1
|
Liu Y, Cai X, Shi B, Mo Y, Zhang J, Luo W, Yu B, Li X. Mechanisms and Therapeutic Prospects of Microglia-Astrocyte Interactions in Neuropathic Pain Following Spinal Cord Injury. Mol Neurobiol 2025; 62:4654-4676. [PMID: 39470872 DOI: 10.1007/s12035-024-04562-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/16/2024] [Indexed: 11/01/2024]
Abstract
Neuropathic pain is a prevalent and debilitating condition experienced by the majority of individuals with spinal cord injury (SCI). The complex pathophysiology of neuropathic pain, involving continuous activation of microglia and astrocytes, reactive gliosis, and altered neuronal plasticity, poses significant challenges for effective treatment. This review focuses on the pivotal roles of microglia and astrocytes, the two major glial cell types in the central nervous system, in the development and maintenance of neuropathic pain after SCI. We highlight the extensive bidirectional interactions between these cells, mediated by the release of inflammatory mediators, neurotransmitters, and neurotrophic factors, which contribute to the amplification of pain signaling. Understanding the microglia-astrocyte crosstalk and its impact on neuronal function is crucial for developing novel therapeutic strategies targeting neuropathic pain. In addition, this review discusses the fundamental biology, post-injury pain roles, and therapeutic prospects of microglia and astrocytes in neuropathic pain after SCI and elucidates the specific signaling pathways involved. We also speculated that the extracellular matrix (ECM) can affect the glial cells as well. Furthermore, we also mentioned potential targeted therapies, challenges, and progress in clinical trials, as well as new biomarkers and therapeutic targets. Finally, other relevant cell interactions in neuropathic pain and the role of glial cells in other neuropathic pain conditions have been discussed. This review serves as a comprehensive resource for further investigations into the microglia-astrocyte interaction and the detailed mechanisms of neuropathic pain after SCI, with the aim of improving therapeutic efficacy.
Collapse
Affiliation(s)
- Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xintong Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bowen Shi
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yajie Mo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianmin Zhang
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenting Luo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bodong Yu
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xi Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
2
|
Goltash S, Khodr R, Bui TV, Laliberte AM. An optogenetic mouse model of hindlimb spasticity after spinal cord injury. Exp Neurol 2025; 386:115157. [PMID: 39863244 DOI: 10.1016/j.expneurol.2025.115157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Spasticity is a common comorbidity of spinal cord injury (SCI), disrupting motor function and resulting in significant discomfort. While elements of post-SCI spasticity can be assessed using pre-clinical SCI models, the robust measurement of spasticity severity can be difficult due to its periodic and spontaneous appearance. Electrical stimulation of sensory afferents can elicit spasticity-associated motor responses, such as spasms; however, placing surface electrodes on the hindlimbs of awake animals can induce stress or encumbrance that could influence the expression of behaviour. Therefore, we have generated a mouse model of SCI-related spasticity that utilizes optogenetics to activate a subset of cutaneous VGLUT2+ sensory afferents to produce reliable incidences of spasticity-associated responses in the hindlimb. To examine the efficacy of this optogenetic SCI spasticity model, a T9-T10 complete transection injury was performed in Islet1-Cre+/-;VGLUT2-Flp+/-;CreON-FlpON-CatCh+/- mice, followed by the implantation of EMG electrodes into the left and right gastrocnemius and tibialis anterior muscles. EMG recordings were performed during episodic optogenetic stimulation (1-2 sessions per week until 5 weeks post-injury (wpi); n = 10 females, 5 males). A subset of these mice (n = 3 females, 2 males) was also tested at 10 wpi. During each recording session, an optic fiber coupled to a 470 nm wavelength LED was used to deliver 9 × 100 ms light pulses to the palmar surface of each hind paw. The results of these recordings demonstrated significant increases in the amplitude of EMG responses to the light stimulus from 2 wpi to 10 wpi, suggesting increased excitability of cutaneous sensorimotor pathways. Interestingly, this effect was significantly greater in the female cohort than in the males. Incidences of prolonged involuntary muscle contraction in response to the stimulus (fictive spasms) were also detected through EMG and visual observation during the testing period, supporting the presence of spasticity. As such, the optogenetic mouse model developed for this study appears to elicit spasticity-associated behaviours in SCI mice reliably and may be valuable for studying SCI-related limb spasticity mechanisms and therapeutic.
Collapse
Affiliation(s)
- Sara Goltash
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Riham Khodr
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Tuan V Bui
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, Ontario, Canada.
| | - Alex M Laliberte
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
3
|
Crul TC, Hakbijl – van der Wind AJ, van Laake – Geelen CM, Visser-Meily JMA, Post MWM, Stolwijk-Swüste JM. Use and experienced effectiveness of non-pharmacological treatments for chronic spinal cord injury related pain in The Netherlands: A cross-sectional survey. J Spinal Cord Med 2025; 48:199-207. [PMID: 38695723 PMCID: PMC11864019 DOI: 10.1080/10790268.2024.2345448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/26/2025] Open
Abstract
CONTEXT/OBJECTIVE Chronic pain is a common secondary condition in spinal cord injury (SCI). Pharmacological interventions to reduce pain are associated with side effects. The reported effects of non-pharmacological treatments are unclear. This study aims to examine the self-reported presence and type of pain, and the use, effectiveness and side effects of non-pharmacological treatments for pain. DESIGN Cross-sectional survey regarding SCI-related pain and non-pharmacological treatments. SETTING Community, the Netherlands. PARTICIPANTS Outpatients with SCI from two rehabilitation centers. INTERVENTIONS Not applicable. OUTCOME MEASURES Self-reported presence and type of pain, use, effectiveness and side effects of non-pharmacological treatments. RESULTS A total of 371 patients (41.5%) returned the questionnaire. Median time since onset of SCI was 7 years. Pain following SCI was reported by 262 patients (70.6%). Neuropathic pain was reported most often (74.4%), followed by musculoskeletal pain (51.5%). Of patients with pain, 204 (77.9%) reported past or current use of non-pharmacological treatments. Non-pharmacological treatments used most were physiotherapy (67.6%), physical exercise (44.7%) and massage (22.5%). Of patients using non-pharmacological treatments, 152 patients (74.5%) reported the effect of their treatment. Most treatments for which the effect was reported, were described as moderately effective. Most side effects were reported for cannabis. CONCLUSION Patients with SCI experiencing pain often use non-pharmacological treatments. Most treatments were described as moderately effective. Research on specific non-pharmacological treatments and different types of pain separately is needed to further determine the effectiveness of non-pharmacological treatments.
Collapse
Affiliation(s)
- Tim C Crul
- Centre of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Centre, University Medical Centre Utrecht, and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
| | - Aline J Hakbijl – van der Wind
- Centre of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Centre, University Medical Centre Utrecht, and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
| | - Charlotte M van Laake – Geelen
- Adelante Centre of Expertise in Rehabilitation and Audiology, Hoensbroek, The Netherlands
- Department of Rehabilitation Medicine, Research School CAPHRI, Maastricht University, Maastricht, The Netherlands
| | - Johanna MA Visser-Meily
- Centre of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Centre, University Medical Centre Utrecht, and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
- Department of Rehabilitation, Physical Therapy Science & Sports, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel WM Post
- Centre of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Centre, University Medical Centre Utrecht, and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
- University of Groningen, University Medical Centre Groningen, Centre for Rehabilitation, Groningen, The Netherlands
| | - Janneke M Stolwijk-Swüste
- Centre of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Centre, University Medical Centre Utrecht, and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
| |
Collapse
|
4
|
Rau J, Joseph R, Weise L, Bryan J, Wardeh J, Konda A, Duplessis L, Hook MA. Acute Opioid Administration Undermines Recovery after SCI: Adverse Effects Are Not Restricted to Morphine. J Neurotrauma 2025. [PMID: 39912807 DOI: 10.1089/neu.2024.0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
Previous studies have shown that administration of high doses of morphine in the acute phase of spinal cord injury (SCI) significantly undermines locomotor recovery and increases symptoms of chronic pain in a rat spinal contusion model. Similarly, SCI patients treated with high doses of opioid for the first 24 h postinjury have increased symptoms of chronic pain 1 year later. Whether these adverse effects are driven by morphine only or all opioids compromise recovery after SCI, however, is unknown. Based on our previous findings we hypothesized that activation of the kappa opioid receptor (KOR) is key in the morphine-induced attenuation of locomotor recovery after SCI. Thus, we posited that opioids that engage KOR-mediated signaling pathways (morphine, oxycodone) would undermine recovery, and clinically relevant opioids with less KOR activity (fentanyl and buprenorphine) would not. To test this, we compared the effects of the clinically relevant opioids on locomotor recovery and pain in a male rat spinal contusion model. Rats were given a moderate spinal contusion injury followed by 7 days of intravenous morphine, oxycodone, fentanyl, buprenorphine, or saline, and recovery was assessed for 28 days. All opioids produced analgesia on tests of thermal, mechanical, and incremented shock reactivity. However, tolerance developed rapidly with buprenorphine administration, particularly with daily administrations of 5 morphine milligram equivalent (MME) buprenorphine. Opioid-induced hyperalgesia (OIH) also developed across days following administration of higher doses (10 MME, 20 MME) of morphine and oxycodone. Fentanyl and buprenorphine did not produce OIH. Contrary to our hypothesis, however, we found that high doses of all opioids reduced recovery of locomotor function. Unlike the other opioids, the effects of buprenorphine on locomotor recovery appeared transient, but it also produced chronic pain. Morphine, oxycodone, and buprenorphine decreased reactivity thresholds on tests of mechanical and incremented shock stimulation. In sum, all opioids undermined long-term recovery in the rat model. Further interrogation of the molecular mechanisms driving the adverse effects is essential. This study provides critical insight into pain management strategies in the acute phase of SCI and potential long-term consequences of early opioid administration.
