1
|
Tashiro S, Shibata S, Nagoshi N, Zhang L, Yamada S, Tsuji T, Nakamura M, Okano H. Do Pharmacological Treatments Act in Collaboration with Rehabilitation in Spinal Cord Injury Treatment? A Review of Preclinical Studies. Cells 2024; 13:412. [PMID: 38474376 PMCID: PMC10931131 DOI: 10.3390/cells13050412] [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: 01/17/2024] [Revised: 02/18/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
There is no choice other than rehabilitation as a practical medical treatment to restore impairments or improve activities after acute treatment in people with spinal cord injury (SCI); however, the effect is unremarkable. Therefore, researchers have been seeking effective pharmacological treatments. These will, hopefully, exert a greater effect when combined with rehabilitation. However, no review has specifically summarized the combinatorial effects of rehabilitation with various medical agents. In the current review, which included 43 articles, we summarized the combinatorial effects according to the properties of the medical agents, namely neuromodulation, neurotrophic factors, counteraction to inhibitory factors, and others. The recovery processes promoted by rehabilitation include the regeneration of tracts, neuroprotection, scar tissue reorganization, plasticity of spinal circuits, microenvironmental change in the spinal cord, and enforcement of the musculoskeletal system, which are additive, complementary, or even synergistic with medication in many cases. However, there are some cases that lack interaction or even demonstrate competition between medication and rehabilitation. A large fraction of the combinatorial mechanisms remains to be elucidated, and very few studies have investigated complex combinations of these agents or targeted chronically injured spinal cords.
Collapse
Affiliation(s)
- Syoichi Tashiro
- Department of Rehabilitation Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
- Department of Rehabilitation Medicine, Faculty of Medicine, Kyorin University, Tokyo 181-8611, Japan
| | - Shinsuke Shibata
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Liang Zhang
- Department of Rehabilitation Medicine, Faculty of Medicine, Kyorin University, Tokyo 181-8611, Japan
| | - Shin Yamada
- Department of Rehabilitation Medicine, Faculty of Medicine, Kyorin University, Tokyo 181-8611, Japan
| | - Tetsuya Tsuji
- Department of Rehabilitation Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Tokyo 160-8582, Japan
| |
Collapse
|
2
|
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
|
3
|
Grau JW, Hudson KE, Tarbet MM, Strain MM. Behavioral studies of spinal conditioning: The spinal cord is smarter than you think it is. JOURNAL OF EXPERIMENTAL PSYCHOLOGY. ANIMAL LEARNING AND COGNITION 2022; 48:435-457. [PMID: 35901417 PMCID: PMC10391333 DOI: 10.1037/xan0000332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In 1988 Robert Rescorla published an article in the Annual Review of Neuroscience that addressed the circumstances under which learning occurs, some key methodological issues, and what constitutes an example of learning. The article has inspired a generation of neuroscientists, opening the door to a wider range of learning phenomena. After reviewing the historical context for his article, its key points are briefly reviewed. The perspective outlined enabled the study of learning in simpler preparations, such as the spinal cord. The period after 1988 revealed that pain (nociceptive) stimuli can induce a lasting sensitization of spinal cord circuits, laying down a kind of memory mediated by signal pathways analogous to those implicated in brain dependent learning and memory. Evidence suggests that the spinal cord is sensitive to instrumental response-outcome (R-O) relations, that learning can induce a peripheral modification (muscle memory) that helps maintain the learned response, and that learning can promote adaptive plasticity (a form of metaplasticity). Conversely, exposure to uncontrollable stimulation disables the capacity to learn. Spinal cord neurons can also abstract that stimuli occur in a regular (predictable) manner, a capacity that appears linked to a neural oscillator (central pattern generator). Disrupting communication with the brain has been shown to transform how GABA affects neuronal function (an example of ionic plasticity), releasing a brake that enables plasticity. We conclude by presenting a framework for understanding these findings and the implications for the broader study of learning. (PsycInfo Database Record (c) 2022 APA, all rights reserved).
Collapse
Affiliation(s)
- James W. Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX, 77843 USA
| | - Kelsey E. Hudson
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX, 77843 USA
| | - Megan M. Tarbet
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Misty M. Strain
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX 78229
| |
Collapse
|
4
|
Hudson KE, Grau JW. Ionic Plasticity: Common Mechanistic Underpinnings of Pathology in Spinal Cord Injury and the Brain. Cells 2022; 11:2910. [PMID: 36139484 PMCID: PMC9496934 DOI: 10.3390/cells11182910] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
The neurotransmitter GABA is normally characterized as having an inhibitory effect on neural activity in the adult central nervous system (CNS), which quells over-excitation and limits neural plasticity. Spinal cord injury (SCI) can bring about a modification that weakens the inhibitory effect of GABA in the central gray caudal to injury. This change is linked to the downregulation of the potassium/chloride cotransporter (KCC2) and the consequent rise in intracellular Cl- in the postsynaptic neuron. As the intracellular concentration increases, the inward flow of Cl- through an ionotropic GABA-A receptor is reduced, which decreases its hyperpolarizing (inhibitory) effect, a modulatory effect known as ionic plasticity. The loss of GABA-dependent inhibition enables a state of over-excitation within the spinal cord that fosters aberrant motor activity (spasticity) and chronic pain. A downregulation of KCC2 also contributes to the development of a number of brain-dependent pathologies linked to states of neural over-excitation, including epilepsy, addiction, and developmental disorders, along with other diseases such as hypertension, asthma, and irritable bowel syndrome. Pharmacological treatments that target ionic plasticity have been shown to bring therapeutic benefits.
Collapse
Affiliation(s)
- Kelsey E. Hudson
- Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - James W. Grau
- Psychological & Brain Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
5
|
Freyermuth-Trujillo X, Segura-Uribe JJ, Salgado-Ceballos H, Orozco-Barrios CE, Coyoy-Salgado A. Inflammation: A Target for Treatment in Spinal Cord Injury. Cells 2022; 11:2692. [PMID: 36078099 PMCID: PMC9454769 DOI: 10.3390/cells11172692] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/16/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a significant cause of disability, and treatment alternatives that generate beneficial outcomes and have no side effects are urgently needed. SCI may be treatable if intervention is initiated promptly. Therefore, several treatment proposals are currently being evaluated. Inflammation is part of a complex physiological response to injury or harmful stimuli induced by mechanical, chemical, or immunological agents. Neuroinflammation is one of the principal secondary changes following SCI and plays a crucial role in modulating the pathological progression of acute and chronic SCI. This review describes the main inflammatory events occurring after SCI and discusses recently proposed potential treatments and therapeutic agents that regulate inflammation after insult in animal models.
Collapse
Affiliation(s)
- Ximena Freyermuth-Trujillo
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico
| | - Julia J. Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City CP 06720, Mexico
| | - Hermelinda Salgado-Ceballos
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| | - Carlos E. Orozco-Barrios
- CONACyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| | - Angélica Coyoy-Salgado
- CONACyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| |
Collapse
|
6
|
Fauss GNK, Hudson KE, Grau JW. Role of Descending Serotonergic Fibers in the Development of Pathophysiology after Spinal Cord Injury (SCI): Contribution to Chronic Pain, Spasticity, and Autonomic Dysreflexia. BIOLOGY 2022; 11:234. [PMID: 35205100 PMCID: PMC8869318 DOI: 10.3390/biology11020234] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 12/12/2022]
Abstract
As the nervous system develops, nerve fibers from the brain form descending tracts that regulate the execution of motor behavior within the spinal cord, incoming sensory signals, and capacity to change (plasticity). How these fibers affect function depends upon the transmitter released, the receptor system engaged, and the pattern of neural innervation. The current review focuses upon the neurotransmitter serotonin (5-HT) and its capacity to dampen (inhibit) neural excitation. A brief review of key anatomical details, receptor types, and pharmacology is provided. The paper then considers how damage to descending serotonergic fibers contributes to pathophysiology after spinal cord injury (SCI). The loss of serotonergic fibers removes an inhibitory brake that enables plasticity and neural excitation. In this state, noxious stimulation can induce a form of over-excitation that sensitizes pain (nociceptive) circuits, a modification that can contribute to the development of chronic pain. Over time, the loss of serotonergic fibers allows prolonged motor drive (spasticity) to develop and removes a regulatory brake on autonomic function, which enables bouts of unregulated sympathetic activity (autonomic dysreflexia). Recent research has shown that the loss of descending serotonergic activity is accompanied by a shift in how the neurotransmitter GABA affects neural activity, reducing its inhibitory effect. Treatments that target the loss of inhibition could have therapeutic benefit.
