Methods Mol Biol 2012; 851:171-183. [PMID: 22351090 PMCID: PMC5652297 DOI: 10.1007/978-1-61779-561-9_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Central neuropathic pain is associated with many disease states including multiple sclerosis, stroke, and spinal cord injury, and is poorly managed. One type of central neuropathic pain that is particularly debilitating and challenging to treat is pain that occurs below the level of injury (below-level pain). The study of central neuropathic pain is commonly performed using animal models of stroke and spinal cord injury. Most of the spinal cord injury models currently being used were originally developed to model the gross physiological impact of clinical spinal cord injury. In contrast, the T13/L1 dorsal root avulsion model of spinal cord injury described here was developed specifically for the study of central pain, and as such, was developed to minimize confounding complications, such as paralysis, urinary tract infections, and autotomy. As such, this model induces robust and reliable hindpaw mechanical allodynia. Two versions of the model are described. The first is optimal for testing systemically administered pharmacological manipulations. The second was developed to accommodate intrathecal application of pharmacological manipulations. This model provides an additional means by which to investigate central pain states associated with spinal cord injury, including below-level pain. Finally, a brief discussion of at-level pain measurement is described as it has been suggested in the literature that the mechanisms underlying below- and at-level pain are different.
Collapse
Affiliation(s)
- Julie Wieseler
- Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA.
| | | | | | | | | |
Collapse
|
86
|
Tan AM, Chang YW, Zhao P, Hains BC, Waxman SG. Rac1-regulated dendritic spine remodeling contributes to neuropathic pain after peripheral nerve injury. Exp Neurol 2011; 232:222-33. [DOI: 10.1016/j.expneurol.2011.08.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 08/11/2011] [Accepted: 08/18/2011] [Indexed: 10/17/2022]
|
87
|
Melemedjian OK, Price TJ. Dendritic spine plasticity as an underlying mechanism of neuropathic pain: commentary on Tan et al. Exp Neurol 2011; 233:740-4. [PMID: 22119623 DOI: 10.1016/j.expneurol.2011.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/07/2011] [Accepted: 11/10/2011] [Indexed: 01/01/2023]
|
88
|
Tan AM, Waxman SG. Spinal cord injury, dendritic spine remodeling, and spinal memory mechanisms. Exp Neurol 2011; 235:142-51. [PMID: 21925174 DOI: 10.1016/j.expneurol.2011.08.026] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 08/17/2011] [Accepted: 08/25/2011] [Indexed: 01/27/2023]
Abstract
Spinal cord injury (SCI) often results in the development of neuropathic pain, which can persist for months and years after injury. Although many aberrant changes to sensory processing contribute to the development of chronic pain, emerging evidence demonstrates that mechanisms similar to those underlying classical learning and memory can contribute to central sensitization, a phenomenon of amplified responsiveness to stimuli in nociceptive dorsal horn neurons. Notably, dendritic spines have emerged as major players in learning and memory, providing a structural substrate for how the nervous system modifies connections to form and store information. Until now, most information regarding dendritic spines has been obtained from studies in the brain. Recent experimental data in the spinal cord, however, demonstrate that Rac1-regulated dendritic spine remodeling occurs on second-order wide dynamic range neurons and accompanies neuropathic pain after SCI. Thus, SCI-induced synaptic potentiation engages a putative spinal memory mechanism. A compelling, novel possibility for pain research is that a synaptic model of long-term memory storage could explain the persistent nature of neuropathic pain. Such a conceptual bridge between pain and memory could guide the development of more effective strategies for treatment of chronic pain after injury to the nervous system.
Collapse
Affiliation(s)
- Andrew M Tan
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | |
Collapse
|
89
|
Wieseler J, Ellis AL, McFadden A, Brown K, Starnes C, Maier SF, Watkins LR, Falci S. Below level central pain induced by discrete dorsal spinal cord injury. J Neurotrauma 2011; 27:1697-707. [PMID: 20649467 DOI: 10.1089/neu.2010.1311] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Central neuropathic pain occurs with multiple sclerosis, stroke, and spinal cord injury (SCI). Models of SCI are commonly used to study central neuropathic pain and are excellent at modeling gross physiological changes. Our goal was to develop a rat model of central neuropathic pain by traumatizing a discrete region of the dorsal spinal cord, thereby avoiding issues including paralysis, urinary tract infection, and autotomy. To this end, dorsal root avulsion was pursued. The model was developed by first determining the number of avulsed dorsal roots sufficient to induce below-level hindpaw mechanical allodynia. This was optimally achieved by unilateral T13 and L1 avulsion, which resulted in tissue damage confined to Lissauer's tract, dorsal horn, and dorsal columns, at the site of avulsion, with no gross physical changes at other spinal levels. Behavior following avulsion was compared to that following rhizotomy of the T13 and L1 dorsal roots, a commonly used model of neuropathic pain. Avulsion induced below-level allodynia that was more robust and enduring than that seen after rhizotomy. This, plus the lack of direct spinal cord damage associated with rhizotomy, suggests that avulsion is not synonymous with rhizotomy, and that avulsion (but not rhizotomy) is a model of central neuropathic pain. The new model described here is the first to use discrete dorsal horn damage by dorsal root avulsion to create below-level bilateral central neuropathic pain.