Collapse
Affiliation(s)
- Josephina Rau
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, College Station, Texas, USA
| | - Rose Joseph
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Lara Weise
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Jessica Bryan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, College Station, Texas, USA
| | - Jad Wardeh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Alekya Konda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Landon Duplessis
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, College Station, Texas, USA
| |
Collapse
|
5
|
Mishra S, Mishra Y, Kumar A. Marine-derived bioactive compounds for neuropathic pain: pharmacology and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03667-7. [PMID: 39797987 DOI: 10.1007/s00210-024-03667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 01/13/2025]
Abstract
Neuropathic pain, a challenging condition often associated with diabetes, trauma, or chemotherapy, impairs patients' quality of life. Current treatments often provide inconsistent relief and notable adverse effects, highlighting the urgent need for safer and more effective alternatives. This review investigates marine-derived bioactive compounds as potential novel therapies for neuropathic pain management. Marine organisms, including fungi, algae, cone snails, sponges, soft corals, tunicates, and fish, produce a diverse range of secondary metabolites with significant pharmacological properties. These include peptides (e.g., conopeptides, piscidin 1), non-peptides (e.g., guanidinium toxins, astaxanthin, docosahexaenoic acid, fucoidan, apigenin, fumagillin, aaptamine, flexibilide, excavatolide B, capnellenes, austrasulfones, lemnalol), and crude extracts (e.g., Spirulina platensis, Dunaliella salina, Cliothosa aurivilli). These compounds exhibit diverse mechanisms of action, such as modulating ion channels (e.g., transient receptor potential channels, voltage-gated sodium, calcium, and potassium channels, and G protein-coupled inwardly rectifying potassium channels), interacting with cell-surface receptors (e.g., nicotinic acetylcholine, NMDA, kainate, GABAB, and neurotensin receptors), inhibiting norepinephrine transporters, reducing oxidative stress, and attenuating neuroinflammation. These effects collectively contribute to alleviating nerve degeneration and symptoms of neuropathic pain, including hyperalgesia, allodynia, and associated psychomotor disturbances. Marine-derived bioactive compounds represent promising alternatives to conventional neuropathic pain treatments, to advance their development and assess their integration into neuropathic pain management strategies.
Collapse
Affiliation(s)
- Swapnil Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Yogesh Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India.
| |
Collapse
|
6
|
Hu J, Fan W, Xu Y, Li X, Zhang H, Li S, Xue L. Maladaptive changes in the homeostasis of AEA-TRPV1/CB1R induces pain-related hyperactivity of nociceptors after spinal cord injury. Cell Biosci 2025; 15:2. [PMID: 39789637 PMCID: PMC11720958 DOI: 10.1186/s13578-025-01345-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Neuropathic pain resulting from spinal cord injury (SCI) is associated with persistent hyperactivity of primary nociceptors. Anandamide (AEA) has been reported to modulate neuronal excitability and synaptic transmission through activation of cannabinoid type-1 receptors (CB1Rs) and transient receptor potential vanilloid 1 (TRPV1). However, the role of AEA and these receptors in the hyperactivity of nociceptors after SCI remains unclear. RESULTS In this study, we investigated the effects of AEA and its receptors on the hyperexcitability of mouse dorsal root ganglion (DRG) neurons after SCI. Using a whole-cell patch-clamp technique, we found that the timing of SCI-induced hyperexcitability in nociceptors paralleled an increase in the endocannabinoid AEA content. The expression of TRPV1 and CB1R was also upregulated at different time points after SCI. High-dose extracellular administration of AEA increased the excitability of naive DRG neurons, leading to the transition from a rapidly accommodating (RA) hypoexcitable state to a highly excitable non-accommodating (NA) state. These AEA-induced transitions were facilitated by increased TRPV1 transcription. Pharmacological and Ca2+ imaging experiments revealed that AEA induced hyperexcitability in nociceptors after SCI via the AEA-TRPV1-Ca2+ pathway, whereas activation of CB1Rs reduced SCI-induced hyperexcitability and maintained cytosolic Ca2+ concentration ([Ca2+]cyto) at low levels in the early stages of SCI. As the AEA and TRPV1 levels increased after SCI, adaptive neuroprotection transitioned to a maladaptive hyperactive state, leading to sustained pain. CONCLUSIONS Taken together, this study provides new insights into how endocannabinoids regulate nociceptor activity after SCI, offering potential targets for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- JiaQi Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, People's Republic of China
| | - WenYong Fan
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Yue Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - XiaoFei Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - HaoYang Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, People's Republic of China
| | - Lei Xue
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China.
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China.
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
7
|
Avonts BL, Shen Q, Wrobel NJ, Fessler RG, David BT. The relationship between changes in inflammation and locomotor function in sensory phenotypes of central neuropathic pain after spinal cord injury. Pain Rep 2024; 9:e1184. [PMID: 39399305 PMCID: PMC11469887 DOI: 10.1097/pr9.0000000000001184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/19/2024] [Accepted: 07/12/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction Central neuropathic pain (CNP) commonly develops in patients after spinal cord injury (SCI), causing debilitating symptoms and sensory abnormalities to mechanical and thermal stimuli. The biological variability of pain phenotypes in individuals has limited the number of positive outcomes. Thus, it is necessary to investigate the physiological processes contributing to sensory changes that develop over time. Objective To investigate the physiological processes contributing to neuropathic pain sensory changes and locomotor impairments with sensory phenotypes that develop over time. Methods Using the tail flick and von Frey tests, we performed hierarchical clustering to determine the subpopulation of rats that developed thermal and mechanical sensory abnormalities. To measure inflammation as a potential mediator of CNP phenotypes, we used flow cytometry and immunohistochemistry. Finally, to assess the secondary effects on locomotor recovery, up to 8 weeks after injury, we used the CatWalk test to assess multiple parameters of gait. Results The von Frey test showed a subpopulation of SCI rats that were hyposensitive to mechanical stimuli from 6 to 8 weeks after injury. The tail flick test showed a subpopulation of SCI rats that were hypersensitive to thermal stimuli at 1 week and 3 to 8 weeks after injury. Although there were no differences in inflammatory cells between subpopulations, we did see significant changes in locomotor recovery between rats with and without sensory abnormalities. Conclusion The myeloid cell population at large is not affected by mechanical or thermal phenotypes of pain in this model; however, locomotor recovery is impaired depending on the pain phenotype present. Further investigation into acute inflammatory cells may be insightful for predicting the development of pain phenotypes.
Collapse
Affiliation(s)
- Brittany L. Avonts
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Quan Shen
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Neal J. Wrobel
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Richard G. Fessler
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Brian T. David
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| |
Collapse
|
8
|
Starobova H, Alshammari A, Winkler IG, Vetter I. The role of the neuronal microenvironment in sensory function and pain pathophysiology. J Neurochem 2024; 168:3620-3643. [PMID: 36394416 DOI: 10.1111/jnc.15724] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
The high prevalence of pain and the at times low efficacy of current treatments represent a significant challenge to healthcare systems worldwide. Effective treatment strategies require consideration of the diverse pathophysiologies that underlie various pain conditions. Indeed, our understanding of the mechanisms contributing to aberrant sensory neuron function has advanced considerably. However, sensory neurons operate in a complex dynamic microenvironment that is controlled by multidirectional interactions of neurons with non-neuronal cells, including immune cells, neuronal accessory cells, fibroblasts, adipocytes, and keratinocytes. Each of these cells constitute and control the microenvironment in which neurons operate, inevitably influencing sensory function and the pathology of pain. This review highlights the importance of the neuronal microenvironment for sensory function and pain, focusing on cellular interactions in the skin, nerves, dorsal root ganglia, and spinal cord. We discuss the current understanding of the mechanisms by which neurons and non-neuronal cells communicate to promote or resolve pain, and how this knowledge could be used for the development of mechanism-based treatments.
