1
|
Morioka K, Tazoe T, Huie JR, Hayakawa K, Okazaki R, Guandique CF, Almeida CA, Haefeli J, Hamanoue M, Endoh T, Tanaka S, Bresnahan JC, Beattie MS, Ogata T, Ferguson AR. Disuse plasticity limits spinal cord injury recovery. iScience 2025; 28:112180. [PMID: 40224010 PMCID: PMC11987634 DOI: 10.1016/j.isci.2025.112180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
Use-dependent plasticity after spinal cord injury (SCI) enhances neuromotor function, however, the optimal timing to initiate rehabilitation remains controversial. To test impacts of early disuse, we established a rodent model of transient hindlimb suspension in acute phase SCI. Early disuse in the first 2-week after SCI undermined recovery on open-field locomotion, kinematics, and swim tests even after 6-week of normal gravity reloading. Early disuse produced chronic spinal circuit hyper-excitability in H-reflex and interlimb reflex tests. Quantitative synaptoneurosome analysis of lumboventral spinal cords revealed shifts in AMPA receptor (AMPAR) subunit GluA1 localization and serine 881 phosphorylation, reflecting enduring synaptic memories of early disuse stored in the spinal cord. Automated confocal analysis of motoneurons revealed persistent shifts toward GluA2-lacking, calcium-permeable AMPARs in disuse subjects. Unsupervised machine learning associated multidimensional synaptic changes with persistent recovery deficits in SCI. The results argue for early aggressive rehabilitation to prevent disuse plasticity that limits SCI recovery.
Collapse
Affiliation(s)
- Kazuhito Morioka
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute (OTI), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Toshiki Tazoe
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Neural Prosthesis Project, Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - J. Russell Huie
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Kentaro Hayakawa
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Orthopaedic and Spine Surgery, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Rentaro Okazaki
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Orthopaedic Surgery, Saitama Red Cross Hospital, Saitama, Japan
| | - Cristian F. Guandique
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Carlos A. Almeida
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jenny Haefeli
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Makoto Hamanoue
- Department of Physiology, Advanced Medical Research Center, Toho University School of Medicine, Tokyo, Japan
| | - Takashi Endoh
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Faculty of Development and Education, Uekusa Gakuen University, Chiba, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jacqueline C. Bresnahan
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Michael S. Beattie
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Toru Ogata
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Rehabilitation Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Adam R. Ferguson
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
- San Francisco Veterans Affairs Healthcare System (SFVAHCS), San Francisco, CA, USA
| |
Collapse
|
2
|
Shamsnia HS, Peyrovinasab A, Amirlou D, Sirouskabiri S, Rostamian F, Basiri N, Shalmani LM, Hashemi M, Hushmandi K, Abdolghaffari AH. BDNF-TrkB Signaling Pathway in Spinal Cord Injury: Insights and Implications. Mol Neurobiol 2025; 62:1904-1944. [PMID: 39046702 DOI: 10.1007/s12035-024-04381-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024]
Abstract
Spinal cord injury (SCI) is a neurodegenerative disorder that has critical impact on patient's life expectance and life span, and this disorder also leads to negative socioeconomic features. SCI is defined as a firm collision to the spinal cord which leads to the fracture and the dislocation of vertebrae. The current available treatment is surgery. However, it cannot fully treat SCI, and many consequences remain after the surgery. Accordingly, finding new therapeutics is critical. BDNF-TrkB signaling is a vital signaling in neuronal differentiation, survival, overgrowth, synaptic plasticity, etc. Hence, many studies evaluate its impact on various neurodegenerative disorders. There are several studies evaluating this signaling in SCI, and they show promising outcomes. It was shown that various exercises, chemical interventions, etc. had significant positive impact on SCI by affecting BDNF-TrkB signaling pathway. This study aims to accumulate and evaluate these data and inspect whether this signaling is effective or not.
Collapse
Affiliation(s)
- Hedieh Sadat Shamsnia
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amirreza Peyrovinasab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Dorsa Amirlou
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shirin Sirouskabiri
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Rostamian
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nasim Basiri
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Leila Mohaghegh Shalmani
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | | | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St, P. O. Box: 19419-33111, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
3
|
Sanati M, Manavi MA, Noruzi M, Behmadi H, Akbari T, Jalali S, Sharifzadeh M, Khoobi M. Carbohydrates and neurotrophic factors: A promising partnership for spinal cord injury rehabilitation. BIOMATERIALS ADVANCES 2025; 166:214054. [PMID: 39332344 DOI: 10.1016/j.bioadv.2024.214054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
Spinal cord injury (SCI) leaves a temporary or enduring motor, sensory, and autonomic function loss, significantly impacting the patient's quality of life. Given their biocompatibility, bioactivity, and tunable attributes, three-dimensional scaffolds frequently employ carbohydrates to facilitate spinal cord regeneration. These scaffolds have also been engineered to be novel local delivery platforms that present distinct advantages in the targeted transportation of drug candidates to the damaged spinal cord, ensuring the right dosage and duration of administration. Neurotrophic factors have emerged as promising therapeutic candidates, preserved neuron survival and encouraged severed axons repair, although their local and continuous delivery is believed to produce considerable spinal cord rehabilitation. This study aims to discuss breakthroughs in scaffold engineering, exploiting carbohydrates as an essential part of their structure, and highlight their impact on spinal cord regeneration and sustained neurotrophic factors delivery to treat SCI.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Amin Manavi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Noruzi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
| | - Homayoon Behmadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Sara Jalali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, 1417614411 Tehran, Iran.
| |
Collapse
|
4
|
Jang K, Garraway SM. TrkB Agonist (7,8-DHF)-Induced Responses in Dorsal Root Ganglia Neurons Are Decreased after Spinal Cord Injury: Implication for Peripheral Pain Mechanisms. eNeuro 2025; 12:ENEURO.0219-24.2024. [PMID: 39753357 PMCID: PMC11728855 DOI: 10.1523/eneuro.0219-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 11/08/2024] [Accepted: 12/04/2024] [Indexed: 01/15/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB) are known to contribute to both protective and pronociceptive processes. However, their contribution to neuropathic pain after spinal cord injury (SCI) needs further investigation. In a recent study utilizing TrkBF616A mice, it was shown that systemic pharmacogenetic inhibition of TrkB signaling with 1NM-PP1 (1NMP) immediately after SCI delayed the onset of pain hypersensitivity, implicating maladaptive TrkB signaling in pain after SCI. To examine potential neural mechanisms underlying the behavioral outcome, patch-clamp recording was performed in small-diameter dissociated thoracic (T) dorsal root ganglia (DRG) neurons to evaluate TrkB signaling in uninjured mice and after T10 contusion SCI. Bath-applied 7,8-dihydroxyflavone (7,8-DHF), a selective TrkB agonist, induced a robust inward current in neurons from uninjured mice, which was attenuated by 1NMP treatment. SCI also decreased 7,8-DHF-induced current while increasing the latency to its peak amplitude. Western blot revealed a concomitant decrease in TrkB expression in DRGs adjacent to the spinal lesion. Analyses of cellular and membrane properties showed that SCI increased neuronal excitability, evident by an increase in resting membrane potential and the number of spiking neurons. However, SCI did not increase spontaneous firing in DRG neurons. These results suggest that SCI induced changes in TrkB activation in DRG neurons even though these alterations are likely not contributing to pain hypersensitivity by nociceptor hyperexcitability. Overall, this reveals complex interactions involving TrkB signaling and provides an opportunity to investigate other, presumably peripheral, mechanisms by which TrkB contributes to pain hypersensitivity after SCI.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, Georgia 30322
| | - Sandra M Garraway
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
5
|
Li J, Kang W, Wang X, Pan F. Progress in treatment of pathological neuropathic pain after spinal cord injury. Front Neurol 2024; 15:1430288. [PMID: 39606699 PMCID: PMC11600731 DOI: 10.3389/fneur.2024.1430288] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Pathological neuropathic pain is a common complication following spinal cord injury. Due to its high incidence, prolonged duration, tenacity, and limited therapeutic efficacy, it has garnered increasing attention from both basic researchers and clinicians. The pathogenesis of neuropathic pain after spinal cord injury is multifaceted, involving factors such as structural and functional alterations of the central nervous system, pain signal transduction, and inflammatory effects, posing significant challenges to clinical management. Currently, drugs commonly employed in treating spinal cord injury induced neuropathic pain include analgesics, anticonvulsants, antidepressants, and antiepileptics. However, a subset of patients often experiences suboptimal therapeutic responses or severe adverse reactions. Therefore, emerging treatments are emphasizing a combination of pharmacological and non-pharmacological approaches to enhance neuropathic pain management. We provide a comprehensive review of past literature, which aims to aim both the mechanisms and clinical interventions for pathological neuropathic pain following spinal cord injury, offering novel insights for basic science research and clinical practice in spinal cord injury treatment.
Collapse
Affiliation(s)
- Jian Li
- Department of Orthopedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Xuanwu Jinan Hospital, Jinan, China
| | - Wenqing Kang
- Department of Neurology, Yidu Central Hospital of Weifang, Weifang, Shandong, China
| | - Xi Wang
- Department of Orthopedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Fang Pan
- Department of rehabilitation, Shandong Rehabilitation Hospital, Jinan, Shandong, China
| |
Collapse
|
6
|
Grau JW, Hudson KE, Johnston DT, Partipilo SR. Updating perspectives on spinal cord function: motor coordination, timing, relational processing, and memory below the brain. Front Syst Neurosci 2024; 18:1184597. [PMID: 38444825 PMCID: PMC10912355 DOI: 10.3389/fnsys.2024.1184597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Those studying neural systems within the brain have historically assumed that lower-level processes in the spinal cord act in a mechanical manner, to relay afferent signals and execute motor commands. From this view, abstracting temporal and environmental relations is the province of the brain. Here we review work conducted over the last 50 years that challenges this perspective, demonstrating that mechanisms within the spinal cord can organize coordinated behavior (stepping), induce a lasting change in how pain (nociceptive) signals are processed, abstract stimulus-stimulus (Pavlovian) and response-outcome (instrumental) relations, and infer whether stimuli occur in a random or regular manner. The mechanisms that underlie these processes depend upon signal pathways (e.g., NMDA receptor mediated plasticity) analogous to those implicated in brain-dependent learning and memory. New data show that spinal cord injury (SCI) can enable plasticity within the spinal cord by reducing the inhibitory effect of GABA. It is suggested that the signals relayed to the brain may contain information about environmental relations and that spinal cord systems can coordinate action in response to descending signals from the brain. We further suggest that the study of stimulus processing, learning, memory, and cognitive-like processing in the spinal cord can inform our views of brain function, providing an attractive model system. Most importantly, the work has revealed new avenues of treatment for those that have suffered a SCI.