Collapse
Affiliation(s)
- Julie Wieseler
- Department of Psychology and Neuroscience and the Center for Neurosciences, Campus Box 345, University of Colorado at Boulder, Boulder, Colorado 80309-0345, USA.
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Neuropathic pain- and glial derived neurotrophic factor-associated regulation of cadherins in spinal circuits of the dorsal horn. Pain 2011; 152:924-935. [DOI: 10.1016/j.pain.2011.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 11/24/2010] [Accepted: 01/10/2011] [Indexed: 12/29/2022]
|
91
|
Berger JV, Knaepen L, Janssen SPM, Jaken RJP, Marcus MAE, Joosten EAJ, Deumens R. Cellular and molecular insights into neuropathy-induced pain hypersensitivity for mechanism-based treatment approaches. ACTA ACUST UNITED AC 2011; 67:282-310. [PMID: 21440003 DOI: 10.1016/j.brainresrev.2011.03.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 02/16/2011] [Accepted: 03/18/2011] [Indexed: 12/15/2022]
Abstract
Neuropathic pain is currently being treated by a range of therapeutic interventions that above all act to lower neuronal activity in the somatosensory system (e.g. using local anesthetics, calcium channel blockers, and opioids). The present review highlights novel and often still largely experimental treatment approaches based on insights into pathological mechanisms, which impact on the spinal nociceptive network, thereby opening the 'gate' to higher brain centers involved in the perception of pain. Cellular and molecular mechanisms such as ectopia, sensitization of nociceptors, phenotypic switching, structural plasticity, disinhibition, and neuroinflammation are discussed in relation to their involvement in pain hypersensitivity following either peripheral neuropathies or spinal cord injury. A mechanism-based treatment approach may prove to be successful in effective treatment of neuropathic pain, but requires more detailed insights into the persistence of cellular and molecular pain mechanisms which renders neuropathic pain unremitting. Subsequently, identification of the therapeutic window-of-opportunities for each specific intervention in the particular peripheral and/or central neuropathy is essential for successful clinical trials. Most of the cellular and molecular pain mechanisms described in the present review suggest pharmacological interference for neuropathic pain management. However, also more invasive treatment approaches belong to current and/or future options such as neuromodulatory interventions (including spinal cord stimulation) and cell or gene therapies, respectively.
Collapse
Affiliation(s)
- Julie V Berger
- Department of Anesthesiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
92
|
The animal model of spinal cord injury as an experimental pain model. J Biomed Biotechnol 2011; 2011:939023. [PMID: 21436995 PMCID: PMC3062973 DOI: 10.1155/2011/939023] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 12/19/2010] [Indexed: 12/25/2022] Open
Abstract
Pain, which remains largely unsolved, is one of the most crucial problems for spinal cord injury patients. Due to sensory problems, as well as motor dysfunctions, spinal cord injury research has proven to be complex and difficult. Furthermore, many types of pain are associated with spinal cord injury, such as neuropathic, visceral, and musculoskeletal pain. Many animal models of spinal cord injury exist to emulate clinical situations, which could help to determine common mechanisms of pathology. However, results can be easily misunderstood and falsely interpreted. Therefore, it is important to fully understand the symptoms of human spinal cord injury, as well as the various spinal cord injury models and the possible pathologies. The present paper summarizes results from animal models of spinal cord injury, as well as the most effective use of these models.