Collapse
Affiliation(s)
- Hana Starobova
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Ammar Alshammari
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Ingrid G Winkler
- Mater Research Institute, The University of Queensland, Queensland, South Brisbane, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- The School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
9
|
Liu L, Liang Z, Zhang L, Feng Z, Cao F, Zhang Y, Yang X, Zhang L, Wang J, Zhu Q. Corticothalamic input derived from corticospinal neurons contributes to chronic neuropathic pain after spinal cord injury. Exp Neurol 2024; 381:114923. [PMID: 39142366 DOI: 10.1016/j.expneurol.2024.114923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
Neuropathic pain is a significant and persistent issue for individuals with spinal cord injuries (SCI), severely impacting their quality of life. While changes at the peripheral and spinal levels are known to contribute to SCI-related pain, whether and how supraspinal centers contribute to post SCI chronic neuropathic pain is poorly understood. Here, we first validated delayed development of chronic neuropathic pain in mice with moderate contusion SCI. To identify supraspinal regions involved in the pathology of neuropathic pain after SCI, we next performed an activity dependent genetic screening and identified multiple cortical and subcortical regions that were activated by innocuous tactile stimuli at a late stage following contusion SCI. Notably, chemogenetic inactivation of pain trapped neurons in the lateral thalamus alleviated neuropathic pain and reduced tactile stimuli evoked cortical overactivation. Retrograde tracing showed that contusion SCI led to enhanced corticothalamic axonal sprouting and over-activation of corticospinal neurons. Mechanistically, ablation or silencing of corticospinal neurons prevented the establishment or maintenance of chronic neuropathic pain following contusion SCI. These results highlighted a corticospinal-lateral thalamic feed-forward loop whose activation is required for the development and maintenance of chronic neuropathic pain after SCI. Our data thus shed lights into the central mechanisms underlying chronic neuropathic pain associated with SCI and the development of novel therapeutic avenues to treat refractory pain caused by traumatic brain or spinal cord injuries.
Collapse
Affiliation(s)
- Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fei Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunjian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijie Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qing Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
Sirucek L, De Schoenmacker I, Scheuren PS, Lütolf R, Gorrell LM, Langenfeld A, Baechler M, Rosner J, Wirth B, Hubli M, Schweinhardt P. Indication for spinal sensitization in chronic low back pain: mechanical hyperalgesia adjacent to but not within the most painful body area. Pain Rep 2024; 9:e1166. [PMID: 38910867 PMCID: PMC11191021 DOI: 10.1097/pr9.0000000000001166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction In 85% of patients with chronic low back pain (CLBP), no specific pathoanatomical cause can be identified. Besides primary peripheral drivers within the lower back, spinal or supraspinal sensitization processes might contribute to the patients' pain. Objectives The present study conceptualized the most painful area (MP) of patients with nonspecific CLBP as primarily affected area and assessed signs of peripheral, spinal, and supraspinal sensitization using quantitative sensory testing (QST) in MP, a pain-free area adjacent to MP (AD), and a remote, pain-free control area (CON). Methods Fifty-nine patients with CLBP (51 years, SD = 16.6, 22 female patients) and 35 pain-free control participants individually matched for age, sex, and testing areas (49 years, SD = 17.5, 19 female participants) underwent a full QST protocol in MP and a reduced QST protocol assessing sensory gain in AD and CON. Quantitative sensory testing measures, except paradoxical heat sensations and dynamic mechanical allodynia (DMA), were Z-transformed to the matched control participants and tested for significance using Z-tests (α = 0.001). Paradoxical heat sensations and DMA occurrence were compared between cohorts using Fisher's exact tests (α = 0.05). The same analyses were performed with a high-pain and a low-pain CLBP subsample (50% quantile). Results Patients showed cold and vibration hypoesthesia in MP (all Ps < 0.001) and mechanical hyperalgesia (P < 0.001) and more frequent DMA (P = 0.044) in AD. The results were mainly driven by the high-pain CLBP subsample. In CON, no sensory alterations were observed. Conclusion Mechanical hyperalgesia and DMA adjacent to but not within MP, the supposedly primarily affected area, might reflect secondary hyperalgesia originating from spinal sensitization in patients with CLBP.
Collapse
Affiliation(s)
- Laura Sirucek
- Department of Chiropractic Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Iara De Schoenmacker
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Biomedical Data Science Lab, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Paulina Simonne Scheuren
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Robin Lütolf
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Lindsay Mary Gorrell
- Department of Chiropractic Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Anke Langenfeld
- Department of Chiropractic Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Mirjam Baechler
- Department of Chiropractic Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Jan Rosner
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Brigitte Wirth
- Department of Chiropractic Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Michèle Hubli
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Petra Schweinhardt
- Department of Chiropractic Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Xu GY, Maskey M, Wu Z, Yang Q. Timed sulfonylurea modulation improves locomotor and sensory dysfunction following spinal cord injury. Front Pharmacol 2024; 15:1440198. [PMID: 39148545 PMCID: PMC11324438 DOI: 10.3389/fphar.2024.1440198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Traumatic spinal cord injury (SCI) results in immediate tissue necrosis and delayed secondary expansion of neurological damage, often resulting in lifelong paralysis, neurosensory dysfunction, and chronic pain. Progressive hemorrhagic necrosis (PHN) and excessive excitation are the main sources of secondary neural injury. Recent approaches to attenuate PHN by glibenclamide can improve locomotor function after SCI. However, use of glibenclamide can exacerbate development of SCI-induced chronic pain by inhibiting KATP channels to increase neuronal excitation and glial activation. In this study, we explored a treatment strategy involving administration of glibenclamide, which suppresses PHN, and diazoxide, which protects against neuronal excitation and inflammation, at different time intervals following spinal cord contusion. Our goal was to determine whether this combined approach enhances both sensory and motor function. Contusive SCI was induced at spinal segment T10 in adult rats. We found that KATP channels opener, diazoxide, decreased the hyperexcitability of primary sensory neurons after SCI by electrophysiology. Timed application of glibenclamide and diazoxide following contusion significantly improved locomotor function and mitigated development of SCI-induced chronic pain, as shown by behavioral evidence. Finally, we found that timed application of glibenclamide and diazoxide attenuates the inflammatory activity in the spinal cord and increases the survival of spinal matters following SCI. These preclinical studies introduce a promising potential treatment strategy to address SCI-induced dysfunction.
Collapse
Affiliation(s)
- Guo-Ying Xu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Manjit Maskey
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Zizhen Wu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Qing Yang
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
12
|
Wu J, Lin F, Chen B. Daphnoretin inhibited SCI-induced inflammation and activation of NF-κB pathway in spinal dorsal horn. Aging (Albany NY) 2024; 16:9680-9691. [PMID: 38843384 PMCID: PMC11210226 DOI: 10.18632/aging.205893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/16/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVE Spinal cord injury (SCI) is a devastating disease for which there is no safe and effective treatment at present. Daphnoretin is a natural discoumarin compound isolated from Wikstroemia indica with various pharmacological activities. Our study aimed to investigate the role of Daphnoretin in NF-κB pathway activation and inflammatory response after SCI. METHODS A mouse SCI model was constructed, and the Basso Mouse Scale Score and subscore were used to evaluate the effect of Daphnoretin on the movement capacity of mice. The effect of Daphnoretin on the activation of glial cells in the mouse model and BV2 cells was observed by immunofluorescence. PCR and ELISA were used to detect the expression of inflammatory factors, and Western blot was performed to detect the protein expression associated with NF-κB pathway. RESULTS Daphnoretin inhibited the loss of movement ability and the activation of glial cells in mice after SCI, and it also inhibited the activation of NF-κB pathway and the expression of inflammatory factors TNF-α and IL-1β in vivo and in vitro. CONCLUSIONS Daphnoretin can inhibit the activation of NF-κB pathway and the inflammatory response induced by SCI. Our study demonstrates the potential of Daphnoretin on clinical application for the treatment of SCI.