Collapse
Affiliation(s)
| | | | - James W. Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77843, USA; (G.N.K.F.); (K.E.H.)
| |
Collapse
|
7
|
Baine RE, Johnston DT, Strain MM, Henwood MK, Davis JA, Reynolds JA, Giles ED, Grau JW. Noxious Stimulation Induces Acute Hemorrhage and Impairs Long-Term Recovery after Spinal Cord Injury (SCI) in Female Rats: Evidence Estrous Cycle May Have a Modulatory Effect. Neurotrauma Rep 2022; 3:70-86. [PMID: 35112109 PMCID: PMC8804264 DOI: 10.1089/neur.2021.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Spinal cord injuries (SCIs) are often the result of traumatic accidents, which also produce multiple other injuries (polytrauma). Nociceptive input from associated injuries has been shown to significantly impair recovery post-SCI. Historically, work in our laboratory has focused exclusively on male animals; however, increasing incidence of SCI in females requires research to determine whether pain (nociceptive) input poses the same risk to their recovery. Some animal studies have shown that females demonstrate greater tissue preservation and better locomotor recovery post-SCI. Given this, we examined the effect of sex on SCI recovery in two pain models—intermittent electrical stimulation (shock) to the tail or capsaicin injection to the hindpaw. Female rats received a lower thoracic contusion injury and were exposed to noxious stimulation the next day. The acute effect of noxious input on cardiovascular function, locomotor performance, and hemorrhage were assessed. Treatment with capsaicin or noxious electrical stimulation disrupted locomotor performance, increased blood pressure, and disrupted stepping. Additional experiments examined the long-term consequences of noxious input, demonstrating that both noxious electrical stimulation and capsaicin impair long-term recovery in female rats. Interestingly, injury had a greater effect on behavioral performance when progesterone and estrogen were low (metestrus). Conversely, nociceptive input led to a greater disruption in locomotor performance and produced a greater rise in blood pressure in animals injured during estrus.
Collapse
Affiliation(s)
- Rachel E. Baine
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - David T. Johnston
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - Misty M. Strain
- Department of Cellular and Integrative Physiology, University of Texas Health Science, San Antonio, Texas, USA
| | - Melissa K. Henwood
- Department of Neuroscience, Cell Biology, Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jacob A. Davis
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - Joshua A. Reynolds
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - Erin D. Giles
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - James W. Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
8
|
Begenisic T, Pavese C, Aiachini B, Nardone A, Rossi D. Dynamics of biomarkers across the stages of traumatic spinal cord injury - implications for neural plasticity and repair. Restor Neurol Neurosci 2021; 39:339-366. [PMID: 34657853 DOI: 10.3233/rnn-211169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) is a complex medical condition causing significant physical disability and psychological distress. While the adult spinal cord is characterized by poor regenerative potential, some recovery of neurological function is still possible through activation of neural plasticity mechanisms. We still have limited knowledge about the activation of these mechanisms in the different stages after human SCI. OBJECTIVE In this review, we discuss the potential role of biomarkers of SCI as indicators of the plasticity mechanisms at work during the different phases of SCI. METHODS An extensive review of literature related to SCI pathophysiology, neural plasticity and humoral biomarkers was conducted by consulting the PubMed database. Research and review articles from SCI animal models and SCI clinical trials published in English until January 2021 were reviewed. The selection of candidates for humoral biomarkers of plasticity after SCI was based on the following criteria: 1) strong evidence supporting involvement in neural plasticity (mandatory); 2) evidence supporting altered expression after SCI (optional). RESULTS Based on selected findings, we identified two main groups of potential humoral biomarkers of neural plasticity after SCI: 1) neurotrophic factors including: Brain derived neurotrophic factor (BDNF), Nerve growth factor (NGF), Neurotrofin-3 (NT-3), and Insulin-like growth factor 1 (IGF-1); 2) other factors including: Tumor necrosis factor-alpha (TNF-α), Matrix Metalloproteinases (MMPs), and MicroRNAs (miRNAs). Plasticity changes associated with these biomarkers often can be both adaptive (promoting functional improvement) and maladaptive. This dual role seems to be influenced by their concentrations and time-window during SCI. CONCLUSIONS Further studies of dynamics of biomarkers across the stages of SCI are necessary to elucidate the way in which they reflect the remodeling of neural pathways. A better knowledge about the mechanisms underlying plasticity could guide the selection of more appropriate therapeutic strategies to enhance positive spinal network reorganization.
Collapse
Affiliation(s)
- Tatjana Begenisic
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Chiara Pavese
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.,Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Beatrice Aiachini
- Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Antonio Nardone
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.,Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| |
Collapse
|
9
|
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
|
10
|
Huie JR, Ferguson AR, Kyritsis N, Pan JZ, Irvine KA, Nielson JL, Schupp PG, Oldham MC, Gensel JC, Lin A, Segal MR, Ratan RR, Bresnahan JC, Beattie MS. Machine intelligence identifies soluble TNFa as a therapeutic target for spinal cord injury. Sci Rep 2021; 11:3442. [PMID: 33564058 PMCID: PMC7873211 DOI: 10.1038/s41598-021-82951-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic spinal cord injury (SCI) produces a complex syndrome that is expressed across multiple endpoints ranging from molecular and cellular changes to functional behavioral deficits. Effective therapeutic strategies for CNS injury are therefore likely to manifest multi-factorial effects across a broad range of biological and functional outcome measures. Thus, multivariate analytic approaches are needed to capture the linkage between biological and neurobehavioral outcomes. Injury-induced neuroinflammation (NI) presents a particularly challenging therapeutic target, since NI is involved in both degeneration and repair. Here, we used big-data integration and large-scale analytics to examine a large dataset of preclinical efficacy tests combining five different blinded, fully counter-balanced treatment trials for different acute anti-inflammatory treatments for cervical spinal cord injury in rats. Multi-dimensional discovery, using topological data analysis (TDA) and principal components analysis (PCA) revealed that only one showed consistent multidimensional syndromic benefit: intrathecal application of recombinant soluble TNFα receptor 1 (sTNFR1), which showed an inverse-U dose response efficacy. Using the optimal acute dose, we showed that clinically-relevant 90 min delayed treatment profoundly affected multiple biological indices of NI in the first 48 h after injury, including reduction in pro-inflammatory cytokines and gene expression of a coherent complex of acute inflammatory mediators and receptors. Further, a 90 min delayed bolus dose of sTNFR1 reduced the expression of NI markers in the chronic perilesional spinal cord, and consistently improved neurological function over 6 weeks post SCI. These results provide validation of a novel strategy for precision preclinical drug discovery that is likely to improve translation in the difficult landscape of CNS trauma, and confirm the importance of TNFα signaling as a therapeutic target.