Collapse
Affiliation(s)
- James W. Grau
- Lab of Dr. James Grau, Department of Psychological and Brain Sciences, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | | | | | | |
Collapse
|
7
|
Noble DJ, Dongmo R, Parvin S, Martin KK, Garraway SM. C-low threshold mechanoreceptor activation becomes sufficient to trigger affective pain in spinal cord-injured mice in association with increased respiratory rates. Front Integr Neurosci 2022; 16:1081172. [PMID: 36619238 PMCID: PMC9811591 DOI: 10.3389/fnint.2022.1081172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
The mechanisms of neuropathic pain after spinal cord injury (SCI) are not fully understood. In addition to the plasticity that occurs within the injured spinal cord, peripheral processes, such as hyperactivity of primary nociceptors, are critical to the expression of pain after SCI. In adult rats, truncal stimulation within the tuning range of C-low threshold mechanoreceptors (C-LTMRs) contributes to pain hypersensitivity and elevates respiratory rates (RRs) after SCI. This suggests that C-LTMRs, which normally encode pleasant, affiliative touch, undergo plasticity to transmit pain sensation following injury. Because tyrosine hydroxylase (TH) expression is a specific marker of C-LTMRs, in the periphery, here we used TH-Cre adult mice to investigate more specifically the involvement of C-LTMRs in at-level pain after thoracic contusion SCI. Using a modified light-dark chamber conditioned place aversion (CPA) paradigm, we assessed chamber preferences and transitions between chambers at baseline, and in response to mechanical and optogenetic stimulation of C-LTMRs. In parallel, at baseline and select post-surgical timepoints, mice underwent non-contact RR recordings and von Frey assessment of mechanical hypersensitivity. The results showed that SCI mice avoided the chamber associated with C-LTMR stimulation, an effect that was more pronounced with optical stimulation. They also displayed elevated RRs at rest and during CPA training sessions. Importantly, these changes were restricted to chronic post-surgery timepoints, when hindpaw mechanical hypersensitivity was also evident. Together, these results suggest that C-LTMR afferent plasticity, coexisting with potentially facilitatory changes in breathing, drives at-level affective pain following SCI in adult mice.
Collapse
|
8
|
Martin KK, Noble DJ, Parvin S, Jang K, Garraway SM. Pharmacogenetic inhibition of TrkB signaling in adult mice attenuates mechanical hypersensitivity and improves locomotor function after spinal cord injury. Front Cell Neurosci 2022; 16:987236. [PMID: 36226073 PMCID: PMC9548551 DOI: 10.3389/fncel.2022.987236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) signals through tropomyosin receptor kinase B (TrkB), to exert various types of plasticity. The exact involvement of BDNF and TrkB in neuropathic pain states after spinal cord injury (SCI) remains unresolved. This study utilized transgenic TrkBF616 mice to examine the effect of pharmacogenetic inhibition of TrkB signaling, induced by treatment with 1NM-PP1 (1NMP) in drinking water for 5 days, on formalin-induced inflammatory pain, pain hypersensitivity, and locomotor dysfunction after thoracic spinal contusion. We also examined TrkB, ERK1/2, and pERK1/2 expression in the lumbar spinal cord and trunk skin. The results showed that formalin-induced pain responses were robustly attenuated in 1NMP-treated mice. Weekly assessment of tactile sensitivity with the von Frey test showed that treatment with 1NMP immediately after SCI blocked the development of mechanical hypersensitivity up to 4 weeks post-SCI. Contrastingly, when treatment started 2 weeks after SCI, 1NMP reversibly and partially attenuated hind-paw hypersensitivity. Locomotor scores were significantly improved in the early-treated 1NMP mice compared to late-treated or vehicle-treated SCI mice. 1NMP treatment attenuated SCI-induced increases in TrkB and pERK1/2 levels in the lumbar cord but failed to exert similar effects in the trunk skin. These results suggest that early onset TrkB signaling after SCI contributes to maladaptive plasticity that leads to spinal pain hypersensitivity and impaired locomotor function.
Collapse
Affiliation(s)
| | | | | | | | - Sandra M. Garraway
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
9
|
Parvin S, Williams CR, Jarrett SA, Garraway SM. Spinal Cord Injury Increases Pro-inflammatory Cytokine Expression in Kidney at Acute and Sub-chronic Stages. Inflammation 2021; 44:2346-2361. [PMID: 34417952 PMCID: PMC8616867 DOI: 10.1007/s10753-021-01507-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/21/2021] [Indexed: 11/26/2022]
Abstract
Accumulating evidence supports that spinal cord injury (SCI) produces robust inflammatory plasticity. We previously showed that the pro-inflammatory cytokine tumor necrosis factor (TNF)α is increased in the spinal cord after SCI. SCI also induces a systemic inflammatory response that can impact peripheral organ functions. The kidney plays an important role in maintaining cardiovascular health. However, SCI-induced inflammatory response in the kidney and the subsequent effect on renal function have not been well characterized. This study investigated the impact of high and low thoracic (T) SCI on C-fos, TNFα, interleukin (IL)-1β, and IL-6 expression in the kidney at acute and sub-chronic timepoints. Adult C57BL/6 mice received a moderate contusion SCI or sham procedures at T4 or T10. Uninjured mice served as naïve controls. mRNA levels of the proinflammatory cytokines IL-1β, IL-6, TNFα, and C-fos, and TNFα and C-fos protein expression were assessed in the kidney and spinal cord 1 day and 14 days post-injury. The mRNA levels of all targets were robustly increased in the kidney and spinal cord, 1 day after both injuries. Whereas IL-6 and TNFα remained elevated in the spinal cord at 14 days after SCI, C-fos, IL-6, and TNFα levels were sustained in the kidney only after T10 SCI. TNFα protein was significantly upregulated in the kidney 1 day after both T4 and T10 SCI. Overall, these results clearly demonstrate that SCI induces robust systemic inflammation that extends to the kidney. Hence, the presence of renal inflammation can substantially impact renal pathophysiology and function after SCI.
Collapse
Affiliation(s)
- Shangrila Parvin
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
| | - Clintoria R. Williams
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
- Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH USA
| | - Simone A. Jarrett
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
| | - Sandra M. Garraway
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
- Department of Physiology, Emory University School of Medicine, 615 Michael Street, Suite 605G, Atlanta, GA 30322 USA
| |
Collapse
|
10
|
Strain MM, Johnston DT, Baine RE, Reynolds JA, Huang YJ, Henwood MK, Fauss GN, Davis JA, Miranda RC, West CR, Grau JW. Hemorrhage and Locomotor Deficits Induced by Pain Input after Spinal Cord Injury Are Partially Mediated by Changes in Hemodynamics. J Neurotrauma 2021; 38:3406-3430. [PMID: 34652956 DOI: 10.1089/neu.2021.0219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nociceptive input diminishes recovery and increases lesion area after a spinal cord injury (SCI). Recent work has linked these effects to the expansion of hemorrhage at the site of injury. The current article examines whether these adverse effects are linked to a pain-induced rise in blood pressure (BP) and/or flow. Male rats with a low-thoracic SCI were treated with noxious input (electrical stimulation [shock] or capsaicin) soon after injury. Locomotor recovery and BP were assessed throughout. Tissues were collected 3 h, 24 h, or 21 days later. Both electrical stimulation and capsaicin undermined locomotor function and increased the area of hemorrhage. Changes in BP/flow varied depending on type of noxious input, with only shock producing changes in BP. Providing behavioral control over the termination of noxious stimulation attenuated the rise in BP and hemorrhage. Pretreatment with the α-1 adrenergic receptor inverse agonist, prazosin, reduced the stimulation-induced rise in BP and hemorrhage. Prazosin also attenuated the adverse effect that noxious stimulation has on long-term recovery. Administration of the adrenergic agonist, norepinephrine 1 day after injury induced an increase in BP and disrupted locomotor function, but had little effect on hemorrhage. Further, inducing a rise in BP/flow using norepinephrine undermined long-term recovery and increased tissue loss. Mediational analyses suggest that the pain-induced rise in blood flow may foster hemorrhage after SCI. Increased BP appears to act through an independent process to adversely affect locomotor performance, tissue sparing, and long-term recovery.
Collapse
Affiliation(s)
- Misty M Strain
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - David T Johnston
- Cellular and Behavioral Neuroscience, Department of Psychology, and College of Medicine, Texas A&M University, College Station, Texas, USA
| | - Rachel E Baine
- Cellular and Behavioral Neuroscience, Department of Psychology, and College of Medicine, Texas A&M University, College Station, Texas, USA
| | - Joshua A Reynolds
- Cellular and Behavioral Neuroscience, Department of Psychology, and College of Medicine, Texas A&M University, College Station, Texas, USA
| | | | - Melissa K Henwood
- Cellular and Behavioral Neuroscience, Department of Psychology, and College of Medicine, Texas A&M University, College Station, Texas, USA
| | - Gizelle N Fauss
- Cellular and Behavioral Neuroscience, Department of Psychology, and College of Medicine, Texas A&M University, College Station, Texas, USA
| | - Jacob A Davis
- Cellular and Behavioral Neuroscience, Department of Psychology, and College of Medicine, Texas A&M University, College Station, Texas, USA
| | - Rajesh C Miranda
- Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, College Station, Texas, USA
| | - Christopher R West
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, and College of Medicine, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
11
|
Jeffrey-Gauthier R, Bouyer J, Piché M, Côté MP, Leblond H. Locomotor deficits induced by lumbar muscle inflammation involve spinal microglia and are independent of KCC2 expression in a mouse model of complete spinal transection. Exp Neurol 2021; 338:113592. [PMID: 33388315 PMCID: PMC7904639 DOI: 10.1016/j.expneurol.2020.113592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 12/03/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
Spinal cord injury (SCI) is associated with damage to musculoskeletal tissues of the spine. Recent findings show that pain and inflammatory processes caused by musculoskeletal injury mediate plastic changes in the spinal cord. These changes could impede the adaptive plastic changes responsible for functional recovery. The underlying mechanism remains unclear, but may involve the microglia-BDNF-KCC2 pathway, which is implicated in sensitization of dorsal horn neurons in neuropathic pain and in the regulation of spinal excitability by step-training. In the present study, we examined the effects of step-training and lumbar muscle inflammation induced by complete Freund's adjuvant (CFA) on treadmill locomotion in a mouse model of complete spinal transection. The impact on locomotor recovery of each of these interventions alone or in combination were examined in addition to changes in microglia and KCC2 expression in the dorsal and ventral horns of the sublesional spinal cord. Results show that angular motion at the hip, knee and ankle joint during locomotion were decreased by CFA injection and improved by step-training. Moreover, CFA injection enhanced the expression of the microglial marker Iba1 in both ventral and dorsal horns, with or without step-training. However, this change was not associated with a modulation of KCC2 expression, suggesting that locomotor deficits induced by inflammation are independent of KCC2 expression in the sublesional spinal cord. These results indicate that musculoskeletal injury hinders locomotor recovery after SCI and that microglia is involved in this effect.