Collapse
Affiliation(s)
- Jiazhang Wu
- Department of Orthopaedics, Fuzhou Second General Hospital, School of Clinical Medicine, Fujian Medical University, Fuzhou 350007, China
- Department of Orthopaedics, Fuzhou Second Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou 350007, China
- Fujian Provincial Clinical Medical Research Center for First Aid and Rehabilitation in Orthopaedic Trauma, Fuzhou Trauma Medical Center, Fuzhou 350007, China
| | - Fengfei Lin
- Department of Orthopaedics, Fuzhou Second General Hospital, School of Clinical Medicine, Fujian Medical University, Fuzhou 350007, China
- Department of Orthopaedics, Fuzhou Second Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou 350007, China
- Fujian Provincial Clinical Medical Research Center for First Aid and Rehabilitation in Orthopaedic Trauma, Fuzhou Trauma Medical Center, Fuzhou 350007, China
| | - Bin Chen
- Department of Orthopaedics, Fuzhou Second General Hospital, School of Clinical Medicine, Fujian Medical University, Fuzhou 350007, China
- Department of Orthopaedics, Fuzhou Second Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou 350007, China
- Fujian Provincial Clinical Medical Research Center for First Aid and Rehabilitation in Orthopaedic Trauma, Fuzhou Trauma Medical Center, Fuzhou 350007, China
| |
Collapse
|
13
|
Türk Börü Ü, Kadir Sarıtaş Z, Görücü Özbek F, Bölük C, Acar H, Koç Y, Zeytin Demiral G. Alterations in the spinal cord, trigeminal nerve ganglion, and infraorbital nerve through inducing compression of the dorsal horn region at the upper cervical cord in trigeminal neuralgia. Brain Res 2024; 1832:148842. [PMID: 38447599 DOI: 10.1016/j.brainres.2024.148842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Idiopathic trigeminal neuralgia (TN) cases encountered frequently in daily practice indicate significant gaps that still need to be illuminated in the etiopathogenesis. In this study, a novel TN animal model was developed by compressing the dorsal horn (DH) of the upper cervical spinal cord. METHODS Eighteen rabbits were equally divided into three groups, namely control (CG), sham (SG), and spinal cord compression (SCC) groups. External pressure was applied to the left side at the C3 level in the SCC group. Dorsal hemilaminectomy was performed in the SG, and the operative side was closed without compression. No procedure was implemented in the control group. Samples from the SC, TG, and ION were taken after seven days. For the histochemical staining, damage and axons with myelin were scored using Hematoxylin and Eosin and Toluidine Blue, respectively. Immunohistochemistry, nuclei, apoptotic index, astrocyte activity, microglial labeling, and CD11b were evaluated. RESULTS Mechanical allodynia was observed on the ipsilateral side in the SCC group. In addition, both the TG and ION were partially damaged from SC compression, which resulted in significant histopathological changes and increased the expression of all markers in both the SG and SCC groups compared to that in the CG. There was a notable increase in tissue damage, an increase in the number of apoptotic nuclei, an increase in the apoptotic index, an indication of astrocytic gliosis, and an upsurge in microglial cells. Significant increases were noted in the SG group, whereas more pronounced significant increases were observed in the SCC group. Transmission electron microscopy revealed myelin damage, mitochondrial disruption, and increased anchoring particles. Similar changes were observed to a lesser extent in the contralateral spinal cord. CONCLUSION Ipsilateral trigeminal neuropathic pain was developed due to upper cervical SCC. The clinical finding is supported by immunohistochemical and ultrastructural changes. Thus, alterations in the DH due to compression of the upper cervical region should be considered as a potential cause of idiopathic TN.
Collapse
Affiliation(s)
- Ülkü Türk Börü
- Department of Neurology University of Afyonkarahisar Health Sciences, Afyonkarahisar, Turkey
| | - Zülfükar Kadir Sarıtaş
- Department of Surgery, Faculty of Veterinary Medicine, University of Afyon Kocatepe, Afyonkarahisar, Turkey
| | - Fatma Görücü Özbek
- Department of Surgery, Faculty of Veterinary Medicine, University of Afyon Kocatepe, Afyonkarahisar, Turkey
| | - Cem Bölük
- Department of Neurology and Clinical Neurophysiology, Sanliurfa Training and Research Hospital, Sanliurfa, Turkey.
| | - Hakan Acar
- Department of Neurology University of Afyonkarahisar Health Sciences, Afyonkarahisar, Turkey
| | - Yusuf Koç
- Department of Surgery, Faculty of Veterinary Medicine, University of Afyon Kocatepe, Afyonkarahisar, Turkey
| | - Gökçe Zeytin Demiral
- Department of Neurology University of Afyonkarahisar Health Sciences, Afyonkarahisar, Turkey
| |
Collapse
|
14
|
Sliwinski C, Heutehaus L, Taberner FJ, Weiss L, Kampanis V, Tolou-Dabbaghian B, Cheng X, Motsch M, Heppenstall PA, Kuner R, Franz S, Lechner SG, Weidner N, Puttagunta R. Contribution of mechanoreceptors to spinal cord injury-induced mechanical allodynia. Pain 2024; 165:1336-1347. [PMID: 38739766 PMCID: PMC11090032 DOI: 10.1097/j.pain.0000000000003139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/29/2023] [Accepted: 10/27/2023] [Indexed: 05/16/2024]
Abstract
ABSTRACT Evidence from previous studies supports the concept that spinal cord injury (SCI)-induced neuropathic pain (NP) has its neural roots in the peripheral nervous system. There is uncertainty about how and to which degree mechanoreceptors contribute. Sensorimotor activation-based interventions (eg, treadmill training) have been shown to reduce NP after experimental SCI, suggesting transmission of pain-alleviating signals through mechanoreceptors. The aim of the present study was to understand the contribution of mechanoreceptors with respect to mechanical allodynia in a moderate mouse contusion SCI model. After genetic ablation of tropomyosin receptor kinase B expressing mechanoreceptors before SCI, mechanical allodynia was reduced. The identical genetic ablation after SCI did not yield any change in pain behavior. Peptidergic nociceptor sprouting into lamina III/IV below injury level as a consequence of SCI was not altered by either mechanoreceptor ablation. However, skin-nerve preparations of contusion SCI mice 7 days after injury yielded hyperexcitability in nociceptors, not in mechanoreceptors, which makes a substantial direct contribution of mechanoreceptors to NP maintenance unlikely. Complementing animal data, quantitative sensory testing in human SCI subjects indicated reduced mechanical pain thresholds, whereas the mechanical detection threshold was not altered. Taken together, early mechanoreceptor ablation modulates pain behavior, most likely through indirect mechanisms. Hyperexcitable nociceptors seem to be the main drivers of SCI-induced NP. Future studies need to focus on injury-derived factors triggering early-onset nociceptor hyperexcitability, which could serve as targets for more effective therapeutic interventions.
Collapse
Affiliation(s)
- Christopher Sliwinski
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Laura Heutehaus
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Lisa Weiss
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Vasileios Kampanis
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Bahardokht Tolou-Dabbaghian
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Xing Cheng
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie Motsch
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Steffen Franz
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan G. Lechner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Department of Anesthesiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Laboratory of Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
15
|
Hecht JS, Moore KLJ, Roberts RF. Individuals With Prior Chronic Pain and Long-Term Opioid Treatment May Experience Persistence of That Pain Even After Subsequent Complete Cervical Spinal Cord Injury: Suggestions From a Prospective Case-Controlled Study. Arch Rehabil Res Clin Transl 2024; 6:100338. [PMID: 39006114 PMCID: PMC11240028 DOI: 10.1016/j.arrct.2024.100338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Objective To determine whether chronic pain persists after complete spinal cord injury (SCI). Design Prospective observational study regarding the outcome of pre-existent chronic pain of inpatients admitted with new clinically diagnosed complete cervical SCI. For patients who acknowledged chronic pain of ≥3 years duration before the SCI, further questions explored whether they still experienced that pain, whether they were experiencing current posttraumatic pain, and whether they had any past exposure to opioids. The included patients were identified during the initial consultation in the trauma center for treatment of the SCI. Setting Level I trauma center. Participants From a total of 49 participants with acute cervical SCI with clinically diagnosed complete motor and sensory tetraplegia admitted between 2018 and 2020, 7 were selected on the basis of a history of chronic pain. Intervention Collected complete history and performed physical examination with serial follow-ups during the acute hospital stay until death or discharge. Main Outcome Measures The primary outcome was a finding of chronic pain experienced before new clinical diagnosis of complete SCI, compared with whether or not that pain continued after the SCI injury. The secondary outcome was the relation of persistent pain with opioid use; it was formulated after data collection. Results Among 49 patients with clinically diagnosed complete cervical SCIs, 7 had experienced prior chronic pain. Four participants experienced a continuation of the prior pain after their complete tetraplegia (4/7), whereas 3 participants did not (3/7). All the participants with continued pain had been previously treated with opioids, whereas those whose pain ceased had not received chronic opioid therapy. Conclusions There may be a unique form of chronic pain that is based in the brain, irrespective of peripheral pain or spinal mechanisms. Otherwise healthy people with longstanding antecedent chronic pain whose pain persists after acute clinically complete SCI with tetraplegia may provide a new model for evaluation of brain-based pain. Opioids may be requisite for this type of pain.