Collapse
Affiliation(s)
- J R Huie
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, CA, USA
| | - A R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, CA, USA.
- San Francisco Veterans Affairs Medical Center, San Francisco, USA.
| | - N Kyritsis
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, CA, USA
| | - J Z Pan
- Department of Anesthesiology, University of California San Francisco, San Francisco, USA
| | - K-A Irvine
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Anesthesia, Perioperative Medicine and Pain, Stanford University, Stanford, CA, USA
| | - J L Nielson
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, USA
- Institute for Health Informatics, University of Minnesota, Minneapolis, USA
| | - P G Schupp
- Brain Tumor Research Center, University of California, San Francisco, USA
| | - M C Oldham
- Brain Tumor Research Center, University of California, San Francisco, USA
| | - J C Gensel
- SCoBIRC, University of Kentucky, Lexington, USA
| | - A Lin
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, CA, USA
| | - M R Segal
- Department of Epidemiology and Biostatistics, Center for Bioinformatics and Molecular Biostatistics, University of California San Francisco, San Francisco, USA
| | - R R Ratan
- Department of Neurology and Neuroscience, Burke-Cornell Medical Research Institute, Weill Medical College of Cornell University, New York, USA
| | - J C Bresnahan
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, CA, USA
| | - M S Beattie
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), University of California, San Francisco, CA, USA.
| |
Collapse
|
11
|
Hoy KC, Strain MM, Turtle JD, Lee KH, Huie JR, Hartman JJ, Tarbet MM, Harlow ML, Magnuson DSK, Grau JW. Evidence That the Central Nervous System Can Induce a Modification at the Neuromuscular Junction That Contributes to the Maintenance of a Behavioral Response. J Neurosci 2020; 40:9186-9209. [PMID: 33097637 PMCID: PMC7687054 DOI: 10.1523/jneurosci.2683-19.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 11/21/2022] Open
Abstract
Neurons within the spinal cord are sensitive to environmental relations and can bring about a behavioral modification without input from the brain. For example, rats that have undergone a thoracic (T2) transection can learn to maintain a hind leg in a flexed position to minimize exposure to a noxious electrical stimulation (shock). Inactivating neurons within the spinal cord with lidocaine, or cutting communication between the spinal cord and the periphery (sciatic transection), eliminates the capacity to learn, which implies that it depends on spinal neurons. Here we show that these manipulations have no effect on the maintenance of the learned response, which implicates a peripheral process. EMG showed that learning augments the muscular response evoked by motoneuron output and that this effect survives a sciatic transection. Quantitative fluorescent imaging revealed that training brings about an increase in the area and intensity of ACh receptor labeling at the neuromuscular junction (NMJ). It is hypothesized that efferent motoneuron output, in conjunction with electrical stimulation of the tibialis anterior muscle, strengthens the connection at the NMJ in a Hebbian manner. Supporting this, paired stimulation of the efferent nerve and tibialis anterior generated an increase in flexion duration and augmented the evoked electrical response without input from the spinal cord. Evidence is presented that glutamatergic signaling contributes to plasticity at the NMJ. Labeling for vesicular glutamate transporter is evident at the motor endplate. Intramuscular application of an NMDAR antagonist blocked the acquisition/maintenance of the learned response and the strengthening of the evoked electrical response.SIGNIFICANCE STATEMENT The neuromuscular junction (NMJ) is designed to faithfully elicit a muscular contraction in response to neural input. From this perspective, encoding environmental relations (learning) and the maintenance of a behavioral modification over time (memory) are assumed to reflect only modifications upstream from the NMJ, within the CNS. The current results challenge this view. Rats were trained to maintain a hind leg in a flexed position to avoid noxious stimulation. As expected, treatments that inhibit activity within the CNS, or disrupt peripheral communication, prevented learning. These manipulations did not affect the maintenance of the acquired response. The results imply that a peripheral modification at the NMJ contributes to the maintenance of the learned response.
Collapse
Affiliation(s)
- Kevin C Hoy
- Case Comprehensive Cancer Center/Case Western Reserve School of Medicine, Cleveland, Ohio 44106
| | - Misty M Strain
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, Houston, Texas 78234
| | - Joel D Turtle
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas 77843
| | - Kuan H Lee
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas 77843
| | - J Russell Huie
- Department of Neuroscience, University of California San Francisco, San Francisco, California 94110
| | - John J Hartman
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas 77843
| | - Megan M Tarbet
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas 77843
| | - Mark L Harlow
- Department of Biology, Texas A&M University, College Station, Texas 77843
| | - David S K Magnuson
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky 40202
| | - James W Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
12
|
Grau JW, Baine RE, Bean PA, Davis JA, Fauss GN, Henwood MK, Hudson KE, Johnston DT, Tarbet MM, Strain MM. Learning to promote recovery after spinal cord injury. Exp Neurol 2020; 330:113334. [PMID: 32353465 PMCID: PMC7282951 DOI: 10.1016/j.expneurol.2020.113334] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/19/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023]
Abstract
The present review explores the concept of learning within the context of neurorehabilitation after spinal cord injury (SCI). The aim of physical therapy and neurorehabilitation is to bring about a lasting change in function-to encourage learning. Traditionally, it was assumed that the adult spinal cord is hardwired-immutable and incapable of learning. Research has shown that neurons within the lower (lumbosacral) spinal cord can support learning after communication with the brain has been disrupted by means of a thoracic transection. Noxious stimulation can sensitize nociceptive circuits within the spinal cord, engaging signal pathways analogous to those implicated in brain-dependent learning and memory. After a spinal contusion injury, pain input can fuel hemorrhage, increase the area of tissue loss (secondary injury), and undermine long-term recovery. Neurons within the spinal cord are sensitive to environmental relations. This learning has a metaplastic effect that counters neural over-excitation and promotes adaptive learning through an up-regulation of brain-derived neurotrophic factor (BDNF). Exposure to rhythmic stimulation, treadmill training, and cycling also enhances the expression of BDNF and counters the development of nociceptive sensitization. SCI appears to enable plastic potential within the spinal cord by down-regulating the Cl- co-transporter KCC2, which reduces GABAergic inhibition. This enables learning, but also fuels over-excitation and nociceptive sensitization. Pairing epidural stimulation with activation of motor pathways also promotes recovery after SCI. Stimulating motoneurons in response to activity within the motor cortex, or a targeted muscle, has a similar effect. It is suggested that a neurofunctionalist approach can foster the discovery of processes that impact spinal function and how they may be harnessed to foster recovery after SCI.
Collapse
Affiliation(s)
- James W Grau
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - Rachel E Baine
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Paris A Bean
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Jacob A Davis
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Gizelle N Fauss
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Melissa K Henwood
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Kelsey E Hudson
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - David T Johnston
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Megan M Tarbet
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Misty M Strain
- Battlefield Pain Research, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, BHT-1, BSA Fort Sam Houston, TX 78234, USA
| |
Collapse
|
13
|
Shang P, Zhang Y, Ma D, Hao Y, Wang X, Xin M, Zhang Y, Zhu M, Feng J. Inflammation resolution and specialized pro-resolving lipid mediators in CNS diseases. Expert Opin Ther Targets 2019; 23:967-986. [PMID: 31711309 DOI: 10.1080/14728222.2019.1691525] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Inflammation resolution induced by specialized pro-resolving lipid mediators (SPMs) is a new concept. The application of SPMs is a promising therapeutic strategy that can potentially supersede anti-inflammatory drugs. Most CNS diseases are associated with hyperreactive inflammatory damage. CNS inflammation causes irreversible neuronal loss and permanent functional impairments. Given the high mortality and morbidity rates, the investigation of therapeutic strategies to ameliorate inflammatory damage is necessary.Areas covered: In this review, we explore inflammation resolution in CNS disorders. We discuss the underlying mechanisms and dynamic changes of SPMs and their precursors in neurological diseases and examine how this can potentially be incorporated into the clinic. References were selected from PubMed; most were published between 2010 and 2019.Expert opinion: Inflammation resolution is a natural process that emerges after acute or chronic inflammation. The evidence that SPMs can effectively ameliorate hyperreactive inflammation, shorten resolution time and accelerate tissue regeneration in CNS disorders. Adjuvants and nanotechnology offer opportunities for SPM drug design; however, more preclinical studies are necessary to investigate basic, critical issues such as safety.