Collapse
Affiliation(s)
- Renaud Jeffrey-Gauthier
- Department of Anatomy, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada; CogNAC Research Group, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada.
| | - Julien Bouyer
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA 19129, United States.
| | - Mathieu Piché
- Department of Anatomy, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada; CogNAC Research Group, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada.
| | - Marie-Pascale Côté
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA 19129, United States.
| | - Hugues Leblond
- Department of Anatomy, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada; CogNAC Research Group, Université du Québec à Trois-Rivières, 3351 boul. des Forges, C.P. 500, Trois-Rivières, QC G9A 5H7, Canada.
| |
Collapse
|
12
|
Li X, Qian Y, Tang K, Li Y, Tao R, Gong C, Huang L, Zou K, Liu L. Inhibition of lncRNA H19/miR-370-3p pathway mitigates neuronal apoptosis in an in vitro model of spinal cord injury (SCI). Transl Neurosci 2021; 12:103-113. [PMID: 33708438 PMCID: PMC7925972 DOI: 10.1515/tnsci-2021-0013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/05/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is the most serious complication of spinal injury, often leading to severe dysfunction of the limbs below the injured segment. Conventional therapy approaches are becoming less and less effective, and gene therapy is a new research direction by now. METHODS The Sprague-Dawley rats were haphazardly assigned to two groups, namely sham group and SCI model group, and lncRNA H19 and miR-370-3p levels were investigated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Correlation between lncRNA H19 and miR-370-3p was ascertained by luciferase report assay and RT-qPCR. After transfection with si-H19, miR-370-3p inhibitor, negative controls (NC), or both, primary spinal neurons were subjected to the simulation of lipopolysaccharide (LPS) for inducing in vitro model of SCI. Cell viability, apoptotic rate, caspase-3 activity, Bax and Bcl-2 protein, ROS generation, TNF-α, IL-1β, and IL-6 protein, as well as IκBα and p65 phosphorylation ratio were evaluated adopting 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), apoptosis, caspase-3 activity, ROS generation, and western blot assays, thereby searching for the specific action mechanism on LPS-induced spinal never injury. RESULTS SCI resulted in lncRNA H19 higher expression and miR-370-3p lower expression. LPS simulation raised a series of cellular biological changes, such as decreased viability, promoted apoptosis, generated ROS, and released inflammatory factors. lncRNA H19 inhibition reversed above LPS-induced changes. Besides, as the downstream target of lncRNA H19, miR-370-3p was oppositely regulated by lncRNA H19. The above biological changes induced by lncRNA H19 inhibition were reversed by miR-370-3p upregulation. Moreover, lncRNA H19 inhibition could block NF-κB pathway through miR-370-3p upregulation. CONCLUSION Inhibition of lncRNA H19/miR-370-3p mitigated spinal neuron apoptosis in an in vitro model of SCI. This provided the possibility for clinical use of gene therapy.
Collapse
Affiliation(s)
- Xin Li
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Yan Qian
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Kaihua Tang
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Yang Li
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Rui Tao
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Chunyan Gong
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Li Huang
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Kaiwen Zou
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| | - Lindong Liu
- Department of Rehabilitation Medicine, Qujing No. 1 Hospital, Yuanlin No. 1 Road, Qilin District, Qujing 655000, Yunnan, China
| |
Collapse
|
13
|
Wada N, Yoshimura N, Kurobe M, Saito T, Tyagi P, Kakizaki H. The early, long‐term inhibition of brain‐derived neurotrophic factor improves voiding, and storage dysfunctions in mice with spinal cord injury. Neurourol Urodyn 2020; 39:1345-1354. [DOI: 10.1002/nau.24385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/23/2020] [Accepted: 04/30/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Naoki Wada
- Department of Renal and Urologic SurgeryAsahikawa Medical UniversityAsahikawa Japan
- Department of UrologyUniversity of Pittsburgh School of MedicinePittsburgh Pennsylvania
| | - Naoki Yoshimura
- Department of UrologyUniversity of Pittsburgh School of MedicinePittsburgh Pennsylvania
| | - Masahiro Kurobe
- Department of UrologyUniversity of Pittsburgh School of MedicinePittsburgh Pennsylvania
| | - Tetsuichi Saito
- Department of UrologyUniversity of Pittsburgh School of MedicinePittsburgh Pennsylvania
| | - Pradeep Tyagi
- Department of UrologyUniversity of Pittsburgh School of MedicinePittsburgh Pennsylvania
| | - Hidehiro Kakizaki
- Department of Renal and Urologic SurgeryAsahikawa Medical UniversityAsahikawa Japan
| |
Collapse
|
14
|
Zhao J, Zhang Q, Zhang B, Xu T, Yin D, Gu W, Bai J. Developmental exposure to lead at environmentally relevant concentrations impaired neurobehavior and NMDAR-dependent BDNF signaling in zebrafish larvae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 257:113627. [PMID: 31796321 DOI: 10.1016/j.envpol.2019.113627] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 06/10/2023]
Abstract
Lead (Pb) is one of the predominant heavy metals in e-waste recycling arears and recognized as a notorious environmental neurotoxic substance. However, whether Pb at environmentally relevant concentrations could cause neurobehavioral alteration and even what kind of signaling pathway Pb exposure would disrupt in zebrafish were not fully uncovered. In the present study, 6 h postfertilization (hpf) zebrafish embryos were exposed to Pb at the concentrations of 0, 5, 10, and 20 μg/L until 144 hpf. Then the neurobehavioral indicators including locomotor, turnings and social behaviors, and the expressions of selected genes concerning brain-derived neurotrophic factor (BDNF) signaling were investigated. The results showed that significant changes were obtained under 20 μg/L Pb exposure. The hypoactivity of zebrafish larvae in locomotor and turning behaviors was induced during the dark period, while hyperactivity was observed in a two-fish social assay during the light period. The significantly downregulation of genes encoding BDNF, its receptor TrkB, and N-methyl-D-aspartate glutamate receptor (NMDAR) suggested the involvement of NMDAR-dependent BDNF signaling pathway. Overall, our study demonstrated that developmental exposure to Pb at environmentally relevant concentrations caused obvious neurobehavioral impairment of zebrafish larvae by disrupting the NMDAR-dependent BDNF signaling, which could exert profound ecological consequences in the real environment.
Collapse
Affiliation(s)
- Jing Zhao
- WEEE Research Centre of Shanghai Polytechnic University, Shanghai Collaborative Innovation Centre for WEEE Recycling, Shanghai, 201209, China; Research Center of Resource Recycling Science and Engineering, Shanghai Polytechnic University, Shanghai, 201209, China
| | - Qing Zhang
- WEEE Research Centre of Shanghai Polytechnic University, Shanghai Collaborative Innovation Centre for WEEE Recycling, Shanghai, 201209, China; Research Center of Resource Recycling Science and Engineering, Shanghai Polytechnic University, Shanghai, 201209, China
| | - Bin Zhang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Ting Xu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China.
| | - Daqiang Yin
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Weihua Gu
- WEEE Research Centre of Shanghai Polytechnic University, Shanghai Collaborative Innovation Centre for WEEE Recycling, Shanghai, 201209, China; Research Center of Resource Recycling Science and Engineering, Shanghai Polytechnic University, Shanghai, 201209, China
| | - Jianfeng Bai
- WEEE Research Centre of Shanghai Polytechnic University, Shanghai Collaborative Innovation Centre for WEEE Recycling, Shanghai, 201209, China; Research Center of Resource Recycling Science and Engineering, Shanghai Polytechnic University, Shanghai, 201209, China
| |
Collapse
|
15
|
Reynolds JA, Henwood MK, Turtle JD, Baine RE, Johnston DT, Grau JW. Brain-Dependent Processes Fuel Pain-Induced Hemorrhage After Spinal Cord Injury. Front Syst Neurosci 2019; 13:44. [PMID: 31551720 PMCID: PMC6746957 DOI: 10.3389/fnsys.2019.00044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/15/2019] [Indexed: 12/15/2022] Open
Abstract
Pain (nociceptive) input caudal to a spinal contusion injury can undermine long-term recovery and increase tissue loss (secondary injury). Prior work suggests that nociceptive stimulation has this effect because it fosters the breakdown of the blood-spinal cord barrier (BSCB) at the site of injury, allowing blood to infiltrate the tissue. The present study examined whether these effects impact tissue rostral and caudal to the site of injury. In addition, the study evaluated whether cutting communication with the brain, by means of a rostral transection, affects the development of hemorrhage. Eighteen hours after rats received a lower thoracic (T11-12) contusion injury, half underwent a spinal transection at T2. Noxious electrical stimulation (shock) was applied 6 h later. Cellular assays showed that, in non-transected rats, nociceptive stimulation increased hemoglobin content, activated pro-inflammatory cytokines and engaged signals related to cell death at the site of injury. These effects were not observed in transected animals. In the next experiment, the spinal transection was performed at the time of contusion injury. Nociceptive stimulation was applied 24 h later and tissue was sectioned for microscopy. In non-transected rats, nociceptive stimulation increased the area of hemorrhage and this effect was blocked by spinal transection. These findings imply that the adverse effect of noxious stimulation depends upon spared ascending fibers and the activation of rostral (brain) systems. If true, stimulation should induce less hemorrhage after a severe contusion injury that blocks transmission to the brain. To test this, rats were given a mild, moderate, or severe, injury and electrical stimulation was applied 24 h later. Histological analyses of longitudinal sections showed that nociceptive stimulation triggered less hemorrhage after a severe contusion injury. The results suggest that brain-dependent processes drive pain-induced hemorrhage after spinal cord injury (SCI).