Collapse
Affiliation(s)
- Jeffrey S. Hecht
- Division of Surgical Rehabilitation, Department of Surgery, University of Tennessee, Knoxville, Knoxville, Tennessee, United States
| | - Kyle L. Johnson Moore
- Office of Research, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Roy F. Roberts
- Division of Trauma, Department of Surgery, University of Tennessee, Knoxville, Knoxville, Tennessee, United States
| |
Collapse
|
16
|
Yang WW, Matyas JJ, Li Y, Lee H, Lei Z, Renn CL, Faden AI, Dorsey SG, Wu J. Dissecting Genetic Mechanisms of Differential Locomotion, Depression, and Allodynia after Spinal Cord Injury in Three Mouse Strains. Cells 2024; 13:759. [PMID: 38727295 PMCID: PMC11083625 DOI: 10.3390/cells13090759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Strain differences have been reported for motor behaviors, and only a subset of spinal cord injury (SCI) patients develop neuropathic pain, implicating genetic or genomic contribution to this condition. Here, we evaluated neuropsychiatric behaviors in A/J, BALB/c, and C57BL/6 male mice and tested genetic or genomic alterations following SCI. A/J and BALB/c naive mice showed significantly less locomotor activity and greater anxiety-like behavior than C57BL/6 mice. Although SCI elicited locomotor dysfunction, C57BL/6 and A/J mice showed the best and the worst post-traumatic recovery, respectively. Mild (m)-SCI mice showed deficits in gait dynamics. All moderate/severe SCI mice exhibited similar degrees of anxiety/depression. mSCI in BALB/c and A/J mice resulted in depression, whereas C57BL/6 mice did not exhibit depression. mSCI mice had significantly lower mechanical thresholds than their controls, indicating high cutaneous hypersensitivity. C57BL/6, but not A/J and BLAB/c mice, showed significantly lower heat thresholds than their controls. C57BL/6 mice exhibited spontaneous pain. RNAseq showed that genes in immune responses and wound healing were upregulated, although A/J mice showed the largest increase. The cell cycle and the truncated isoform of trkB genes were robustly elevated in SCI mice. Thus, different genomics are associated with post-traumatic recovery, underscoring the likely importance of genetic factors in SCI.
Collapse
Affiliation(s)
- Wendy W. Yang
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.W.Y.); (J.J.M.); (Y.L.); (Z.L.); (A.I.F.)
| | - Jessica J. Matyas
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.W.Y.); (J.J.M.); (Y.L.); (Z.L.); (A.I.F.)
| | - Yun Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.W.Y.); (J.J.M.); (Y.L.); (Z.L.); (A.I.F.)
| | - Hangnoh Lee
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Zhuofan Lei
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.W.Y.); (J.J.M.); (Y.L.); (Z.L.); (A.I.F.)
| | - Cynthia L. Renn
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD 21201, USA; (C.L.R.); (S.G.D.)
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.W.Y.); (J.J.M.); (Y.L.); (Z.L.); (A.I.F.)
| | - Susan G. Dorsey
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD 21201, USA; (C.L.R.); (S.G.D.)
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (W.W.Y.); (J.J.M.); (Y.L.); (Z.L.); (A.I.F.)
| |
Collapse
|
17
|
Altaf N, Rehman NU, Karim N, Khan I, Halim SA, Alotaibi BS, Hamad RS, Batiha GES, Tayyeb JZ, Turkistani A, Khan A, Al-Harrasi A. Attenuation of Streptozotocin-Induced Diabetic Neuropathic Allodynia by Flavone Derivative Through Modulation of GABA-ergic Mechanisms and Endogenous Biomarkers. Neurochem Res 2024; 49:980-997. [PMID: 38170385 DOI: 10.1007/s11064-023-04078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/19/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
Diabetic neuropathic pain is one of the most devasting disorders of peripheral nervous system. The loss of GABAergic inhibition is associated with the development of painful diabetic neuropathy. The current study evaluated the potential of 3-Hydroxy-2-methoxy-6-methyl flavone (3-OH-2'MeO6MF), to ameliorate peripheral neuropathic pain using an STZ-induced hyperglycemia rat model. The pain threshold was assessed by tail flick, cold, mechanical allodynia, and formalin test on days 0, 14, 21, and 28 after STZ administration accompanied by evaluation of several biochemical parameters. Administration of 3-OH-2'-MeO6MF (1,10, 30, and 100 mg/kg, i.p) significantly enhanced the tail withdrawal threshold in tail-flick and tail cold allodynia tests. 3-OH-2'-MeO6MF also increased the paw withdrawal threshold in mechanical allodynia and decreased paw licking time in the formalin test. Additionally, 3-OH-2'-MeO6MF also attenuated the increase in concentrations of myeloperoxidase (MPO), thiobarbituric acid reactive substances (TBARS), nitrite, TNF-α, and IL 6 along with increases in glutathione (GSH). Pretreatment of pentylenetetrazole (PTZ) (40 mg/kg, i.p.) abolished the antinociceptive effect of 3-OH-2'-MeO6MF in mechanical allodynia. Besides, the STZ-induced alterations in the GABA concentration and GABA transaminase activity attenuated by 3-OH-2'-MeO6MF treatment suggest GABAergic mechanisms. Molecular docking also authenticates the involvement of α2β2γ2L GABA-A receptors and GABA-T enzyme in the antinociceptive activities of 3-OH-2'-MeO6MF.
Collapse
Affiliation(s)
- Nouman Altaf
- Department of Pharmacy, University of Malakand, Chakdara, Lower Dir, KPK, Pakistan
| | - Najeeb Ur Rehman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat al Mouz, Initial Campus, 616, Nizwa, Sultanate of Oman
| | - Nasiara Karim
- Department of Pharmacy, University of Malakand, Chakdara, Lower Dir, KPK, Pakistan.
- Department of Pharmacy, University of Peshawar, Peshawar, KPK, Pakistan.
| | - Imran Khan
- Department of Pharmacy, University of Swabi, Swabi, KPK, Pakistan
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat al Mouz, Initial Campus, 616, Nizwa, Sultanate of Oman
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, 31982, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, 12411, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| | - Jehad Zuhair Tayyeb
- Department of Clinical Biochemistry, College of Medicine, University of Jeddah, 23890, Jeddah, Saudi Arabia
| | - Areej Turkistani
- Department of Pharmacology and Toxicology, College of Medicine, Taif University, 21944, Taif, Kingdom of Saudi Arabia
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat al Mouz, Initial Campus, 616, Nizwa, Sultanate of Oman.
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat al Mouz, Initial Campus, 616, Nizwa, Sultanate of Oman.
| |
Collapse
|
18
|
Song Y, Xue T, Guo S, Yu Z, Yun C, Zhao J, Song Z, Liu Z. Inhibition of aquaporin-4 and its subcellular localization attenuates below-level central neuropathic pain by regulating astrocyte activation in a rat spinal cord injury model. Neurotherapeutics 2024; 21:e00306. [PMID: 38237380 DOI: 10.1016/j.neurot.2023.e00306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/19/2023] [Indexed: 03/24/2024] Open
Abstract
The mechanisms of central neuropathic pain (CNP) caused by spinal cord injury have not been sufficiently studied. We have found that the upregulation of astrocytic aquaporin-4 (AQP4) aggravated peripheral neuropathic pain after spinal nerve ligation in rats. Using a T13 spinal cord hemisection model, we showed that spinal AQP4 was markedly upregulated after SCI and mainly expressed in astrocytes in the spinal dorsal horn (SDH). Inhibition of AQP4 with TGN020 suppressed astrocyte activation, attenuated the development and maintenance of below-level CNP and promoted motor function recovery in vivo. In primary astrocyte cultures, TGN020 also changed cell morphology, diminished cell proliferation and suppressed astrocyte activation. Moreover, T13 spinal cord hemisection induced cell-surface abundance of the AQP4 channel and perivascular localization in the SDH. Targeted inhibition of AQP4 subcellular localization with trifluoperazine effectively diminished astrocyte activation in vitro and further ablated astrocyte activation, attenuated the development and maintenance of below-level CNP, and accelerated functional recovery in vivo. Together, these results provide mechanistic insights into the roles of AQP4 in the development and maintenance of below-level CNP. Intervening with AQP4, including targeting AQP4 subcellular localization, might emerge as a promising agent to prevent chronic CNP after SCI.