Collapse
Affiliation(s)
- Pei Shang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Di Ma
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yulei Hao
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyu Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Meiying Xin
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yunhai Zhang
- Jiangsu Key Laboratory of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Mingqin Zhu
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiachun Feng
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
14
|
Strain MM, Hook MA, Reynolds JD, Huang YJ, Henwood MK, Grau JW. A brief period of moderate noxious stimulation induces hemorrhage and impairs locomotor recovery after spinal cord injury. Physiol Behav 2019; 212:112695. [PMID: 31647990 DOI: 10.1016/j.physbeh.2019.112695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 10/25/2022]
Abstract
Spinal cord injury (SCI) is often accompanied by additional tissue damage (polytrauma) that provides a source of pain input. Our studies suggest that this pain input may be detrimental to long-term recovery. In a rodent model, we have shown that engaging pain (nociceptive) fibers caudal to a lower thoracic contusion SCI impairs recovery of locomotor function and increases tissue loss (secondary injury) and hemorrhage at the site of injury. In these studies, nociceptive fibers were activated using intermittent electrical stimulation. The stimulation parameters were derived from earlier studies demonstrating that 6 min of noxious stimulation, at an intensity (1.5 mA) that engages unmyelinated C (pain) fibers, induces a form of maladaptive plasticity within the lumbosacral spinal cord. We hypothesized that both shorter bouts of nociceptive input and lower intensities of stimulation will decrease locomotor function and increase spinal cord hemorrhage when rats have a spinal cord contusion. To test this, the present study exposed rats to electrical stimulation 24 h after a moderate lower thoracic contusion SCI. One group of rats received 1.5 mA stimulation for 0, 14.4, 72, or 180 s. Another group received six minutes of stimulation at 0, 0.17, 0.5, and 1.5 mA. Just 72 s of stimulation induced an acute disruption in motor performance, increased hemorrhage, and undermined the recovery of locomotor function. Likewise, less intense (0.5 mA) stimulation produced an acute disruption in motor performance, fueled hemorrhage, and impaired long-term recovery. The results imply that a brief period of moderate pain input can trigger hemorrhage after SCI and undermine long-term recovery. This highlights the importance of managing nociceptive signals after concurrent peripheral and central nervous system injuries.
Collapse
Affiliation(s)
- Misty M Strain
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - Michelle A Hook
- Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Joshua D Reynolds
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Yung-Jen Huang
- ChemPartner, 998 Halei Rd., Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai, 201203 China
| | - Melissa K Henwood
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
15
|
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
|
16
|
Ren H, Chen X, Tian M, Zhou J, Ouyang H, Zhang Z. Regulation of Inflammatory Cytokines for Spinal Cord Injury Repair Through Local Delivery of Therapeutic Agents. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800529. [PMID: 30479916 PMCID: PMC6247077 DOI: 10.1002/advs.201800529] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/06/2018] [Indexed: 05/29/2023]
Abstract
The balance of inflammation is critical to the repair of spinal cord injury (SCI), which is one of the most devastating traumas in human beings. Inflammatory cytokines, the direct mediators of local inflammation, have differential influences on the repair of the injured spinal cord. Some inflammatory cytokines are demonstrated beneficial to spinal cord repair in SCI models, while some detrimental. Various animal researches have revealed that local delivery of therapeutic agents efficiently regulates inflammatory cytokines and promotes repair from SCI. Quite a few clinical studies have also shown the promotion of repair from SCI through regulation of inflammatory cytokines. However, local delivery of a single agent affects only a part of the inflammatory cytokines that need to be regulated. Meanwhile, different individuals have differential profiles of inflammatory cytokines. Therefore, future studies may aim to develop personalized strategies of locally delivered therapeutic agent cocktails for effective and precise regulation of inflammation, and substantial functional recovery from SCI.
Collapse
Affiliation(s)
- Hao Ren
- The Third Affiliated Hospital of Guangzhou Medical UniversityNo. 63 Duobao RoadGuangzhou510150P. R. China
| | - Xuri Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Mengya Tian
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Jing Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Zhiyong Zhang
- Translational Research Center for Regenerative Medicine and 3D Printing TechnologiesGuangzhou Medical UniversityNo. 63 Duobao RoadGuangzhou510150P. R. China
| |
Collapse
|
17
|
Juvenile stress leads to long-term immunological metaplasticity-like effects on inflammatory responses in adulthood. Neurobiol Learn Mem 2018; 154:12-21. [DOI: 10.1016/j.nlm.2017.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 09/19/2017] [Accepted: 09/25/2017] [Indexed: 12/11/2022]
|
18
|
Alt EU, Barabadi Z, Pfnür A, Ochoa JE, Daneshimehr F, Lang LM, Lin D, Braun SE, Chandrasekar B, Izadpanah R. TRAF3IP2, a novel therapeutic target in glioblastoma multiforme. Oncotarget 2018; 9:29772-29788. [PMID: 30038719 PMCID: PMC6049871 DOI: 10.18632/oncotarget.25710] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/13/2018] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme (glioblastoma) remains one of the deadliest cancers. Pro-inflammatory and pro-tumorigenic mediators present in tumor microenvironment (TME) facilitate communication between tumor cells and adjacent non-malignant cells, resulting in glioblastoma growth. Since a majority of these mediators are products of NF-κB- and/or AP-1-responsive genes, and as TRAF3 Interacting Protein 2 (TRAF3IP2) is an upstream regulator of both transcription factors, we hypothesized that targeting TRAF3IP2 blunts tumor growth by inhibiting NF-κB and pro-inflammatory/pro-tumorigenic mediators. Our in vitro data demonstrate that similar to primary glioblastoma tumor tissues, malignant glioblastoma cell lines (U87 and U118) express high levels of TRAF3IP2. Silencing TRAF3IP2 expression inhibits basal and inducible NF-κB activation, induction of pro-inflammatory mediators, clusters of genes involved in cell cycle progression and angiogenesis, and formation of spheroids. Additionally, silencing TRAF3IP2 significantly increases apoptosis. In vivo studies indicate TRAF3IP2-silenced U87 cells formed smaller tumors. Additionally, treating existing tumors formed by wild type U87 cells with lentiviral TRAF3IP2 shRNA markedly regresses their size. Analysis of residual tumors revealed reduced expression of pro-inflammatory/pro-tumorigenic/pro-angiogenic mediators and kinesins. In contrast, the expression of IL-10, an anti-inflammatory cytokine, was increased. Together, these novel data indicate that TRAF3IP2 is a master regulator of malignant signaling in glioblastoma, and its targeting modulates the TME and inhibits tumor growth by suppressing the expression of mediators involved in inflammation, angiogenesis, growth, and malignant transformation. Our data identify TRAF3IP2 as a potential therapeutic target in glioblastoma growth and dissemination.