Collapse
|
93
|
Nesic O, Sundberg LM, Herrera JJ, Mokkapati VUL, Lee J, Narayana PA. Vascular endothelial growth factor and spinal cord injury pain. J Neurotrauma 2011; 27:1793-803. [PMID: 20698758 DOI: 10.1089/neu.2010.1351] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF)-A mRNA was previously identified as one of the significantly upregulated transcripts in spinal cord injured tissue from adult rats that developed allodynia. To characterize the role of VEGF-A in the development of pain in spinal cord injury (SCI), we analyzed mechanical allodynia in SCI rats that were treated with either vehicle, VEGF-A isoform 165 (VEGF(165)), or neutralizing VEGF(165)-specific antibody. We have observed that exogenous administration of VEGF(165) increased both the number of SCI rats that develop persistent mechanical allodynia, and the level of hypersensitivity to mechanical stimuli. Our analysis identified excessive and aberrant growth of myelinated axons in dorsal horns and dorsal columns of chronically injured spinal cords as possible mechanisms for both SCI pain and VEGF(165)-induced amplification of SCI pain, suggesting that elevated endogenous VEGF(165) may have a role in the development of allodynia after SCI. However, the neutralizing VEGF(165) antibody showed no effect on allodynia or axonal sprouting after SCI. It is possible that another endogenous VEGF isoform activates the same signaling pathway as the exogenously-administered 165 isoform and contributes to SCI pain. Our transcriptional analysis revealed that endogenous VEGF(188) is likely to be the isoform involved in the development of allodynia after SCI. To the best of our knowledge, this is the first study to suggest a possible link between VEGF, nonspecific sprouting of myelinated axons, and mechanical allodynia following SCI.
Collapse
Affiliation(s)
- Olivera Nesic
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555-1072, USA.
| | | | | | | | | | | |
Collapse
|
94
|
Gwak YS, Hulsebosch CE. GABA and central neuropathic pain following spinal cord injury. Neuropharmacology 2011; 60:799-808. [PMID: 21216257 DOI: 10.1016/j.neuropharm.2010.12.030] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 12/02/2010] [Accepted: 12/22/2010] [Indexed: 12/31/2022]
Abstract
Spinal cord injury induces maladaptive synaptic transmission in the somatosensory system that results in chronic central neuropathic pain. Recent literature suggests that glial-neuronal interactions are important modulators in synaptic transmission following spinal cord injury. Neuronal hyperexcitability is one of the predominant phenomenon caused by maladaptive synaptic transmission via altered glial-neuronal interactions after spinal cord injury. In the somatosensory system, spinal inhibitory neurons counter balance the enhanced synaptic transmission from peripheral input. For a decade, the literature suggests that hypofunction of GABAergic inhibitory tone is an important factor in the enhanced synaptic transmission that often results in neuronal hyperexcitability in dorsal horn neurons following spinal cord injury. Neurons and glial cells synergistically control intracellular chloride ion gradients via modulation of chloride transporters, extracellular glutamate and GABA concentrations via uptake mechanisms. Thus, the intracellular "GABA-glutamate-glutamine cycle" is maintained for normal physiological homeostasis. However, hyperexcitable neurons and glial activation after spinal cord injury disrupts the balance of chloride ions, glutamate and GABA distribution in the spinal dorsal horn and results in chronic neuropathic pain. In this review, we address spinal cord injury induced mechanisms in hypofunction of GABAergic tone that results in chronic central neuropathic pain. This article is part of a Special Issue entitled 'Synaptic Plasticity & Interneurons'.
Collapse
Affiliation(s)
- Young S Gwak
- Department of Neuroscience & Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1043, USA.
| | | |
Collapse
|
95
|
Abstract
Chronic pain is a major challenge to clinical practice and basic science. The peripheral and central neural networks that mediate nociception show extensive plasticity in pathological disease states. Disease-induced plasticity can occur at both structural and functional levels and is manifest as changes in individual molecules, synapses, cellular function and network activity. Recent work has yielded a better understanding of communication within the neural matrix of physiological pain and has also brought important advances in concepts of injury-induced hyperalgesia and tactile allodynia and how these might contribute to the complex, multidimensional state of chronic pain. This review focuses on the molecular determinants of network plasticity in the central nervous system (CNS) and discusses their relevance to the development of new therapeutic approaches.