Collapse
Affiliation(s)
- Yu Song
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Tao Xue
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Shiwu Guo
- Department of Orthopedics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, China
| | - Zhen Yu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Chengming Yun
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Jie Zhao
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China
| | - Zhiwen Song
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Zhiyuan Liu
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213003, China; Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China; The Wujin Clinical College of Xuzhou Medical University, Changzhou 213003, China.
| |
Collapse
|
19
|
Lai A, Iliff D, Zaheer K, Gansau J, Laudier DM, Zachariou V, Iatridis JC. Annulus Fibrosus Injury Induces Acute Neuroinflammation and Chronic Glial Response in Dorsal Root Ganglion and Spinal Cord-An In Vivo Rat Discogenic Pain Model. Int J Mol Sci 2024; 25:1762. [PMID: 38339040 PMCID: PMC10855200 DOI: 10.3390/ijms25031762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Chronic painful intervertebral disc (IVD) degeneration (i.e., discogenic pain) is a major source of global disability needing improved knowledge on multiple-tissue interactions and how they progress in order improve treatment strategies. This study used an in vivo rat annulus fibrosus (AF) injury-driven discogenic pain model to investigate the acute and chronic changes in IVD degeneration and spinal inflammation, as well as sensitization, inflammation, and remodeling in dorsal root ganglion (DRG) and spinal cord (SC) dorsal horn. AF injury induced moderate IVD degeneration with acute and broad spinal inflammation that progressed to DRG to SC changes within days and weeks, respectively. Specifically, AF injury elevated macrophages in the spine (CD68) and DRGs (Iba1) that peaked at 3 days post-injury, and increased microglia (Iba1) in SC that peaked at 2 weeks post-injury. AF injury also triggered glial responses with elevated GFAP in DRGs and SC at least 8 weeks post-injury. Spinal CD68 and SC neuropeptide Substance P both remained elevated at 8 weeks, suggesting that slow and incomplete IVD healing provides a chronic source of inflammation with continued SC sensitization. We conclude that AF injury-driven IVD degeneration induces acute spinal, DRG, and SC inflammatory crosstalk with sustained glial responses in both DRGs and SC, leading to chronic SC sensitization and neural plasticity. The known association of these markers with neuropathic pain suggests that therapeutic strategies for discogenic pain need to target both spinal and nervous systems, with early strategies managing acute inflammatory processes, and late strategies targeting chronic IVD inflammation, SC sensitization, and remodeling.
Collapse
Affiliation(s)
- Alon Lai
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Denise Iliff
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kashaf Zaheer
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jennifer Gansau
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Damien M Laudier
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Venetia Zachariou
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA 02118, USA
| | - James C Iatridis
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
20
|
Saunders MN, Griffin KV, Kalashnikova I, Kolpek D, Smith DR, Saito E, Cummings BJ, Anderson AJ, Shea LD, Park J. Biodegradable nanoparticles targeting circulating immune cells reduce central and peripheral sensitization to alleviate neuropathic pain following spinal cord injury. Pain 2024; 165:92-101. [PMID: 37463227 PMCID: PMC10787809 DOI: 10.1097/j.pain.0000000000002989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/26/2023] [Indexed: 07/20/2023]
Abstract
ABSTRACT Neuropathic pain is a critical source of comorbidity following spinal cord injury (SCI) that can be exacerbated by immune-mediated pathologies in the central and peripheral nervous systems. In this article, we investigate whether drug-free, biodegradable, poly(lactide- co -glycolide) (PLG) nanoparticle treatment mitigates the development of post-SCI neuropathic pain in female mice. Our results show that acute treatment with PLG nanoparticles following thoracic SCI significantly reduces tactile and cold hypersensitivity scores in a durable fashion. Nanoparticles primarily reduce peripheral immune-mediated mechanisms of neuropathic pain, including neuropathic pain-associated gene transcript frequency, transient receptor potential ankyrin 1 nociceptor expression, and MCP-1 (CCL2) chemokine production in the subacute period after injury. Altered central neuropathic pain mechanisms during this period are limited to reduced innate immune cell cytokine expression. However, in the chronic phase of SCI, nanoparticle treatment induces changes in both central and peripheral neuropathic pain signaling, driving reductions in cytokine production and other immune-relevant markers. This research suggests that drug-free PLG nanoparticles reprogram peripheral proalgesic pathways subacutely after SCI to reduce neuropathic pain outcomes and improve chronic central pain signaling.
Collapse
Affiliation(s)
- Michael N Saunders
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Kate V Griffin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Irina Kalashnikova
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
| | - Daniel Kolpek
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Brian J Cummings
- Department of Anatomy and Neurobiology, University of California, Irvine, CA USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA USA
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, CA USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Jonghyuck Park
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY USA
| |
Collapse
|
21
|
Jang K, Garraway SM. A review of dorsal root ganglia and primary sensory neuron plasticity mediating inflammatory and chronic neuropathic pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100151. [PMID: 38314104 PMCID: PMC10837099 DOI: 10.1016/j.ynpai.2024.100151] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
Pain is a sensory state resulting from complex integration of peripheral nociceptive inputs and central processing. Pain consists of adaptive pain that is acute and beneficial for healing and maladaptive pain that is often persistent and pathological. Pain is indeed heterogeneous, and can be expressed as nociceptive, inflammatory, or neuropathic in nature. Neuropathic pain is an example of maladaptive pain that occurs after spinal cord injury (SCI), which triggers a wide range of neural plasticity. The nociceptive processing that underlies pain hypersensitivity is well-studied in the spinal cord. However, recent investigations show maladaptive plasticity that leads to pain, including neuropathic pain after SCI, also exists at peripheral sites, such as the dorsal root ganglia (DRG), which contains the cell bodies of sensory neurons. This review discusses the important role DRGs play in nociceptive processing that underlies inflammatory and neuropathic pain. Specifically, it highlights nociceptor hyperexcitability as critical to increased pain states. Furthermore, it reviews prior literature on glutamate and glutamate receptors, voltage-gated sodium channels (VGSC), and brain-derived neurotrophic factor (BDNF) signaling in the DRG as important contributors to inflammatory and neuropathic pain. We previously reviewed BDNF's role as a bidirectional neuromodulator of spinal plasticity. Here, we shift focus to the periphery and discuss BDNF-TrkB expression on nociceptors, non-nociceptor sensory neurons, and non-neuronal cells in the periphery as a potential contributor to induction and persistence of pain after SCI. Overall, this review presents a comprehensive evaluation of large bodies of work that individually focus on pain, DRG, BDNF, and SCI, to understand their interaction in nociceptive processing.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Sandra M. Garraway
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
22
|
Willits AB, Kader L, Eller O, Roberts E, Bye B, Strope T, Freudenthal BD, Umar S, Chintapalli S, Shankar K, Pei D, Christianson J, Baumbauer KM, Young EE. Spinal cord injury-induced neurogenic bowel: A role for host-microbiome interactions in bowel pain and dysfunction. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100156. [PMID: 38601267 PMCID: PMC11004406 DOI: 10.1016/j.ynpai.2024.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
Background and aims Spinal cord injury (SCI) affects roughly 300,000 Americans with 17,000 new cases added annually. In addition to paralysis, 60% of people with SCI develop neurogenic bowel (NB), a syndrome characterized by slow colonic transit, constipation, and chronic abdominal pain. The knowledge gap surrounding NB mechanisms after SCI means that interventions are primarily symptom-focused and largely ineffective. The goal of the present studies was to identify mechanism(s) that initiate and maintain NB after SCI as a critical first step in the development of evidence-based, novel therapeutic treatment options. Methods Following spinal contusion injury at T9, we observed alterations in bowel structure and function reflecting key clinical features of NB. We then leveraged tissue-specific whole transcriptome analyses (RNAseq) and fecal 16S rRNA amplicon sequencing in combination with histological, molecular, and functional (Ca2+ imaging) approaches to identify potential mechanism(s) underlying the generation of the NB phenotype. Results In agreement with prior reports focused on SCI-induced changes in the skin, we observed a rapid and persistent increase in expression of calcitonin gene-related peptide (CGRP) expression in the colon. This is suggestive of a neurogenic inflammation-like process engaged by antidromic activity of below-level primary afferents following SCI. CGRP has been shown to disrupt colon homeostasis and negatively affect peristalsis and colon function. As predicted, contusion SCI resulted in increased colonic transit time, expansion of lymphatic nodules, colonic structural and genomic damage, and disruption of the inner, sterile intestinal mucus layer corresponding to increased CGRP expression in the colon. Gut microbiome colonization significantly shifted over 28 days leading to the increase in Anaeroplasma, a pathogenic, gram-negative microbe. Moreover, colon specific vagal afferents and enteric neurons were hyperresponsive after SCI to different agonists including fecal supernatants. Conclusions Our data suggest that SCI results in overexpression of colonic CGRP which could alter colon structure and function. Neurogenic inflammatory-like processes and gut microbiome dysbiosis can also sensitize vagal afferents, providing a mechanism for visceral pain despite the loss of normal sensation post-SCI. These data may shed light on novel therapeutic interventions targeting this process to prevent NB development in patients.