Collapse
Affiliation(s)
- Eckhard U Alt
- Applied Stem Cell Laboratory, Medicine/Heart and Vascular Institute, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Zahra Barabadi
- Applied Stem Cell Laboratory, Medicine/Heart and Vascular Institute, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Andreas Pfnür
- Applied Stem Cell Laboratory, Medicine/Heart and Vascular Institute, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Joana E Ochoa
- Department of Surgery, Tulane University Health Science Center, New Orleans, Louisiana, USA
| | - Fatemeh Daneshimehr
- Applied Stem Cell Laboratory, Medicine/Heart and Vascular Institute, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Lea M Lang
- Applied Stem Cell Laboratory, Medicine/Heart and Vascular Institute, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Dong Lin
- Applied Stem Cell Laboratory, Medicine/Heart and Vascular Institute, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Stephen E Braun
- Division of Regenerative Medicine, Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Bysani Chandrasekar
- Department of Medicine, University of Missouri School of Medicine and Harry S. Truman Veterans Memorial Hospital, Columbia, Missouri, USA
| | - Reza Izadpanah
- Applied Stem Cell Laboratory, Medicine/Heart and Vascular Institute, Tulane University Health Sciences Center, New Orleans, Louisiana, USA.,Department of Surgery, Tulane University Health Science Center, New Orleans, Louisiana, 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
|
Magerl W, Hansen N, Treede RD, Klein T. The human pain system exhibits higher-order plasticity (metaplasticity). Neurobiol Learn Mem 2018; 154:112-120. [PMID: 29631001 DOI: 10.1016/j.nlm.2018.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 02/21/2018] [Accepted: 04/05/2018] [Indexed: 01/10/2023]
Abstract
The human pain system can be bidirectionally modulated by high-frequency (HFS; 100 Hz) and low-frequency (LFS; 1 Hz) electrical stimulation of nociceptors leading to long-term potentiation or depression of pain perception (pain-LTP or pain-LTD). Here we show that priming a test site by very low-frequency stimulation (VLFS; 0.05 Hz) prevented pain-LTP probably by elevating the threshold (set point) for pain-LTP induction. Conversely, prior HFS-induced pain-LTP was substantially reversed by subsequent VLFS, suggesting that preceding HFS had primed the human nociceptive system for pain-LTD induction by VLFS. In contrast, the pain elicited by the pain-LTP-precipitating conditioning HFS stimulation remained unaffected. In aggregate these experiments demonstrate that the human pain system expresses two forms of higher-order plasticity (metaplasticity) acting in either direction along the pain-LTD to pain-LTP continuum with similar shifts in thresholds for LTD and LTP as in synaptic plasticity, indicating intriguing new mechanisms for the prevention of pain memory and the erasure of hyperalgesia related to an already established pain memory trace. There were no apparent gender differences in either pain-LTP or metaplasticity of pain-LTP. However, individual subjects appeared to present with an individual balance of pain-LTD to pain-LTP (a pain plasticity "fingerprint").
Collapse
Affiliation(s)
- Walter Magerl
- Department of Neurophysiology, Center of Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl-University Heidelberg, Ludolf Krehl-Str. 13-17, 68167 Mannheim, Germany.
| | - Niels Hansen
- Department of Neurophysiology, Center of Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl-University Heidelberg, Ludolf Krehl-Str. 13-17, 68167 Mannheim, Germany; Department of Psychiatry and Psychotherapy & Department of Epileptology, University Hospital Bonn, Sigmund-Freud-Straße 25, 53105 Bonn, Germany
| | - Rolf-Detlef Treede
- Department of Neurophysiology, Center of Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl-University Heidelberg, Ludolf Krehl-Str. 13-17, 68167 Mannheim, Germany
| | - Thomas Klein
- Department of Neurophysiology, Center of Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl-University Heidelberg, Ludolf Krehl-Str. 13-17, 68167 Mannheim, Germany
| |
Collapse
|
22
|
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]
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
Huang YJ, Lee KH, Murphy L, Garraway SM, Grau JW. Acute spinal cord injury (SCI) transforms how GABA affects nociceptive sensitization. Exp Neurol 2016; 285:82-95. [PMID: 27639636 PMCID: PMC5926208 DOI: 10.1016/j.expneurol.2016.09.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 10/21/2022]
Abstract
Noxious input can sensitize pain (nociceptive) circuits within the spinal cord, inducing a lasting increase in spinal cord neural excitability (central sensitization) that is thought to contribute to chronic pain. The development of spinally-mediated central sensitization is regulated by descending fibers and GABAergic interneurons. The current study provides evidence that spinal cord injury (SCI) transforms how GABA affects nociceptive transmission within the spinal cord, recapitulating an earlier developmental state wherein GABA has an excitatory effect. In spinally transected rats, noxious electrical stimulation and inflammation induce enhanced mechanical reactivity (EMR), a behavioral index of nociceptive sensitization. Pretreatment with the GABAA receptor antagonist bicuculline blocked these effects. Peripheral application of an irritant (capsaicin) also induced EMR. Both the induction and maintenance of this effect were blocked by bicuculline. Cellular indices of central sensitization [c-fos expression and ERK phosphorylation (pERK)] were also attenuated. In intact (sham operated) rats, bicuculline had the opposite effect. Pretreatment with a GABA agonist (muscimol) attenuated nociceptive sensitization in intact, but not spinally injured, rats. The effect of SCI on GABA function was linked to a reduction in the Cl- transporter, KCC2, leading to a reduction in intracellular Cl- that would attenuate GABA-mediated inhibition. Pharmacologically blocking the KCC2 channel (with i.t. DIOA) in intact rats mimicked the effect of SCI. Conversely, a pharmacological treatment (bumetanide) that should increase intracellular Cl- levels blocked the effect of SCI. The results suggest that GABAergic neurons drive, rather than inhibit, the development of nociceptive sensitization after spinal injury.
Collapse
Affiliation(s)
- Yung-Jen Huang
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - Kuan H Lee
- Center for Pain Research, Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lauren Murphy
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - James W Grau
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
27
|
Anti-Inflammatory Mechanism of Neural Stem Cell Transplantation in Spinal Cord Injury. Int J Mol Sci 2016; 17:ijms17091380. [PMID: 27563878 PMCID: PMC5037660 DOI: 10.3390/ijms17091380] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 01/08/2023] Open
Abstract
Neural stem cell (NSC) transplantation has been proposed to promote functional recovery after spinal cord injury. However, a detailed understanding of the mechanisms of how NSCs exert their therapeutic plasticity is lacking. We transplanted mouse NSCs into the injured spinal cord seven days after SCI, and the Basso Mouse Scale (BMS) score was performed to assess locomotor function. The anti-inflammatory effects of NSC transplantation was analyzed by immunofluorescence staining of neutrophil and macrophages and the detection of mRNA levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6) and interleukin-12 (IL-12). Furthermore, bone marrow-derived macrophages (BMDMs) were co-cultured with NSCs and followed by analyzing the mRNA levels of inducible nitric oxide synthase (iNOS), TNF-α, IL-1β, IL-6 and IL-10 with quantitative real-time PCR. The production of TNF-α and IL-1β by BMDMs was examined using the enzyme-linked immunosorbent assay (ELISA). Transplanted NSCs had significantly increased BMS scores (p < 0.05). Histological results showed that the grafted NSCs migrated from the injection site toward the injured area. NSCs transplantation significantly reduced the number of neutrophils and iNOS+/Mac-2+ cells at the epicenter of the injured area (p < 0.05). Meanwhile, mRNA levels of TNF-α, IL-1β, IL-6 and IL-12 in the NSCs transplantation group were significantly decreased compared to the control group. Furthermore, NSCs inhibited the iNOS expression of BMDMs and the release of inflammatory factors by macrophages in vitro (p < 0.05). These results suggest that NSC transplantation could modulate SCI-induced inflammatory responses and enhance neurological function after SCI via reducing M1 macrophage activation and infiltrating neutrophils. Thus, this study provides a new insight into the mechanisms responsible for the anti-inflammatory effect of NSC transplantation after SCI.