Collapse
|
96
|
Validity of acute and chronic tactile sensory testing after spinal cord injury in rats. Exp Neurol 2010; 225:366-76. [PMID: 20643128 DOI: 10.1016/j.expneurol.2010.07.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 07/10/2010] [Accepted: 07/13/2010] [Indexed: 11/22/2022]
Abstract
Spinal cord injury (SCI) impairs sensory systems causing allodynia. Measuring the development of allodynia in rodent models of SCI is challenging due to spinal shock and marked motor impairments. Assessment of SCI-induced allodynia is not standardized across labs, making interpretation of results difficult. Therefore, we validated sensory threshold assessment after SCI and developed a novel assessment of allodynia prior to motor recovery in a rat SCI model. One hundred fifty-six Sprague-Dawley rats received T8 laminectomy or mild to moderate SCI using the OSU SCI device (0.3 mm to 1.3 mm cord displacement). To determine tactile thresholds, von Frey hairs (VFH) were applied in Up-Down or ascending order to the dorsal or plantar hindpaw. The most efficient and valid procedures that maintain high sensitivity and specificity were identified. Ten Up-Down VFH applications yielded stable thresholds; reducing the risk of threshold decay and unnecessary exposure to painful stimuli. Importantly, distraction of SCI-rats with food revealed differential decay of thresholds than when distraction is not provided. The new test uses dorsal VFH stimulation and is independent of trunk or hindlimb control. Acute dorsal VFH thresholds collected before recovery of hindlimb weight support accurately predicted plantar VFH thresholds measured at late timepoints (chi(2)=8.479; p<0.05). Thus, standardized testing early after SCI using the dorsal VFH test or later using 10 stimuli in the Up-Down test produces valid measures of tactile sensation across many SCI severities. Early detection of allodynia in experimental SCI will allow identification of mechanisms responsible for pain development and determine targets for therapeutic interventions.
Collapse
|
97
|
|
98
|
At the sharp end of spines: anesthetic effects on synaptic remodeling in the developing brain. Anesthesiology 2010; 112:521-3. [PMID: 20124984 DOI: 10.1097/aln.0b013e3181cd795d] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
99
|
BDNF-hypersecreting human mesenchymal stem cells promote functional recovery, axonal sprouting, and protection of corticospinal neurons after spinal cord injury. J Neurosci 2010; 29:14932-41. [PMID: 19940189 DOI: 10.1523/jneurosci.2769-09.2009] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) derived from bone marrow has been shown to improve functional outcome in spinal cord injury (SCI). We transplanted MSCs derived from human bone marrow (hMSCs) to study their potential therapeutic effect in SCI in the rat. In addition to hMSCs, we used gene-modified hMSCs to secrete brain-derived neurotrophic factor (BDNF-hMSCs). After a dorsal transection lesion was induced at T9, cells were microinjected on each side of the transection site. Fluorogold (FG) was injected into the epicenter of the lesion cavity to identify transected corticospinal tract (CST) neurons. At 5 weeks after transplantation, the animals were perfused. Locomotor recovery improvement was observed for the BDNF-hMSC group, but not in the hMSC group. Structurally there was increased sprouting of injured corticospinal tract and serotonergic projections after hMSC and BDNF-hMSC transplantation. Moreover, an increased number of serotonergic fibers was observed in spinal gray matter including the ventral horn at and below the level of the lesion, indicating increased innervation in the terminal regions of a descending projection important for locomotion. Stereological quantification was performed on the brains to determine neuronal density in primary motor (M1) cortex. The number of FG backfilled cells demonstrated an increased cell survival of CST neurons in M1 cortex in both the hMSC and BDNF-hMSC groups at 5 weeks, but the increase for the BDNF-hMSC group was greater. These results indicate that transplantation of hMSCs hypersecreting BDNF results in structural changes in brain and spinal cord, which are associated with improved functional outcome in acute SCI.
Collapse
|
100
|
Tan AM, Choi JS, Waxman SG, Hains BC. Dendritic Spine Remodeling After Spinal Cord Injury Alters Neuronal Signal Processing. J Neurophysiol 2009; 102:2396-409. [DOI: 10.1152/jn.00095.2009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Central sensitization, a prolonged hyperexcitability of dorsal horn nociceptive neurons, is a major contributor to abnormal pain processing after spinal cord injury (SCI). Dendritic spines are micron-sized dendrite protrusions that can regulate the efficacy of synaptic transmission. Here we used a computational approach to study whether changes in dendritic spine shape, density, and distribution can individually, or in combination, adversely modify the input–output function of a postsynaptic neuron to create a hyperexcitable neuronal state. The results demonstrate that a conversion from thin-shaped to more mature, mushroom-shaped spine structures results in enhanced synaptic transmission and fidelity, improved frequency-following ability, and reduced inhibitory gating effectiveness. Increasing the density and redistributing spines toward the soma results in a greater probability of action potential activation. Our results demonstrate that changes in dendritic spine morphology, documented in previous studies on spinal cord injury, contribute to the generation of pain following SCI.
Collapse
Affiliation(s)
- Andrew M. Tan
- Departments of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jin-Sung Choi
- Departments of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Departments of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Bryan C. Hains
- Departments of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|