Collapse
Affiliation(s)
- Adam B. Willits
- Department of Anesthesiology, Pain and Perioperative Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Leena Kader
- Department of Anesthesiology, Pain and Perioperative Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | - Olivia Eller
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Emily Roberts
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Bailey Bye
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | - Taylor Strope
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Bret D. Freudenthal
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sree Chintapalli
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dong Pei
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, United States
| | - Julie Christianson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Kyle M. Baumbauer
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Erin E. Young
- Department of Anesthesiology, Pain and Perioperative Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
23
|
De Schoenmacker I, Sirucek L, Scheuren PS, Lütolf R, Gorrell LM, Brunner F, Curt A, Rosner J, Schweinhardt P, Hubli M. Sensory phenotypes in complex regional pain syndrome and chronic low back pain-indication of common underlying pathomechanisms. Pain Rep 2023; 8:e1110. [PMID: 38027464 PMCID: PMC10653599 DOI: 10.1097/pr9.0000000000001110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction First-line pain treatment is unsatisfactory in more than 50% of chronic pain patients, likely because of the heterogeneity of mechanisms underlying pain chronification. Objectives This cross-sectional study aimed to better understand pathomechanisms across different chronic pain cohorts, regardless of their diagnoses, by identifying distinct sensory phenotypes through a cluster analysis. Methods We recruited 81 chronic pain patients and 63 age-matched and sex-matched healthy controls (HC). Two distinct chronic pain cohorts were recruited, ie, complex regional pain syndrome (N = 20) and low back pain (N = 61). Quantitative sensory testing (QST) was performed in the most painful body area to investigate somatosensory changes related to clinical pain. Furthermore, QST was conducted in a pain-free area to identify remote sensory alterations, indicating more widespread changes in somatosensory processing. Results Two clusters were identified based on the QST measures in the painful area, which did not represent the 2 distinct pain diagnoses but contained patients from both cohorts. Cluster 1 showed increased pain sensitivities in the painful and control area, indicating central sensitization as a potential pathomechanism. Cluster 2 showed a similar sensory profile as HC in both tested areas. Hence, either QST was not sensitive enough and more objective measures are needed to detect sensitization within the nociceptive neuraxis or cluster 2 may not have pain primarily because of sensitization, but other factors such as psychosocial ones are involved. Conclusion These findings support the notion of shared pathomechanisms irrespective of the pain diagnosis. Conversely, different mechanisms might contribute to the pain of patients with the same diagnosis.
Collapse
Affiliation(s)
- Iara De Schoenmacker
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Laura Sirucek
- Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
- Department of Chiropractic Medicine, Integrative Spinal Research Group, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Paulina S. Scheuren
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Robin Lütolf
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Lindsay M. Gorrell
- Department of Chiropractic Medicine, Integrative Spinal Research Group, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Florian Brunner
- Physical Medicine and Rheumatology, Balgrist University Hospital, Zurich, Switzerland
| | - Armin Curt
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Jan Rosner
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Petra Schweinhardt
- Department of Chiropractic Medicine, Integrative Spinal Research Group, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- Alan Edward Center for Research on Pain, McGill University, Montreal, QC, Canada
| | - Michèle Hubli
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Walters ET. Exaptation and Evolutionary Adaptation in Nociceptor Mechanisms Driving Persistent Pain. BRAIN, BEHAVIOR AND EVOLUTION 2023; 98:314-330. [PMID: 38035556 PMCID: PMC10922759 DOI: 10.1159/000535552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Several evolutionary explanations have been proposed for why chronic pain is a major clinical problem. One is that some mechanisms important for driving chronic pain, while maladaptive for modern humans, were adaptive because they enhanced survival. Evidence is reviewed for persistent nociceptor hyperactivity (PNH), known to promote chronic pain in rodents and humans, being an evolutionarily adaptive response to significant bodily injury, and primitive molecular mechanisms related to cellular injury and stress being exapted (co-opted or repurposed) to drive PNH and consequent pain. SUMMARY PNH in a snail (Aplysia californica), squid (Doryteuthis pealeii), fruit fly (Drosophila melanogaster), mice, rats, and humans has been documented as long-lasting enhancement of action potential discharge evoked by peripheral stimuli, and in some of these species as persistent extrinsically driven ongoing activity and/or intrinsic spontaneous activity (OA and SA, respectively). In mammals, OA and SA are often initiated within the protected nociceptor soma long after an inducing injury. Generation of OA or SA in nociceptor somata may be very rare in invertebrates, but prolonged afterdischarge in nociceptor somata readily occurs in sensitized Aplysia. Evidence for the adaptiveness of injury-induced PNH has come from observations of decreased survival of injured squid exposed to predators when PNH is blocked, from plausible survival benefits of chronic sensitization after severe injuries such as amputation, and from the functional coherence and intricacy of mammalian PNH mechanisms. Major contributions of cAMP-PKA signaling (with associated calcium signaling) to the maintenance of PNH both in mammals and molluscs suggest that this ancient stress signaling system was exapted early during the evolution of nociceptors to drive hyperactivity following bodily injury. Vertebrates have retained core cAMP-PKA signaling modules for PNH while adding new extracellular modulators (e.g., opioids) and cAMP-regulated ion channels (e.g., TRPV1 and Nav1.8 channels). KEY MESSAGES Evidence from multiple phyla indicates that PNH is a physiological adaptation that decreases the risk of attacks on injured animals. Core cAMP-PKA signaling modules make major contributions to the maintenance of PNH in molluscs and mammals. This conserved signaling has been linked to ancient cellular responses to stress, which may have been exapted in early nociceptors to drive protective hyperactivity that can persist while bodily functions recover after significant injury.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
25
|
Li J, Tian C, Yuan S, Yin Z, Wei L, Chen F, Dong X, Liu A, Wang Z, Wu T, Tian C, Niu L, Wang L, Wang P, Xie W, Cao F, Shen H. Neuropathic pain following spinal cord hemisection induced by the reorganization in primary somatosensory cortex and regulated by neuronal activity of lateral parabrachial nucleus. CNS Neurosci Ther 2023; 29:3269-3289. [PMID: 37170721 PMCID: PMC10580357 DOI: 10.1111/cns.14258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/04/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023] Open
Abstract
AIMS Neuropathic pain after spinal cord injury (SCI) remains a common and thorny problem, influencing the life quality severely. This study aimed to elucidate the reorganization of the primary sensory cortex (S1) and the regulatory mechanism of the lateral parabrachial nucleus (lPBN) in the presence of allodynia or hyperalgesia after left spinal cord hemisection injury (LHS). METHODS Through behavioral tests, we first identified mechanical allodynia and thermal hyperalgesia following LHS. We then applied two-photon microscopy to observe calcium activity in S1 during mechanical or thermal stimulation and long-term spontaneous calcium activity after LHS. By slice patch clamp recording, the electrophysiological characteristics of neurons in lPBN were explored. Finally, exploiting chemogenetic activation or inhibition of the neurons in lPBN, allodynia or hyperalgesia was regulated. RESULTS The calcium activity in left S1 was increased during mechanical stimulation of right hind limb and thermal stimulation of tail, whereas in right S1 it was increased only with thermal stimulation of tail. The spontaneous calcium activity in right S1 changed more dramatically than that in left S1 after LHS. The lPBN was also activated after LHS, and exploiting chemogenetic activation or inhibition of the neurons in lPBN could induce or alleviate allodynia and hyperalgesia in central neuropathic pain. CONCLUSION The neuronal activity changes in S1 are closely related to limb pain, which has accurate anatomical correspondence. After LHS, the spontaneously increased functional connectivity of calcium transient in left S1 is likely causing the mechanical allodynia in right hind limb and increased neuronal activity in bilateral S1 may induce thermal hyperalgesia in tail. This state of allodynia and hyperalgesia can be regulated by lPBN.