Collapse
Affiliation(s)
- Joshua A Reynolds
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| | - Melissa K Henwood
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| | - Joel D Turtle
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| | - Rachel E Baine
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| | - David T Johnston
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| | - James W Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
16
|
Brakel K, Hook MA. SCI and depression: Does inflammation commandeer the brain? Exp Neurol 2019; 320:112977. [PMID: 31203113 DOI: 10.1016/j.expneurol.2019.112977] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
The incidence of depression is almost twice as high in the spinally injured population compared to the general population. While this incidence has long been attributed to the psychological, economic, and social burdens that accompany spinal cord injury (SCI), data from animal studies indicate that the biology of SCI may play an important role in the development of depression. Inflammation has been shown to impact stress response in rodents and humans, and inflammatory cytokines have been associated with depression for decades. The inflammation inherent to SCI may disrupt necessary mechanisms of mental homeostasis, such as serotonin production, dopamine production, and the hypothalamic pituitary adrenal axis. Additionally, gut dysbiosis that occurs after SCI can exacerbate inflammation and may cause further mood and behavior changes. These mediators combined may significantly contribute to the rise in depression seen after SCI. Currently, there are no therapies specific to depression after SCI. Elucidation of the molecular pathways that contribute to SCI-specific depression is crucial for the understanding of this disease and its potential treatments.
Collapse
Affiliation(s)
- Kiralyn Brakel
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Medical Research and Education Building, Ste. 1005, 8447 Riverside Pkwy, Bryan, TX 77807, United States; Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474 College Station, TAMU, TX, United States.
| | - Michelle A Hook
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Medical Research and Education Building, Ste. 1005, 8447 Riverside Pkwy, Bryan, TX 77807, United States; Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474 College Station, TAMU, TX, United States
| |
Collapse
|
17
|
Martin KK, Parvin S, Garraway SM. Peripheral Inflammation Accelerates the Onset of Mechanical Hypersensitivity after Spinal Cord Injury and Engages Tumor Necrosis Factor α Signaling Mechanisms. J Neurotrauma 2019; 36:2000-2010. [PMID: 30520675 DOI: 10.1089/neu.2018.5953] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Previously, we showed that noxious stimulation of the tail produces numerous detrimental effects after spinal cord injury (SCI), including an earlier onset and increased magnitude of mechanical hypersensitivity. Expanding on these observations, this study sought to determine whether localized peripheral inflammation similarly impacts the expression of mechanical hypersensitivity after SCI. Adult rats received a moderate contusion injury at the thoracic level (Tl0) or sham surgery, and were administered complete Freund's adjuvant (CFA) or vehicle in one hindpaw 24 hours later. Examination of locomotor recovery (Basso, Beattie, and Bresnahan [BBB] score) showed no adverse effect of CFA. Mechanical testing with von Frey hairs was done at time-points ranging from 1 h to 28 days after CFA or vehicle treatment, and rats were sacrificed at 1, 7, or 28 days for cellular assessment. Unlike vehicle-treated SCI rats where mechanical hypersensitivity emerged at 14 days, CFA-treated SCI rats showed mechanical hypersensitivity as early as 1 h after CFA administration, which lasted at least 28 days. CFA-treated sham subjects also showed an early onset of mechanical hypersensitivity, but this was maintained up to 7 days after treatment. Cellular assessments revealed congruent findings. Expression levels of c-fos, tumor necrosis factor α (TNFα), TNF receptors, and members of the TNFα signaling pathway such as caspase 8 and phosphorylated extracellular related kinase (pERK) were preferentially upregulated in the lumbar spinal cord of SCI-CFA rats. Meanwhile, c-jun was significantly increased in both CFA-treated groups. Overall, these results together with our previous reports, suggest that peripheral noxious input after SCI facilitates the development of pain by mechanisms that may require TNFα signaling.
Collapse
Affiliation(s)
- Karmarcha K Martin
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Shangrila Parvin
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
18
|
Brumley MR, Strain MM, Devine N, Bozeman AL. The Spinal Cord, Not to Be Forgotten: the Final Common Path for Development, Training and Recovery of Motor Function. Perspect Behav Sci 2018; 41:369-393. [PMID: 31976401 DOI: 10.1007/s40614-018-00177-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Research on learning, memory, and neural plasticity has long focused on the brain. However, the spinal cord also exhibits these phenomena to a remarkable degree. Following a spinal cord injury, the isolated spinal cord in vivo can adapt to the environment and benefit from training. The amount of plasticity or recovery of function following a spinal injury often depends on the age at which the injury occurs. In this overview, we discuss learning in the spinal cord, including associative conditioning, neural mechanisms, development, and applications to clinical populations. We take an integrated approach to the spinal cord, one that combines basic and experimental information about experience-dependent learning in animal models to clinical treatment of spinal cord injuries in humans. From such an approach, an important goal is to better inform therapeutic treatments for individuals with spinal cord injuries, as well as develop a more accurate and complete account of spinal cord and behavioral functioning.
Collapse
Affiliation(s)
- Michele R Brumley
- 1Department of Psychology, Idaho State University, 921 South 8th Avenue, Stop 8112, Pocatello, ID 83209-8112 USA
| | - Misty M Strain
- 2United States Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX USA
| | - Nancy Devine
- 3Department of Physical and Occupational Therapy, Idaho State University, Pocatello, ID USA
| | - Aimee L Bozeman
- 1Department of Psychology, Idaho State University, 921 South 8th Avenue, Stop 8112, Pocatello, ID 83209-8112 USA
| |
Collapse
|
19
|
Turtle JD, Strain MM, Reynolds JA, Huang YJ, Lee KH, Henwood MK, Garraway SM, Grau JW. Pain Input After Spinal Cord Injury (SCI) Undermines Long-Term Recovery and Engages Signal Pathways That Promote Cell Death. Front Syst Neurosci 2018; 12:27. [PMID: 29977195 PMCID: PMC6021528 DOI: 10.3389/fnsys.2018.00027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/17/2018] [Indexed: 12/19/2022] Open
Abstract
Pain (nociceptive) input caudal to a spinal contusion injury increases tissue loss and impairs long-term recovery. It was hypothesized that noxious stimulation has this effect because it engages unmyelinated pain (C) fibers that produce a state of over-excitation in central pathways. The present article explored this issue by assessing the effect of capsaicin, which activates C-fibers that express the transient receptor potential vanilloid receptor-1 (TRPV1). Rats received a lower thoracic (T11) contusion injury and capsaicin was applied to one hind paw the next day. For comparison, other animals received noxious electrical stimulation at an intensity that engages C fibers. Both forms of stimulation elicited similar levels of c-fos mRNA expression, a cellular marker of nociceptive activation, and impaired long-term behavioral recovery. Cellular assays were then performed to compare the acute effect of shock and capsaicin treatment. Both forms of noxious stimulation increased expression of tumor necrosis factor (TNF) and caspase-3, which promotes apoptotic cell death. Shock, but not capsaicin, enhanced expression of signals related to pyroptotic cell death [caspase-1, inteleukin-1 beta (IL-1ß)]. Pyroptosis has been linked to the activation of the P2X7 receptor and the outward flow of adenosine triphosphate (ATP) through the pannexin-1 channel. Blocking the P2X7 receptor with Brilliant Blue G (BBG) reduced the expression of signals related to pyroptotic cell death in contused rats that had received shock. Blocking the pannexin-1 channel with probenecid paradoxically had the opposite effect. BBG enhanced long-term recovery and lowered reactivity to mechanical stimulation applied to the girdle region (an index of chronic pain), but did not block the adverse effect of nociceptive stimulation. The results suggest that C-fiber input after injury impairs long-term recovery and that this effect may arise because it induces apoptotic cell death.
Collapse
Affiliation(s)
- Joel D Turtle
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Misty M Strain
- United States Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX, United States
| | - Joshua A Reynolds
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Yung-Jen Huang
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Kuan H Lee
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Melissa K Henwood
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - James W Grau
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
20
|
Grau JW, Huang YJ. Metaplasticity within the spinal cord: Evidence brain-derived neurotrophic factor (BDNF), tumor necrosis factor (TNF), and alterations in GABA function (ionic plasticity) modulate pain and the capacity to learn. Neurobiol Learn Mem 2018; 154:121-135. [PMID: 29635030 DOI: 10.1016/j.nlm.2018.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/01/2018] [Accepted: 04/06/2018] [Indexed: 12/15/2022]
Abstract
Evidence is reviewed that behavioral training and neural injury can engage metaplastic processes that regulate adaptive potential. This issue is explored within a model system that examines how training affects the capacity to learn within the lower (lumbosacral) spinal cord. Response-contingent (controllable) stimulation applied caudal to a spinal transection induces a behavioral modification indicative of learning. This behavioral change is not observed in animals that receive stimulation in an uncontrollable manner. Exposure to uncontrollable stimulation also engages a process that disables spinal learning for 24-48 h. Controllable stimulation has the opposite effect; it engages a process that enables learning and prevents/reverses the learning deficit induced by uncontrollable stimulation. These observations suggest that a learning episode can impact the capacity to learn in future situations, providing an example of behavioral metaplasticity. The protective/restorative effect of controllable stimulation has been linked to an up-regulation of brain-derived neurotrophic factor (BDNF). The disruption of learning has been linked to the sensitization of pain (nociceptive) circuits, which is enabled by a reduction in GABA-dependent inhibition. After spinal cord injury (SCI), the co-transporter (KCC2) that regulates the outward flow of Cl- is down-regulated. This causes the intracellular concentration of Cl- to increase, reducing (and potentially reversing) the inward flow of Cl- through the GABA-A receptor. The shift in GABA function (ionic plasticity) increases neural excitability caudal to injury and sets the stage for nociceptive sensitization. The injury-induced shift in KCC2 is related to the loss of descending serotonergic (5HT) fibers that regulate plasticity within the spinal cord dorsal horn through the 5HT-1A receptor. Evidence is presented that these alterations in spinal plasticity impact pain in a brain-dependent task (place conditioning). The findings suggest that ionic plasticity can affect learning potential, shifting a neural circuit from dampened/hard-wired to excitable/plastic.