Collapse
|
28
|
Hansen CN, Norden DM, Faw TD, Deibert R, Wohleb ES, Sheridan JF, Godbout JP, Basso DM. Lumbar Myeloid Cell Trafficking into Locomotor Networks after Thoracic Spinal Cord Injury. Exp Neurol 2016; 282:86-98. [PMID: 27191729 DOI: 10.1016/j.expneurol.2016.05.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/28/2016] [Accepted: 05/13/2016] [Indexed: 01/05/2023]
Abstract
Spinal cord injury (SCI) promotes inflammation along the neuroaxis that jeopardizes plasticity, intrinsic repair and recovery. While inflammation at the injury site is well-established, less is known within remote spinal networks. The presence of bone marrow-derived immune (myeloid) cells in these areas may further impede functional recovery. Previously, high levels of the gelatinase, matrix metalloproteinase-9 (MMP-9) occurred within the lumbar enlargement after thoracic SCI and impeded activity-dependent recovery. Since SCI-induced MMP-9 potentially increases vascular permeability, myeloid cell infiltration may drive inflammatory toxicity in locomotor networks. Therefore, we examined neurovascular reactivity and myeloid cell infiltration in the lumbar cord after thoracic SCI. We show evidence of region-specific recruitment of myeloid cells into the lumbar but not cervical region. Myeloid infiltration occurred with concomitant increases in chemoattractants (CCL2) and cell adhesion molecules (ICAM-1) around lumbar vasculature 24h and 7days post injury. Bone marrow GFP chimeric mice established robust infiltration of bone marrow-derived myeloid cells into the lumbar gray matter 24h after SCI. This cell infiltration occurred when the blood-spinal cord barrier was intact, suggesting active recruitment across the endothelium. Myeloid cells persisted as ramified macrophages at 7days post injury in parallel with increased inhibitory GAD67 labeling. Importantly, macrophage infiltration required MMP-9.
Collapse
Affiliation(s)
- Christopher N Hansen
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Diana M Norden
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Timothy D Faw
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Rochelle Deibert
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Eric S Wohleb
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Division of Biosciences, , The Ohio State University, Columbus, OH 43210, USA.
| | - John F Sheridan
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Division of Biosciences, , The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan P Godbout
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH 43210, USA
| | - D Michele Basso
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
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
|
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
|
31
|
Lee KH, Turtle JD, Huang YJ, Strain MM, Baumbauer KM, Grau JW. Learning about time within the spinal cord: evidence that spinal neurons can abstract and store an index of regularity. Front Behav Neurosci 2015; 9:274. [PMID: 26539090 PMCID: PMC4612497 DOI: 10.3389/fnbeh.2015.00274] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/28/2015] [Indexed: 01/06/2023] Open
Abstract
Prior studies have shown that intermittent noxious stimulation has divergent effects on spinal cord plasticity depending upon whether it occurs in a regular (fixed time, FT) or irregular (variable time, VT) manner: In spinally transected animals, VT stimulation to the tail or hind leg impaired spinal learning whereas an extended exposure to FT stimulation had a restorative/protective effect. These observations imply that lower level systems are sensitive to temporal relations. Using spinally transected rats, it is shown that the restorative effect of FT stimulation emerges after 540 shocks; fewer shocks generate a learning impairment. The transformative effect of FT stimulation is related to the number of shocks administered, not the duration of exposure. Administration of 360 FT shocks induces a learning deficit that lasts 24 h. If a second bout of FT stimulation is given a day after the first, it restores the capacity to learn. This savings effect implies that the initial training episode had a lasting (memory-like) effect. Two bouts of shock have a transformative effect when applied at different locations or at difference frequencies, implying spinal systems abstract and store an index of regularity (rather than a specific interval). 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
| | - Joel D Turtle
- Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University College Station, TX, USA
| | - Yung-Jen Huang
- Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University College Station, TX, USA
| | - Misty M Strain
- 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
|
32
|
Puga DA, Tovar CA, Guan Z, Gensel JC, Lyman MS, McTigue DM, Popovich PG. Stress exacerbates neuron loss and microglia proliferation in a rat model of excitotoxic lower motor neuron injury. Brain Behav Immun 2015; 49:246-54. [PMID: 26100488 PMCID: PMC4567453 DOI: 10.1016/j.bbi.2015.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/01/2015] [Accepted: 06/08/2015] [Indexed: 11/19/2022] Open
Abstract
All individuals experience stress and hormones (e.g., glucocorticoids/GCs) released during stressful events can affect the structure and function of neurons. These effects of stress are best characterized for brain neurons; however, the mechanisms controlling the expression and binding affinity of glucocorticoid receptors in the spinal cord are different than those in the brain. Accordingly, whether stress exerts unique effects on spinal cord neurons, especially in the context of pathology, is unknown. Using a controlled model of focal excitotoxic lower motor neuron injury in rats, we examined the effects of acute or chronic variable stress on spinal cord motor neuron survival and glial activation. New data indicate that stress exacerbates excitotoxic spinal cord motor neuron loss and associated activation of microglia. In contrast, hypertrophy and hyperplasia of astrocytes and NG2+ glia were unaffected or were modestly suppressed by stress. Although excitotoxic lesions cause significant motor neuron loss and stress exacerbates this pathology, overt functional impairment did not develop in the relevant forelimb up to one week post-lesion. These data indicate that stress is a disease-modifying factor capable of altering neuron and glial responses to pathological challenges in the spinal cord.
Collapse
Affiliation(s)
- Denise A Puga
- Center for Brain and Spinal Cord Repair, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States
| | - C Amy Tovar
- Center for Brain and Spinal Cord Repair, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States
| | - Zhen Guan
- Center for Brain and Spinal Cord Repair, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Matthew S Lyman
- Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States
| | - Dana M McTigue
- Center for Brain and Spinal Cord Repair, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States; Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, Ohio 43210, United States
| |
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
|
Early administration of tumor necrosis factor-alpha antagonist promotes survival of transplanted neural stem cells and axon myelination after spinal cord injury in rats. Brain Res 2014; 1575:87-100. [DOI: 10.1016/j.brainres.2014.05.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/30/2014] [Accepted: 05/23/2014] [Indexed: 12/19/2022]
|
37
|
Tumor necrosis factor alpha: a link between neuroinflammation and excitotoxicity. Mediators Inflamm 2014; 2014:861231. [PMID: 24966471 PMCID: PMC4055424 DOI: 10.1155/2014/861231] [Citation(s) in RCA: 481] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/07/2014] [Indexed: 02/08/2023] Open
Abstract
Tumor necrosis factor alpha (TNF-α) is a proinflammatory cytokine that exerts both homeostatic and pathophysiological roles in the central nervous system. In pathological conditions, microglia release large amounts of TNF-α; this de novo production of TNF-α is an important component of the so-called neuroinflammatory response that is associated with several neurological disorders. In addition, TNF-α can potentiate glutamate-mediated cytotoxicity by two complementary mechanisms: indirectly, by inhibiting glutamate transport on astrocytes, and directly, by rapidly triggering the surface expression of Ca+2 permeable-AMPA receptors and NMDA receptors, while decreasing inhibitory GABAA receptors on neurons. Thus, the net effect of TNF-α is to alter the balance of excitation and inhibition resulting in a higher synaptic excitatory/inhibitory ratio. This review summarizes the current knowledge of the cellular and molecular mechanisms by which TNF-α links the neuroinflammatory and excitotoxic processes that occur in several neurodegenerative diseases, but with a special emphasis on amyotrophic lateral sclerosis (ALS). As microglial activation and upregulation of TNF-α expression is a common feature of several CNS diseases, as well as chronic opioid exposure and neuropathic pain, modulating TNF-α signaling may represent a valuable target for intervention.