Collapse
Affiliation(s)
- James W Grau
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843-4235, USA.
| | - Yung-Jen Huang
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843-4235, USA
| |
Collapse
|
21
|
Jeffrey-Gauthier R, Piché M, Leblond H. Lumbar muscle inflammation alters spinally mediated locomotor recovery induced by training in a mouse model of complete spinal cord injury. Neuroscience 2017; 359:69-81. [DOI: 10.1016/j.neuroscience.2017.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/30/2017] [Accepted: 07/06/2017] [Indexed: 02/07/2023]
|
22
|
Grau JW, Huang YJ, Turtle JD, Strain MM, Miranda RC, Garraway SM, Hook MA. When Pain Hurts: Nociceptive Stimulation Induces a State of Maladaptive Plasticity and Impairs Recovery after Spinal Cord Injury. J Neurotrauma 2017; 34:1873-1890. [PMID: 27788626 PMCID: PMC5444485 DOI: 10.1089/neu.2016.4626] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is often accompanied by other tissue damage (polytrauma) that provides a source of pain (nociceptive) input. Recent findings are reviewed that show SCI places the caudal tissue in a vulnerable state that exaggerates the effects nociceptive stimuli and promotes the development of nociceptive sensitization. Stimulation that is both unpredictable and uncontrollable induces a form of maladaptive plasticity that enhances nociceptive sensitization and impairs spinally mediated learning. In contrast, relational learning induces a form of adaptive plasticity that counters these adverse effects. SCI sets the stage for nociceptive sensitization by disrupting serotonergic (5HT) fibers that quell overexcitation. The loss of 5HT can enhance neural excitability by reducing membrane-bound K+-Cl- cotransporter 2, a cotransporter that regulates the outward flow of Cl-. This increases the intracellular concentration of Cl-, which reduces the hyperpolarizing (inhibitory) effect of gamma-aminobutyric acid. Uncontrollable noxious stimulation also undermines the recovery of locomotor function, and increases behavioral signs of chronic pain, after a contusion injury. Nociceptive stimulation has a greater effect if experienced soon after SCI. This adverse effect has been linked to a downregulation in brain-derived neurotrophic factor and an upregulation in the cytokine, tumor necrosis factor. Noxious input enhances tissue loss at the site of injury by increasing the extent of hemorrhage and apoptotic/pyroptotic cell death. Intrathecal lidocaine blocks nociception-induced hemorrhage, cellular indices of cell death, and its adverse effect on behavioral recovery. Clinical implications are discussed.
Collapse
Affiliation(s)
- James W. Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Yung-Jen Huang
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Joel D. Turtle
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Misty M. Strain
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Rajesh C. Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas
| | - Sandra M. Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Michelle A. Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas
| |
Collapse
|
23
|
Fixed spaced stimulation restores adaptive plasticity within the spinal cord: Identifying the eliciting conditions. Physiol Behav 2017; 174:1-9. [PMID: 28238778 DOI: 10.1016/j.physbeh.2017.02.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/22/2017] [Indexed: 01/21/2023]
Abstract
Prior work has shown that neurons within the spinal cord are sensitive to temporal relations and that stimulus regularity impacts nociceptive processing and adaptive plasticity. Application of brief (80ms) shocks (180-900) in a variable manner induces a form of maladaptive plasticity that inhibits spinally-mediated learning and enhances nociceptive reactivity. In contrast, an extended exposure (720-900) to stimuli given at regular (fixed spaced) intervals has a restorative effect that counters nociceptive sensitization and enables learning. The present paper explores the stimulus parameters under which this therapeutic effect of fixed spaced stimulation emerges. Spinally transected rats received variably spaced stimulation (180 shocks) to the sciatic nerve at an intensity (40-V) that recruits pain (C) fibers, producing a form of maladaptive plasticity that impairs spinal learning. As previously shown, exposure to 720 fixed spaced shocks had a therapeutic effect that restored adaptive learning. This therapeutic effect was most robust at a lower shock intensity (20V) and was equally strong irrespective of pulse duration (20-80ms). A restorative effect was observed when stimuli were given at a frequency between 0.5 and 5Hz, but not at a higher (50Hz) or lower (0.05Hz) rate. The results are consistent with prior work implicating neural systems related to the central pattern generator that drives stepping behavior. Clinical implications are discussed.
Collapse
|
24
|
Huie JR, Morioka K, Haefeli J, Ferguson AR. What Is Being Trained? How Divergent Forms of Plasticity Compete To Shape Locomotor Recovery after Spinal Cord Injury. J Neurotrauma 2017; 34:1831-1840. [PMID: 27875927 DOI: 10.1089/neu.2016.4562] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating syndrome that produces dysfunction in motor and sensory systems, manifesting as chronic paralysis, sensory changes, and pain disorders. The multi-faceted and heterogeneous nature of SCI has made effective rehabilitative strategies challenging. Work over the last 40 years has aimed to overcome these obstacles by harnessing the intrinsic plasticity of the spinal cord to improve functional locomotor recovery. Intensive training after SCI facilitates lower extremity function and has shown promise as a tool for retraining the spinal cord by engaging innate locomotor circuitry in the lumbar cord. As new training paradigms evolve, the importance of appropriate afferent input has emerged as a requirement for adaptive plasticity. The integration of kinematic, sensory, and loading force information must be closely monitored and carefully manipulated to optimize training outcomes. Inappropriate peripheral input may produce lasting maladaptive sensory and motor effects, such as central pain and spasticity. Thus, it is important to closely consider the type of afferent input the injured spinal cord receives. Here we review preclinical and clinical input parameters fostering adaptive plasticity, as well as those producing maladaptive plasticity that may undermine neurorehabilitative efforts. We differentiate between passive (hindlimb unloading [HU], limb immobilization) and active (peripheral nociception) forms of aberrant input. Furthermore, we discuss the timing of initiating exposure to afferent input after SCI for promoting functional locomotor recovery. We conclude by presenting a candidate rapid synaptic mechanism for maladaptive plasticity after SCI, offering a pharmacological target for restoring the capacity for adaptive spinal plasticity in real time.
Collapse
Affiliation(s)
- J Russell Huie
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California
| | - Kazuhito Morioka
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California
| | - Jenny Haefeli
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California
| | - Adam R Ferguson
- 1 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California , San Francisco, California.,2 San Francisco Veterans Affairs Medical Center , San Francisco, California
| |
Collapse
|
25
|
Turtle JD, Strain MM, Aceves M, Huang YJ, Reynolds JA, Hook MA, Grau JW. Pain Input Impairs Recovery after Spinal Cord Injury: Treatment with Lidocaine. J Neurotrauma 2016; 34:1200-1208. [PMID: 27912032 DOI: 10.1089/neu.2016.4778] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
More than 90% of spinal cord injuries are caused by traumatic accidents and are often associated with other tissue damage (polytrauma) that can provide a source of continued pain input during recovery. In a clinically relevant spinal cord contusion injury model, prior work has shown that noxious stimulation at an intensity that engages pain (C) fibers soon after injury augments secondary injury and impairs functional recovery. Noxious input increases the expression of pro-inflammatory cytokines (interleukin 1β and 18), cellular signals associated with cell death (caspase 3 and 8), and physiological signs of hemorrhage. Here, it is shown that reducing neural excitability after spinal cord injury (SCI) with the local anesthetic lidocaine (micro-injected by means of a lumbar puncture) blocks these adverse cellular effects. In contrast, treatment with an analgesic dose of morphine had no effect. Contused rats that received nociceptive stimulation soon after injury exhibited poor locomotor recovery, less weight gain, and greater tissue loss at the site of injury. Prophylactic application of lidocaine blocked the adverse effect of nociceptive stimulation on behavioral recovery and reduced tissue loss from secondary injury. The results suggest that quieting neural excitability using lidocaine can reduce the adverse effect of pain input (from polytrauma or surgery) after SCI.
Collapse
Affiliation(s)
- Joel D Turtle
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Misty M Strain
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Miriam Aceves
- 2 Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University , College Station, Texas
| | - Yung-Jen Huang
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Joshua A Reynolds
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Michelle A Hook
- 2 Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University , College Station, Texas
| | - James W Grau
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| |
Collapse
|
26
|
Cornel Iridoid Glycoside Improves Locomotor Impairment and Decreases Spinal Cord Damage in Rats. BIOMED RESEARCH INTERNATIONAL 2016; 2016:6725381. [PMID: 27990434 PMCID: PMC5136393 DOI: 10.1155/2016/6725381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/06/2016] [Accepted: 10/04/2016] [Indexed: 01/21/2023]
Abstract
Purpose. This study was to investigate the effects of cornel iridoid glycoside (CIG) on spinal cord injury (SCI) in rats. Methods. The thoracic cord (at T9) of rats was injured by clip compression for 30 sec. Locomotor function was assessed using the Basso, Beattie, and Bresnahan (BBB) rating scale. Neuroanatomic stereological parameters as well as Nogo-A, p75 neurotrophin receptor (p75NTR), and ROCKII expression were measured by histological processing, immunohistochemistry, and stereological analyses. The axons passing through the lesion site were detected by BDA tracing. Results. Intragastric administration of CIG (60 and 180 mg/kg) improved the locomotor impairment at 10, 17, 24, and 31 days post-injury (dpi) compared with untreated SCI model rats. CIG treatment decreased the volume of the lesion epicenter (LEp) and increased the volume of spared tissue and the number of surviving neurons in the injured spinal cord at 31 dpi. CIG promoted the growth of BDA-positive axons and their passage through the lesion site and decreased the expression of Nogo-A, p75NTR, and ROCKII both in and around the LEp. Conclusion. CIG improved the locomotor impairment, decreased tissue damage, and downregulated the myelin-associated inhibition signaling pathway in SCI rats. The results suggest that CIG may be beneficial for SCI therapy.
Collapse
|
27
|
Aceves M, Bancroft EA, Aceves AR, Hook MA. Nor-Binaltorphimine Blocks the Adverse Effects of Morphine after Spinal Cord Injury. J Neurotrauma 2016; 34:1164-1174. [PMID: 27736318 DOI: 10.1089/neu.2016.4601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Opioids are frequently used for the treatment of pain following spinal cord injury (SCI). Unfortunately, we have shown that morphine administered in the acute phase of SCI results in significant, adverse secondary consequences including compromised locomotor and sensory recovery. Similarly, we showed that selective activation of the κ-opioid receptor (KOR), even at a dose 32-fold lower than morphine, is sufficient to attenuate recovery of locomotor function. In the current study, we tested whether activation of the KOR is necessary to produce morphine's adverse effects using nor-Binaltorphimine (norBNI), a selective KOR antagonist. Rats received a moderate spinal contusion (T12) and 24 h later, baseline locomotor function and nociceptive reactivity were assessed. Rats were then administered norBNI (0, 0.02, 0.08, or 0.32 μmol) followed by morphine (0 or 0.32 μmol). Nociception was reassessed 30 min after drug treatment, and recovery was evaluated for 21 days. The effects of norBNI on morphine-induced attenuation of recovery were dose dependent. At higher doses, norBNI blocked the adverse effects of morphine on locomotor recovery, but analgesia was also significantly decreased. Conversely, at low doses, analgesia was maintained, but the adverse effects on recovery persisted. A moderate dose of norBNI, however, adequately protected against morphine's adverse effects without eliminating its analgesic efficacy. This suggests that activation of the KOR system plays a significant role in the morphine-induced attenuation of recovery. Our research suggests that morphine, and other opioid analgesics, may be contraindicated for the SCI population. Blocking KOR activity may be a viable strategy for improving the safety of clinical opioid use.
Collapse
Affiliation(s)
- Miriam Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Eric A Bancroft
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Alejandro R Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| |
Collapse
|
28
|
Spinal Plasticity and Behavior: BDNF-Induced Neuromodulation in Uninjured and Injured Spinal Cord. Neural Plast 2016; 2016:9857201. [PMID: 27721996 PMCID: PMC5046018 DOI: 10.1155/2016/9857201] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/27/2016] [Accepted: 08/10/2016] [Indexed: 12/26/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophic factor family of signaling molecules. Since its discovery over three decades ago, BDNF has been identified as an important regulator of neuronal development, synaptic transmission, and cellular and synaptic plasticity and has been shown to function in the formation and maintenance of certain forms of memory. Neural plasticity that underlies learning and memory in the hippocampus shares distinct characteristics with spinal cord nociceptive plasticity. Research examining the role BDNF plays in spinal nociception and pain overwhelmingly suggests that BDNF promotes pronociceptive effects. BDNF induces synaptic facilitation and engages central sensitization-like mechanisms. Also, peripheral injury-induced neuropathic pain is often accompanied with increased spinal expression of BDNF. Research has extended to examine how spinal cord injury (SCI) influences BDNF plasticity and the effects BDNF has on sensory and motor functions after SCI. Functional recovery and adaptive plasticity after SCI are typically associated with upregulation of BDNF. Although neuropathic pain is a common consequence of SCI, the relation between BDNF and pain after SCI remains elusive. This article reviews recent literature and discusses the diverse actions of BDNF. We also highlight similarities and differences in BDNF-induced nociceptive plasticity in naïve and SCI conditions.
Collapse
|
29
|
Lee KH, Huang YJ, Grau JW. Learning about Time within the Spinal Cord II: Evidence that Temporal Regularity Is Encoded by a Spinal Oscillator. Front Behav Neurosci 2016; 10:14. [PMID: 26903830 PMCID: PMC4749712 DOI: 10.3389/fnbeh.2016.00014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/28/2016] [Indexed: 11/13/2022] Open
Abstract
How a stimulus impacts spinal cord function depends upon temporal relations. When intermittent noxious stimulation (shock) is applied and the interval between shock pulses is varied (unpredictable), it induces a lasting alteration that inhibits adaptive learning. If the same stimulus is applied in a temporally regular (predictable) manner, the capacity to learn is preserved and a protective/restorative effect is engaged that counters the adverse effect of variable stimulation. Sensitivity to temporal relations implies a capacity to encode time. This study explores how spinal neurons discriminate variable and fixed spaced stimulation. Communication with the brain was blocked by means of a spinal transection and adaptive capacity was tested using an instrumental learning task. In this task, subjects must learn to maintain a hind limb in a flexed position to minimize shock exposure. To evaluate the possibility that a distinct class of afferent fibers provide a sensory cue for regularity, we manipulated the temporal relation between shocks given to two dermatomes (leg and tail). Evidence for timing emerged when the stimuli were applied in a coherent manner across dermatomes, implying that a central (spinal) process detects regularity. Next, we show that fixed spaced stimulation has a restorative effect when half the physical stimuli are randomly omitted, as long as the stimuli remain in phase, suggesting that stimulus regularity is encoded by an internal oscillator Research suggests that the oscillator that drives the tempo of stepping depends upon neurons within the rostral lumbar (L1-L2) region. Disrupting communication with the L1-L2 tissue by means of a L3 transection eliminated the restorative effect of fixed spaced stimulation. Implications of the results for step training and rehabilitation after injury are discussed.
Collapse
Affiliation(s)
- Kuan H Lee
- Department of Neurobiology, Center for Pain Research, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Yung-Jen Huang
- Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University College Station, TX, USA
| | - James W Grau
- Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University College Station, TX, USA
| |
Collapse
|
30
|
Khaing ZZ, Agrawal NK, Park JH, Xin S, Plumton GC, Lee KH, Huang YJ, Niemerski AL, Schmidt CE, Grau JW. Localized and sustained release of brain-derived neurotrophic factor from injectable hydrogel/microparticle composites fosters spinal learning after spinal cord injury. J Mater Chem B 2016; 4:7560-7571. [DOI: 10.1039/c6tb01602b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Injectable hydrogel allows for sustained delivery of growth factor resulting in spinal mediated learning after injury.
Collapse
Affiliation(s)
- Zin Z. Khaing
- Department of Biomedical Engineering
- University of Florida
- Gainesville
- USA
| | - Nikunj K. Agrawal
- Department of Biomedical Engineering
- University of Florida
- Gainesville
- USA
| | - James H. Park
- College of Medicine
- University of Florida
- Gainesville
- USA
| | - Shangjing Xin
- Department of Materials Science and Engineering
- University of Florida
- Gainesville
- USA
| | | | - Kuan H. Lee
- Department of Neurobiology
- University of Pittsburgh School of Medicine
- Pittsburgh
- USA
| | - Yung-Jen Huang
- Department of Psychology
- Texas A&M University
- College Station
- USA
| | | | | | - James W. Grau
- Department of Psychology
- Texas A&M University
- College Station
- USA
| |
Collapse
|
31
|
AMPA Receptor Phosphorylation and Synaptic Colocalization on Motor Neurons Drive Maladaptive Plasticity below Complete Spinal Cord Injury. eNeuro 2015; 2:eN-NWR-0091-15. [PMID: 26668821 PMCID: PMC4677690 DOI: 10.1523/eneuro.0091-15.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/05/2015] [Accepted: 10/05/2015] [Indexed: 12/22/2022] Open
Abstract
Clinical spinal cord injury (SCI) is accompanied by comorbid peripheral injury in 47% of patients. Human and animal modeling data have shown that painful peripheral injuries undermine long-term recovery of locomotion through unknown mechanisms. Peripheral nociceptive stimuli induce maladaptive synaptic plasticity in dorsal horn sensory systems through AMPA receptor (AMPAR) phosphorylation and trafficking to synapses. Here we test whether ventral horn motor neurons in rats demonstrate similar experience-dependent maladaptive plasticity below a complete SCI in vivo. Quantitative biochemistry demonstrated that intermittent nociceptive stimulation (INS) rapidly and selectively increases AMPAR subunit GluA1 serine 831 phosphorylation and localization to synapses in the injured spinal cord, while reducing synaptic GluA2. These changes predict motor dysfunction in the absence of cell death signaling, suggesting an opportunity for therapeutic reversal. Automated confocal time-course analysis of lumbar ventral horn motor neurons confirmed a time-dependent increase in synaptic GluA1 with concurrent decrease in synaptic GluA2. Optical fractionation of neuronal plasma membranes revealed GluA2 removal from extrasynaptic sites on motor neurons early after INS followed by removal from synapses 2 h later. As GluA2-lacking AMPARs are canonical calcium-permeable AMPARs (CP-AMPARs), their stimulus- and time-dependent insertion provides a therapeutic target for limiting calcium-dependent dynamic maladaptive plasticity after SCI. Confirming this, a selective CP-AMPAR antagonist protected against INS-induced maladaptive spinal plasticity, restoring adaptive motor responses on a sensorimotor spinal training task. These findings highlight the critical involvement of AMPARs in experience-dependent spinal cord plasticity after injury and provide a pharmacologically targetable synaptic mechanism by which early postinjury experience shapes motor plasticity.
Collapse
|
32
|
Lentivirus-mediated inhibition of tumour necrosis factor-α improves motor function associated with PRDX6 in spinal cord contusion rats. Sci Rep 2015; 5:8486. [PMID: 25686213 PMCID: PMC4329786 DOI: 10.1038/srep08486] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 01/09/2015] [Indexed: 02/05/2023] Open
Abstract
The recovery of motor function in rats is inhibited following contusion spinal cord injury (cSCI). However, the mechanism of tumour necrosis factor α (TNF-α) in motor function after cSCI associated with peroxiredoxin 6 (PRDX6) remains unknown. We randomly divided rats into four groups: sham, cSCI, vector and lentivirus mediating TNF-α RNA interference (TNF-α-RNAi-LV) group. The Basso, Beattie, Bresnahan (BBB) scale was used to evaluate motor function. Real-time quantitative PCR (qRT-PCR) and western blotting were used to detect the expression of TNF-α and PRDX6, which were located in neurons using immunohistochemistry (IHC) and immunofluorescence. Subsequently, lentiviral-mediated TNF-α was used to determine the role of TNF-αand the relationship of PRDX6 and TNF-α in cSCI. After cSCI, the motor capability of hind limbs disappeared and was followed by recovery of function. IHC analysis indicated that TNF-α and PRDX6 were primarily located in spinal cord neurons. TNF-α interference significantly improved neural behaviour and increased expression of PRDX6. Our study suggests that inhibition of TNF-α can promote the recovery of motor function. The underlying mechanism of TNF-α-promoted motor function may be connected with the up-regulation of PRDX6. This provides a new strategy or target for the clinical treatment of SCI in future.