Collapse
|
38
|
Zhou L, Wang H, Luo J, Xiong K, Zeng L, Chen D, Huang J. Regulatory effects of inhibiting the activation of glial cells on retinal synaptic plasticity. Neural Regen Res 2014; 9:385-393. [PMID: 25206825 PMCID: PMC4146193 DOI: 10.4103/1673-5374.128240] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2014] [Indexed: 01/09/2023] Open
Abstract
Various retinal injuries induced by ocular hypertension have been shown to induce plastic changes in retinal synapses, but the potential regulatory mechanism of synaptic plasticity after retinal injury was still unclear. A rat model of acute ocular hypertension was established by injecting saline intravitreally for an hour, and elevating the intraocular pressure to 14.63 kPa (110 mmHg). Western blot assay and immunofluorescence results showed that synaptophysin expression had a distinct spatiotemporal change that increased in the inner plexiform layer within 1 day and spread across the outer plexiform layer after 3 days. Glial fibrillary acidic protein expression in retinae was greatly increased after 3 days, and reached a peak at 7 days, which was also consistent with the peak time of synaptophysin expression in the outer plexiform layer following the increased intraocular pressure. Fluorocitrate, a glial metabolic inhibitor, was intravitreally injected to inhibit glial cell activation following high intraocular pressure. This significantly inhibited the enhanced glial fibrillary acidic protein expression induced by high intraocular pressure injury. Synaptophysin expression also decreased in the inner plexiform layer within a day and the widened distribution in the outer plexiform layer had disappeared by 3 days. The results suggested that retinal glial cell activation might play an important role in the process of retinal synaptic plasticity induced by acute high intraocular pressure through affecting the expression and distribution of synaptic functional proteins, such as synaptophysin.
Collapse
Affiliation(s)
- Lihong Zhou
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Hui Wang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jia Luo
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Leping Zeng
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Dan Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jufang Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
39
|
Elevated MMP-9 in the lumbar cord early after thoracic spinal cord injury impedes motor relearning in mice. J Neurosci 2013; 33:13101-11. [PMID: 23926264 DOI: 10.1523/jneurosci.1576-13.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Spinal cord injury results in distant pathology around putative locomotor networks that may jeopardize the recovery of locomotion. We previously showed that activated microglia and increased cytokine expression extend at least 10 segments below the injury to influence sensory function. Matrix metalloproteinase-9 (MMP-9) is a potent regulator of acute neuroinflammation. Whether MMP-9 is produced remote to the injury or influences locomotor plasticity remains unexamined. Therefore, we characterized the lumbar enlargement after a T9 spinal cord injury in C57BL/6 (wild-type [WT]) and MMP-9-null (knock-out [KO]) mice. Within 24 h, resident microglia displayed an activated phenotype alongside increased expression of progelatinase MMP-3 in WT mice. By 7 d, increases in active MMP-9 around lumbar vasculature and production of proinflammatory TNF-α were evident. Deletion of MMP-9 attenuated remote microglial activation and restored TNF-α expression to homeostatic levels. To determine whether MMP-9 impedes locomotor plasticity, we delivered lumbar-focused treadmill training in WT and KO mice during early (2-9 d) or late (35-42 d) phases of recovery. Robust behavioral improvements were observed by 7 d, when only trained KO mice stepped in the open field. Locomotor improvements were retained for 4 weeks as identified using state of the art mouse kinematics. Neither training nor MMP-9 depletion alone promoted recovery. The same intervention delivered late was ineffective, suggesting that lesion site sparing is insufficient to facilitate activity-based training and recovery. Our work suggests that by attenuating remote mechanisms of inflammation, acute treadmill training can harness endogenous spinal plasticity to promote robust recovery.
Collapse
|
40
|
Managing inflammation after spinal cord injury through manipulation of macrophage function. Neural Plast 2013; 2013:945034. [PMID: 24288627 PMCID: PMC3833318 DOI: 10.1155/2013/945034] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/17/2013] [Indexed: 12/23/2022] Open
Abstract
Spinal cord injury (SCI) triggers inflammation with activation of innate immune responses that contribute to secondary injury including oligodendrocyte apoptosis, demyelination, axonal degeneration, and neuronal death. Macrophage activation, accumulation, and persistent inflammation occur in SCI. Macrophages are heterogeneous cells with extensive functional plasticity and have the capacity to switch phenotypes by factors present in the inflammatory microenvironment of the injured spinal cord. This review will discuss the role of different polarized macrophages and the potential effect of macrophage-based therapies for SCI.
Collapse
|
41
|
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
|
42
|
Inoue T, Lin A, Ma X, McKenna SL, Creasey GH, Manley GT, Ferguson AR, Bresnahan JC, Beattie MS. Combined SCI and TBI: recovery of forelimb function after unilateral cervical spinal cord injury (SCI) is retarded by contralateral traumatic brain injury (TBI), and ipsilateral TBI balances the effects of SCI on paw placement. Exp Neurol 2013; 248:136-47. [PMID: 23770071 DOI: 10.1016/j.expneurol.2013.06.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 05/23/2013] [Accepted: 06/06/2013] [Indexed: 11/19/2022]
Abstract
A significant proportion (estimates range from 16 to 74%) of patients with spinal cord injury (SCI) have concomitant traumatic brain injury (TBI), and the combination often produces difficulties in planning and implementing rehabilitation strategies and drug therapies. For example, many of the drugs used to treat SCI may interfere with cognitive rehabilitation, and conversely drugs that are used to control seizures in TBI patients may undermine locomotor recovery after SCI. The current paper presents an experimental animal model for combined SCI and TBI to help drive mechanistic studies of dual diagnosis. Rats received a unilateral SCI (75 kdyn) at C5 vertebral level, a unilateral TBI (2.0 mm depth, 4.0 m/s velocity impact on the forelimb sensori-motor cortex), or both SCI+TBI. TBI was placed either contralateral or ipsilateral to the SCI. Behavioral recovery was examined using paw placement in a cylinder, grooming, open field locomotion, and the IBB cereal eating test. Over 6weeks, in the paw placement test, SCI+contralateral TBI produced a profound deficit that failed to recover, but SCI+ipsilateral TBI increased the relative use of the paw on the SCI side. In the grooming test, SCI+contralateral TBI produced worse recovery than either lesion alone even though contralateral TBI alone produced no observable deficit. In the IBB forelimb test, SCI+contralateral TBI revealed a severe deficit that recovered in 3 weeks. For open field locomotion, SCI alone or in combination with TBI resulted in an initial deficit that recovered in 2 weeks. Thus, TBI and SCI affected forelimb function differently depending upon the test, reflecting different neural substrates underlying, for example, exploratory paw placement and stereotyped grooming. Concurrent SCI and TBI had significantly different effects on outcomes and recovery, depending upon laterality of the two lesions. Recovery of function after cervical SCI was retarded by the addition of a moderate TBI in the contralateral hemisphere in all tests, but forepaw placements were relatively increased by an ipsilateral TBI relative to SCI alone, perhaps due to the dual competing injuries influencing the use of both forelimbs. These findings emphasize the complexity of recovery from combined CNS injuries, and the possible role of plasticity and laterality in rehabilitation, and provide a start towards a useful preclinical model for evaluating effective therapies for combine SCI and TBI.