Collapse
|
33
|
Strickland ER, Woller SA, Garraway SM, Hook MA, Grau JW, Miranda RC. Regulatory effects of intermittent noxious stimulation on spinal cord injury-sensitive microRNAs and their presumptive targets following spinal cord contusion. Front Neural Circuits 2014; 8:117. [PMID: 25278846 PMCID: PMC4166958 DOI: 10.3389/fncir.2014.00117] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 09/03/2014] [Indexed: 11/13/2022] Open
Abstract
Uncontrollable nociceptive stimulation adversely affects recovery in spinally contused rats. Spinal cord injury (SCI) results in altered microRNA (miRNA) expression both at, and distal to the lesion site. We hypothesized that uncontrollable nociception further influences SCI-sensitive miRNAs and associated gene targets, potentially explaining the progression of maladaptive plasticity. Our data validated previously described sensitivity of miRNAs to SCI alone. Moreover, following SCI, intermittent noxious stimulation decreased expression of miR124 in dorsal spinal cord 24 h after stimulation and increased expression of miR129-2 in dorsal, and miR1 in ventral spinal cord at 7 days. We also found that brain-derived neurotrophic factor (BDNF) mRNA expression was significantly down-regulated 1 day after SCI alone, and significantly more so, after SCI followed by tailshock. Insulin-like growth factor-1 (IGF-1) mRNA expression was significantly increased at both 1 and 7 days post-SCI, and significantly more so, 7 days post-SCI with shock. MiR1 expression was positively and significantly correlated with IGF-1, but not BDNF mRNA expression. Further, stepwise linear regression analysis indicated that a significant proportion of the changes in BDNF and IGF-1 mRNA expression were explained by variance in two groups of miRNAs, implying co-regulation. Collectively, these data show that uncontrollable nociception which activates sensorimotor circuits distal to the injury site, influences SCI-miRNAs and target mRNAs within the lesion site. SCI-sensitive miRNAs may well mediate adverse consequences of uncontrolled sensorimotor activation on functional recovery. However, their sensitivity to distal sensory input also implicates these miRNAs as candidate targets for the management of SCI and neuropathic pain.
Collapse
Affiliation(s)
- Eric R Strickland
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center Bryan, TX, USA
| | - Sarah A Woller
- Department of Psychology, Texas A&M University, College Station TX, USA
| | - Sandra M Garraway
- Department of Psychology, Texas A&M University, College Station TX, USA
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center Bryan, TX, USA
| | - James W Grau
- Department of Psychology, Texas A&M University, College Station TX, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center Bryan, TX, USA
| |
Collapse
|
34
|
Grau JW, Huie JR, Lee KH, Hoy KC, Huang YJ, Turtle JD, Strain MM, Baumbauer KM, Miranda RM, Hook MA, Ferguson AR, Garraway SM. Metaplasticity and behavior: how training and inflammation affect plastic potential within the spinal cord and recovery after injury. Front Neural Circuits 2014; 8:100. [PMID: 25249941 PMCID: PMC4157609 DOI: 10.3389/fncir.2014.00100] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/31/2014] [Indexed: 12/30/2022] Open
Abstract
Research has shown that spinal circuits have the capacity to adapt in response to training, nociceptive stimulation and peripheral inflammation. These changes in neural function are mediated by physiological and neurochemical systems analogous to those that support plasticity within the hippocampus (e.g., long-term potentiation and the NMDA receptor). As observed in the hippocampus, engaging spinal circuits can have a lasting impact on plastic potential, enabling or inhibiting the capacity to learn. These effects are related to the concept of metaplasticity. Behavioral paradigms are described that induce metaplastic effects within the spinal cord. Uncontrollable/unpredictable stimulation, and peripheral inflammation, induce a form of maladaptive plasticity that inhibits spinal learning. Conversely, exposure to controllable or predictable stimulation engages a form of adaptive plasticity that counters these maladaptive effects and enables learning. Adaptive plasticity is tied to an up-regulation of brain derived neurotrophic factor (BDNF). Maladaptive plasticity is linked to processes that involve kappa opioids, the metabotropic glutamate (mGlu) receptor, glia, and the cytokine tumor necrosis factor (TNF). Uncontrollable nociceptive stimulation also impairs recovery after a spinal contusion injury and fosters the development of pain (allodynia). These adverse effects are related to an up-regulation of TNF and a down-regulation of BDNF and its receptor (TrkB). In the absence of injury, brain systems quell the sensitization of spinal circuits through descending serotonergic fibers and the serotonin 1A (5HT 1A) receptor. This protective effect is blocked by surgical anesthesia. Disconnected from the brain, intracellular Cl- concentrations increase (due to a down-regulation of the cotransporter KCC2), which causes GABA to have an excitatory effect. It is suggested that BDNF has a restorative effect because it up-regulates KCC2 and re-establishes GABA-mediated inhibition.
Collapse
Affiliation(s)
- James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - J Russell Huie
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | - Kuan H Lee
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Kevin C Hoy
- Department of Neurosciences, MetroHealth Medical Center and Case Western Reserve University Cleveland, OH, USA
| | - Yung-Jen Huang
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Joel D Turtle
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Misty M Strain
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | | | - Rajesh M Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center Bryan, TX, USA
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center Bryan, TX, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine Atlanta, GA, USA
| |
Collapse
|
35
|
Garraway SM, Woller SA, Huie JR, Hartman JJ, Hook MA, Miranda RC, Huang YJ, Ferguson AR, Grau JW. Peripheral noxious stimulation reduces withdrawal threshold to mechanical stimuli after spinal cord injury: role of tumor necrosis factor alpha and apoptosis. Pain 2014; 155:2344-59. [PMID: 25180012 DOI: 10.1016/j.pain.2014.08.034] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/12/2014] [Accepted: 08/26/2014] [Indexed: 01/23/2023]
Abstract
We previously showed that peripheral noxious input after spinal cord injury (SCI) inhibits beneficial spinal plasticity and impairs recovery of locomotor and bladder functions. These observations suggest that noxious input may similarly affect the development and maintenance of chronic neuropathic pain, an important consequence of SCI. In adult rats with a moderate contusion SCI, we investigated the effect of noxious tail stimulation, administered 1 day after SCI on mechanical withdrawal responses to von Frey stimuli from 1 to 28 days after treatment. In addition, because the proinflammatory cytokine tumor necrosis factor alpha (TNFα) is implicated in numerous injury-induced processes including pain hypersensitivity, we assessed the temporal and spatial expression of TNFα, TNF receptors, and several downstream signaling targets after stimulation. Our results showed that unlike sham surgery or SCI only, nociceptive stimulation after SCI induced mechanical sensitivity by 24h. These behavioral changes were accompanied by increased expression of TNFα. Cellular assessments of downstream targets of TNFα revealed that nociceptive stimulation increased the expression of caspase 8 and the active subunit (12 kDa) of caspase 3, indicative of active apoptosis at a time point consistent with the onset of mechanical allodynia. In addition, immunohistochemical analysis revealed distinct morphological signs of apoptosis in neurons and microglia at 24h after stimulation. Interestingly, expression of the inflammatory mediator NFκB was unaltered by nociceptive stimulation. These results suggest that noxious input caudal to the level of SCI can increase the onset and expression of behavioral responses indicative of pain, potentially involving TNFα signaling.
Collapse
Affiliation(s)
- Sandra M Garraway
- Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - Sarah A Woller
- Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - J Russell Huie
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, 1001 Potrero Ave, Bldg 1, Room 101, San Francisco, CA 94110, USA
| | - John J Hartman
- Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Michelle A Hook
- Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Medical Research and Education Bldg, 8447 State Highway 47, Bryan, TX 77807-3260, USA
| | - Yung-Jen Huang
- Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, 1001 Potrero Ave, Bldg 1, Room 101, San Francisco, CA 94110, USA
| | - James W Grau
- Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
36
|
Distribution and phenotype of TrkB oligodendrocyte lineage cells in the adult rat spinal cord. Brain Res 2014; 1582:21-33. [PMID: 25072185 DOI: 10.1016/j.brainres.2014.07.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
The distribution and phenotype of a previously undescribed population of nonneuronal cells in the intact spinal cord that expresses TrkB, the cognate receptor for brain derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4), were characterized by examining the extent of co-localization of TrkB with NG2, which identifies oligodendrocyte progenitors (OPCs) or CC1, a marker for mature oligodendrocytes (OLs). All TrkB nonneuronal cells expressed Olig2, confirming their role in the OL lineage. Similar to OPCs and OLs, TrkB cells resided in gray and white matter of the spinal cord in similar abundance. Less than 2% of TrkB cells expressed NG2, while over 80% of TrkB cells in the adult spinal cord co-expressed CC1. Most OPCs did not express detectable levels of TrkB, however a small OPC pool (~5%) showed TrkB immunoreactivity. The majority of mature OLs (~65%) expressed TrkB, but a population of mature OLs (~36%) did not express TrkB at detectable levels, and 17% of TrkB nonneuronal cells did not express NG2 or CC1. Approximately 20% of the TrkB nonneuronal population in the ventral horn resided in close proximity to motor neurons and were categorized as perineuronal. We conclude that TrkB is expressed by several pools of OL lineage cells in the adult spinal cord. These findings are important in understanding the neurotrophin regulation of OL lineage cells in the adult spinal cord.
Collapse
|
37
|
Boyce VS, Mendell LM. Neurotrophins and spinal circuit function. Front Neural Circuits 2014; 8:59. [PMID: 24926235 PMCID: PMC4046666 DOI: 10.3389/fncir.2014.00059] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 05/19/2014] [Indexed: 01/19/2023] Open
Abstract
Work early in the last century emphasized the stereotyped activity of spinal circuits based on studies of reflexes. However, the last several decades have focused on the plasticity of these spinal circuits. These considerations began with studies of the effects of monoamines on descending and reflex circuits. In recent years new classes of compounds called growth factors that are found in peripheral nerves and the spinal cord have been shown to affect circuit behavior in the spinal cord. In this review we will focus on the effects of neurotrophins, particularly nerve growth factor (NGF), brain derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), on spinal circuits. We also discuss evidence that these molecules can modify functions including nociceptive behavior, motor reflexes and stepping behavior. Since these substances and their receptors are normally present in the spinal cord, they could potentially be useful in improving function in disease states and after injury. Here we review recent findings relevant to these translational issues.
Collapse
Affiliation(s)
- Vanessa S Boyce
- Department of Neurobiology and Behavior, Stony Brook University Stony Brook, NY, USA
| | - Lorne M Mendell
- Department of Neurobiology and Behavior, Stony Brook University Stony Brook, NY, USA
| |
Collapse
|
38
|
Luedtke K, Bouchard SM, Woller SA, Funk MK, Aceves M, Hook MA. Assessment of depression in a rodent model of spinal cord injury. J Neurotrauma 2014; 31:1107-21. [PMID: 24564232 DOI: 10.1089/neu.2013.3204] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Despite an increased incidence of depression in patients after spinal cord injury (SCI), there is no animal model of depression after SCI. To address this, we used a battery of established tests to assess depression after a rodent contusion injury. Subjects were acclimated to the tasks, and baseline scores were collected before SCI. Testing was conducted on days 9-10 (acute) and 19-20 (chronic) postinjury. To categorize depression, subjects' scores on each behavioral measure were averaged across the acute and chronic stages of injury and subjected to a principal component analysis. This analysis revealed a two-component structure, which explained 72.2% of between-subjects variance. The data were then analyzed with a hierarchical cluster analysis, identifying two clusters that differed significantly on the sucrose preference, open field, social exploration, and burrowing tasks. One cluster (9 of 26 subjects) displayed characteristics of depression. Using these data, a discriminant function analysis was conducted to derive an equation that could classify subjects as "depressed" on days 9-10. The discriminant function was used in a second experiment examining whether the depression-like symptoms could be reversed with the antidepressant, fluoxetine. Fluoxetine significantly decreased immobility in the forced swim test (FST) in depressed subjects identified with the equation. Subjects that were depressed and treated with saline displayed significantly increased immobility on the FST, relative to not depressed, saline-treated controls. These initial experiments validate our tests of depression, generating a powerful model system for further understanding the relationships between molecular changes induced by SCI and the development of depression.
Collapse
Affiliation(s)
- Kelsey Luedtke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | | | | | | | | | | |
Collapse
|
39
|
Learning from the spinal cord: how the study of spinal cord plasticity informs our view of learning. Neurobiol Learn Mem 2013; 108:155-71. [PMID: 23973905 DOI: 10.1016/j.nlm.2013.08.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 08/01/2013] [Accepted: 08/07/2013] [Indexed: 01/10/2023]
Abstract
The paper reviews research examining whether and how training can induce a lasting change in spinal cord function. A framework for the study of learning, and some essential issues in experimental design, are discussed. A core element involves delayed assessment under common conditions. Research has shown that brain systems can induce a lasting (memory-like) alteration in spinal function. Neurons within the lower (lumbosacral) spinal cord can also adapt when isolated from the brain by means of a thoracic transection. Using traditional learning paradigms, evidence suggests that spinal neurons support habituation and sensitization as well as Pavlovian and instrumental conditioning. At a neurobiological level, spinal systems support phenomena (e.g., long-term potentiation), and involve mechanisms (e.g., NMDA mediated plasticity, protein synthesis) implicated in brain-dependent learning and memory. Spinal learning also induces modulatory effects that alter the capacity for learning. Uncontrollable/unpredictable stimulation disables the capacity for instrumental learning and this effect has been linked to the cytokine tumor necrosis factor (TNF). Predictable/controllable stimulation enables learning and counters the adverse effects of uncontrollable stimulation through a process that depends upon brain-derived neurotrophic factor (BDNF). Finally, uncontrollable, but not controllable, nociceptive stimulation impairs recovery after a contusion injury. A process-oriented approach (neurofunctionalism) is outlined that encourages a broader view of learning phenomena.
Collapse
|
40
|
Coulibaly AP, Gannon SM, Hawk K, Walsh BF, Isaacson LG. Transection of preganglionic axons leads to CNS neuronal plasticity followed by survival and target reinnervation. Auton Neurosci 2013; 179:49-59. [PMID: 23891533 DOI: 10.1016/j.autneu.2013.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/17/2013] [Accepted: 07/04/2013] [Indexed: 10/26/2022]
Abstract
The goals of the present study were to investigate the changes in sympathetic preganglionic neurons following transection of distal axons in the cervical sympathetic trunk (CST) that innervate the superior cervical ganglion (SCG) and to assess changes in the protein expression of brain derived neurotrophic factor (BDNF) and its receptor TrkB in the thoracic spinal cord. At 1 week, a significant decrease in soma volume and reduced soma expression of choline acetyltransferase (ChAT) in the intermediolateral cell column (IML) of T1 spinal cord were observed, with both ChAT-ir and non-immunoreactive neurons expressing the injury marker activating transcription factor 3. These changes were transient, and at later time points, ChAT expression and soma volume returned to control values and the number of ATF3 neurons declined. No evidence for cell loss or neuronal apoptosis was detected at any time point. Protein levels of BDNF and/or full length TrkB in the spinal cord were increased throughout the survival period. In the SCG, both ChAT-ir axons and ChAT protein remained decreased at 16 weeks, but were increased compared to the 10 week time point. These results suggest that though IML neurons show reduced ChAT expression and cell volume at 1 week following CST transection, at later time points, the neurons recovered and exhibited no significant signs of neurodegeneration. The alterations in BDNF and/or TrkB may have contributed to the survival of the IML neurons and the recovery of ChAT expression, as well as to the reinnervation of the SCG.
Collapse
Affiliation(s)
- Aminata P Coulibaly
- Center for Neuroscience and Behavior, Miami University, Oxford, OH 45056, United States; Graduate Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, United States
| | | | | | | | | |
Collapse
|
41
|
Grau JW, Huie JR, Garraway SM, Hook MA, Crown ED, Baumbauer KM, Lee KH, Hoy KC, Ferguson AR. Impact of behavioral control on the processing of nociceptive stimulation. Front Physiol 2012; 3:262. [PMID: 22934018 PMCID: PMC3429038 DOI: 10.3389/fphys.2012.00262] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/23/2012] [Indexed: 12/24/2022] Open
Abstract
How nociceptive signals are processed within the spinal cord, and whether these signals lead to behavioral signs of neuropathic pain, depends upon their relation to other events and behavior. Our work shows that these relations can have a lasting effect on spinal plasticity, inducing a form of learning that alters the effect of subsequent nociceptive stimuli. The capacity of lower spinal systems to adapt, in the absence of brain input, is examined in spinally transected rats that receive a nociceptive shock to the tibialis anterior muscle of one hind leg. If shock is delivered whenever the leg is extended (controllable stimulation), it induces an increase in flexion duration that minimizes net shock exposure. This learning is not observed in subjects that receive the same amount of shock independent of leg position (uncontrollable stimulation). These two forms of stimulation have a lasting, and divergent, effect on subsequent learning: controllable stimulation enables learning whereas uncontrollable stimulation disables it (learning deficit). Uncontrollable stimulation also enhances mechanical reactivity. We review evidence that training with controllable stimulation engages a brain-derived neurotrophic factor (BDNF)-dependent process that can both prevent and reverse the consequences of uncontrollable shock. We relate these effects to changes in BDNF protein and TrkB signaling. Controllable stimulation is also shown to counter the effects of peripheral inflammation (from intradermal capsaicin). A model is proposed that assumes nociceptive input is gated at an early sensory stage. This gate is sensitive to current environmental relations (between proprioceptive and nociceptive input), allowing stimulation to be classified as controllable or uncontrollable. We further propose that the status of this gate is affected by past experience and that a history of uncontrollable stimulation will promote the development of neuropathic pain.
Collapse
Affiliation(s)
- James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University College Station, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee IN, Lin MHC, Chung CY, Lee MH, Weng HH, Yang JT. Chronic cigarette smoke exposure enhances brain-derived neurotrophic factor expression in rats with traumatic brain injury. Metab Brain Dis 2012; 27:197-204. [PMID: 22476923 DOI: 10.1007/s11011-012-9294-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 03/15/2012] [Indexed: 01/12/2023]
Abstract
The involvement of brain-derived neurotrophic factor (BDNF) in regulating neuronal survival during neuron differentiation, growth, and maturation, and during the regeneration of injured nerve cells, has already been documented. In experimental Parkinson's disease, chronic exposure to cigarette smoke increased BDNF levels and survival of dopaminergic neurons. BDNF is also elevated in traumatic brain injury (TBI), where it is potentially involved in post-injury repair and regeneration. The aim of this study was to investigate the effects of chronic exposure to cigarette smoke on BDNF expression and apoptosis in rats with TBI. Three groups of rats were compared: rats with TBI after chronic exposure to cigarette smoke, rats with TBI and no exposure to cigarette smoke, and sham-operated rats. BDNF mRNA expression in the hippocampus increased from 2 to 24 h after TBI, and chronic exposure to cigarette smoke upregulated TBI-induced BDNF mRNA elevation at 0, 2, 4, 12, and 24 h after head injury. The BDNF protein levels generally corresponded to the mRNA levels in the hippocampal region. Compared to the TBI group without smoke exposure, chronic cigarette smoke exposure in rats inhibited the decrease of the Bcl-2/Bax ratio and reduced P53 expression and apoptosis 24 h after TBI. In addition, neuronal damage in the parietal and cingulate cortex 7 days after TBI was less extensive in rats exposed to cigarette smoke. In conclusion, although chronic exposure to cigarette smoke is a risk factor for myocardial and pulmonary disease, cigarette smoke exposure increases BDNF expression after TBI and thereby can play a neuroprotective role.
Collapse
Affiliation(s)
- I-Neng Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Pu Tz City, Chia-Yi, Taiwan
| | | | | | | | | | | |
Collapse
|