Collapse
Affiliation(s)
- Tomoo Inoue
- Department of Neurological Surgery, University of California San Francisco, and San Francisco General Hospital, San Francisco, CA, USA; Brain and Spinal Injury Center, University of California San Francisco, San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Porkka-Heiskanen T. Sleep homeostasis. Curr Opin Neurobiol 2013; 23:799-805. [PMID: 23510741 DOI: 10.1016/j.conb.2013.02.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 10/27/2022]
Abstract
Research on sleep homeostasis aims to answer the question: how does the brain measure the duration and intensity of previous wakefulness in order to increase the duration and intensity of subsequent sleep? The search of regulatory factors has identified a number of potential molecules that increase their concentration in waking and decrease it during sleep. These factors regulate many physiological functions, including energy metabolism, neural plasticity and immune functions and one molecule may participate in the regulation of all these functions. The method to study regulation of sleep homeostasis is experimental prolongation of waking, which is used also to address the question of physiological purpose of sleep: prolonging wakefulness provokes symptoms that tell us what goes wrong during lack of sleep. The interpretation of the role of each identified factor in the regulation of sleep/sleep homeostasis reflects the theoretical background concept of the research. Presently three main concepts are being actively studied: the energy (depletion) hypothesis, the neural plasticity hypothesis and the (immune) defense hypothesis.
Collapse
Affiliation(s)
- Tarja Porkka-Heiskanen
- University of Helsinki, Institute of Biomedicine, Department of Physiology, PO Box 63, 00014 University of Helsinki, Finland.
| |
Collapse
|
44
|
Ferguson AR, Huie JR, Crown ED, Baumbauer KM, Hook MA, Garraway SM, Lee KH, Hoy KC, Grau JW. Maladaptive spinal plasticity opposes spinal learning and recovery in spinal cord injury. Front Physiol 2012; 3:399. [PMID: 23087647 PMCID: PMC3468083 DOI: 10.3389/fphys.2012.00399] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 09/20/2012] [Indexed: 01/23/2023] Open
Abstract
Synaptic plasticity within the spinal cord has great potential to facilitate recovery of function after spinal cord injury (SCI). Spinal plasticity can be induced in an activity-dependent manner even without input from the brain after complete SCI. A mechanistic basis for these effects is provided by research demonstrating that spinal synapses have many of the same plasticity mechanisms that are known to underlie learning and memory in the brain. In addition, the lumbar spinal cord can sustain several forms of learning and memory, including limb-position training. However, not all spinal plasticity promotes recovery of function. Central sensitization of nociceptive (pain) pathways in the spinal cord may emerge in response to various noxious inputs, demonstrating that plasticity within the spinal cord may contribute to maladaptive pain states. In this review we discuss interactions between adaptive and maladaptive forms of activity-dependent plasticity in the spinal cord below the level of SCI. The literature demonstrates that activity-dependent plasticity within the spinal cord must be carefully tuned to promote adaptive spinal training. Prior work from our group has shown that stimulation that is delivered in a limb position-dependent manner or on a fixed interval can induce adaptive plasticity that promotes future spinal cord learning and reduces nociceptive hyper-reactivity. On the other hand, stimulation that is delivered in an unsynchronized fashion, such as randomized electrical stimulation or peripheral skin injuries, can generate maladaptive spinal plasticity that undermines future spinal cord learning, reduces recovery of locomotor function, and promotes nociceptive hyper-reactivity after SCI. We review these basic phenomena, how these findings relate to the broader spinal plasticity literature, discuss the cellular and molecular mechanisms, and finally discuss implications of these and other findings for improved rehabilitative therapies after SCI.
Collapse
Affiliation(s)
- Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ferguson AR, Huie JR, Crown ED, Grau JW. Central nociceptive sensitization vs. spinal cord training: opposing forms of plasticity that dictate function after complete spinal cord injury. Front Physiol 2012; 3:396. [PMID: 23060820 PMCID: PMC3463829 DOI: 10.3389/fphys.2012.00396] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 09/15/2012] [Indexed: 11/13/2022] Open
Abstract
The spinal cord demonstrates several forms of plasticity that resemble brain-dependent learning and memory. Among the most studied form of spinal plasticity is spinal memory for noxious (nociceptive) stimulation. Numerous papers have described central pain as a spinally-stored memory that enhances future responses to cutaneous stimulation. This phenomenon, known as central sensitization, has broad relevance to a range of pathological conditions. Work from the spinal cord injury (SCI) field indicates that the lumbar spinal cord demonstrates several other forms of plasticity, including formal learning and memory. After complete thoracic SCI, the lumbar spinal cord can be trained by delivering stimulation to the hindleg when the leg is extended. In the presence of this response-contingent stimulation the spinal cord rapidly learns to hold the leg in a flexed position, a centrally mediated effect that meets the formal criteria for instrumental (response-outcome) learning. Instrumental flexion training produces a central change in spinal plasticity that enables future spinal learning on both the ipsilateral and contralateral leg. However, if stimulation is given in a response-independent manner, the spinal cord develops central maladaptive plasticity that undermines future spinal learning on both legs. The present paper tests for interactions between spinal cord training and central nociceptive sensitization after complete spinal cord transection. We found that spinal training alters future central sensitization by intradermal formalin (24 h post-training). Conversely intradermal formalin impaired future spinal learning (24 h post-injection). Because formalin-induced central sensitization has been shown to involve NMDA receptor activation, we tested whether pre-treatment with NMDA would also affect spinal learning in manner similar to formalin. We found intrathecal NMDA impaired learning in a dose-dependent fashion, and that this effect endures for at least 24 h. These data provide strong evidence for an opposing relationship between nociceptive plasticity and use-dependent learning in the spinal cord. The present work has clinical implications given recent findings that adaptive spinal training improves recovery in humans with SCI. Nociception below the SCI may undermine this rehabilitation potential.
Collapse
Affiliation(s)
- Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | | | | | | |
Collapse
|
46
|
Hoy KC, Huie JR, Grau JW. AMPA receptor mediated behavioral plasticity in the isolated rat spinal cord. Behav Brain Res 2012; 236:319-326. [PMID: 22982187 DOI: 10.1016/j.bbr.2012.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/31/2012] [Accepted: 09/05/2012] [Indexed: 12/18/2022]
Abstract
Previous research has demonstrated that the spinal cord is capable of a simple form of instrumental learning. Spinally transected rats that receive shock to a hind leg in an extended position quickly learn to maintain the leg in a flexed position, reducing net shock exposure whenever that leg is flexed. Subjects that receive shock independent of leg position (uncontrollable shock) do not exhibit an increase in flexion duration and later fail to learn when tested with controllable shock (learning deficit). The present study examined the role of the ionotropic glutamate receptor α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) in spinal learning. Intrathecal application of the AMPA receptor antagonist CNQX disrupted performance of a spinal instrumental learning in a dose dependent fashion (Experiment 1). CNQX also disrupted the maintenance of the instrumental response (Experiment 2) and blocked the induction of the learning deficit (Experiment 3). Intrathecal application of the agonist AMPA had a non-monotonic effect, producing a slight facilitation of performance at a low dose and disrupting learning at a high concentration (Experiment 4). Within the dose range tested, intrathecal application of AMPA did not have a long-term effect (Experiment 5). The results suggest that AMPA-mediated transmission plays an essential role in both instrumental learning and the induction of the learning deficit.
Collapse
Affiliation(s)
- Kevin C Hoy
- Texas A&M University, College Station, TX 77843-4235, United States.
| | - J Russell Huie
- University of California, San Francisco, CA, United States
| | - James W Grau
- Texas A&M University, College Station, TX 77843-4235, United States
| |
Collapse
|
47
|
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
|