Collapse
Affiliation(s)
- Jing Li
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Chao Tian
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Shiyang Yuan
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Zhenyu Yin
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Liangpeng Wei
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Feng Chen
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Xi Dong
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Aili Liu
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
| | - Zhenhuan Wang
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Tongrui Wu
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Chunxiao Tian
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Lin Niu
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
| | - Lei Wang
- Department of PhysiologyZhuhai Campus of Zunyi Medical UniversityZhuhaiChina
| | - Pu Wang
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Wanyu Xie
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Fujiang Cao
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Hui Shen
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
- Innovation Research Institute of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
26
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Deng Y, Tang S, Cheng J, Zhang X, Jing D, Lin Z, Zhou J. Integrated analysis reveals Atf3 promotes neuropathic pain via orchestrating JunB mediated release of inflammatory cytokines in DRG macrophage. Life Sci 2023; 329:121939. [PMID: 37451398 DOI: 10.1016/j.lfs.2023.121939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/08/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
The dorsal root ganglion (DRG) is actively involved in the development of neuropathic pain (NP), serving as an intermediate station for pain signals from the peripheral nervous system to the central nervous system. The mechanism by which DRG is involved in NP regulation is not fully understood. The immune system plays a pivotal role in the physiological and pathological states of the human body. In recent years, the immune system has been thought to play an increasingly important role in the pathogenesis of NP. The immune system plays a key role in pain through specific immune cells and their immune-related genes (IRGs). However, the mechanism by which IRGs of DRG regulate NP action has not been fully elucidated. Here, we performed Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of IRGs in DRG bulk-RNA sequencing data from spared nerve injury (SNI) model mice and found that their IRGs were enriched in many pathways, especially in the immune response pathway. Subsequently, we analyzed single-cell RNA sequencing (scRNA-seq) data from DRGs extracted from the SNI model and identified eight cell populations. Among them, the highest IRG activity was presented in macrophages. Next, we analyzed the scRNA and bulk-sequencing data and deduced five common transcription factors (TFs) from differentially expressed genes (DEGs). The protein-protein interaction (PPI) network suggested that Atf3 and JunB are closely related. In vitro experiments, we verified that the protein and mRNA expressions of Atf3 and JunB were up-regulated in macrophages after lipopolysaccharide (LPS) stimulation. Moreover, the down-regulation of Atf3 reduced the release of inflammatory cytokines and decreased the protein and mRNA expression levels of JunB. The down-regulation of JunB also reduced the release of inflammatory cytokines. Furthermore, overexpression of JunB attenuated the effect of Atf3 down-regulation in reducing the release of inflammatory cytokines. Therefore, we speculated that Atf3 might promote NP through JunB-mediated release of inflammatory factors in DRG macrophages.
Collapse
Affiliation(s)
- Yingdong Deng
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Simin Tang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Jiurong Cheng
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Xiangsheng Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Danqin Jing
- College of Anesthesiology, Shanxi Medical University, Taiyuan, Shanxi Province 030001, China
| | - Ziqiang Lin
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Jun Zhou
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China.
| |
Collapse
|
28
|
Liu H, Lauzadis J, Gunaratna K, Sipple E, Kaczocha M, Puopolo M. Inhibition of T-Type Calcium Channels With TTA-P2 Reduces Chronic Neuropathic Pain Following Spinal Cord Injury in Rats. THE JOURNAL OF PAIN 2023; 24:1681-1695. [PMID: 37169156 DOI: 10.1016/j.jpain.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
Spinal cord injury (SCI)-induced neuropathic pain (SCI-NP) develops in up to 60 to 70% of people affected by traumatic SCI, leading to a major decline in quality of life and increased risk for depression, anxiety, and addiction. Gabapentin and pregabalin, together with antidepressant drugs, are commonly prescribed to treat SCI-NP, but their efficacy is unsatisfactory. The limited efficacy of current pharmacological treatments for SCI-NP likely reflects our limited knowledge of the underlying mechanism(s) responsible for driving the maintenance of SCI-NP. The leading hypothesis in the field supports a major role for spontaneously active injured nociceptors in driving the maintenance of SCI-NP. Recent data from our laboratory provided additional support for this hypothesis and identified the T-type calcium channels as key players in driving the spontaneous activity of SCI-nociceptors, thus providing a rational pharmacological target to treat SCI-NP. To test whether T-type calcium channels contribute to the maintenance of SCI-NP, male and female SCI and sham rats were treated with TTA-P2 (a blocker of T-type calcium channels) to determine its effects on mechanical hypersensitivity (as measured with the von Frey filaments) and spontaneous ongoing pain (as measured with the conditioned place preference paradigm), and compared them to the effects of gabapentin, a blocker of high voltage-activated calcium channels. We found that both TTA-P2 and gabapentin reduced mechanical hypersensitivity in male and females SCI rats, but surprisingly only TTA-P2 reduced spontaneous ongoing pain in male SCI rats. PERSPECTIVES: SCI-induced neuropathic pain, and in particular the spontaneous ongoing pain component, is notoriously very difficult to treat. Our data provide evidence that inhibition of T-type calcium channels reduces spontaneous ongoing pain in SCI rats, supporting a clinically relevant role for T-type channels in the maintenance of SCI-induced neuropathic pain.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Kavindu Gunaratna
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Erin Sipple
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York.
| |
Collapse
|
29
|
Ivasiuk A, Matvieienko M, Kononenko NI, Duzhyy DE, Korogod SM, Voitenko N, Belan P. Diabetes-Induced Amplification of Nociceptive DRG Neuron Output by Upregulation of Somatic T-Type Ca 2+ Channels. Biomolecules 2023; 13:1320. [PMID: 37759720 PMCID: PMC10526307 DOI: 10.3390/biom13091320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
The development of pain symptoms in peripheral diabetic neuropathy (PDN) is associated with the upregulation of T-type Ca2+ channels (T-channels) in the soma of nociceptive DRG neurons. Moreover, a block of these channels in DRG neurons effectively reversed mechanical and thermal hyperalgesia in animal diabetic models, indicating that T-channel functioning in these neurons is causally linked to PDN. However, no particular mechanisms relating the upregulation of T-channels in the soma of nociceptive DRG neurons to the pathological pain processing in PDN have been suggested. Here we have electrophysiologically identified voltage-gated currents expressed in nociceptive DRG neurons and developed a computation model of the neurons, including peripheral and central axons. Simulations showed substantially stronger sensitivity of neuronal excitability to diabetes-induced T-channel upregulation at the normal body temperature compared to the ambient one. We also found that upregulation of somatic T-channels, observed in these neurons under diabetic conditions, amplifies a single action potential invading the soma from the periphery into a burst of multiple action potentials further propagated to the end of the central axon. We have concluded that the somatic T-channel-dependent amplification of the peripheral nociceptive input to the spinal cord demonstrated in this work may underlie abnormal nociception at different stages of diabetes development.
Collapse
Affiliation(s)
- Arsentii Ivasiuk
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Maksym Matvieienko
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Nikolai I. Kononenko
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Dmytro E. Duzhyy
- Department of Sensory Signaling, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine;
| | - Sergiy M. Korogod
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Nana Voitenko
- Department of Biomedicine and Neuroscience, Kyiv Academic University of NAS of Ukraine, 03142 Kyiv, Ukraine
- Research Center, Dobrobut Academy Medical School, 03022 Kyiv, Ukraine
| | - Pavel Belan
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
- Department of Biomedicine and Neuroscience, Kyiv Academic University of NAS of Ukraine, 03142 Kyiv, Ukraine
| |
Collapse
|
30
|
De Schoenmacker I, Mollo A, Scheuren PS, Sirucek L, Brunner F, Schweinhardt P, Curt A, Rosner J, Hubli M. Central sensitization in CRPS patients with widespread pain: a cross-sectional study. PAIN MEDICINE (MALDEN, MASS.) 2023; 24:974-984. [PMID: 36946277 PMCID: PMC10391588 DOI: 10.1093/pm/pnad040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/08/2023] [Accepted: 03/20/2023] [Indexed: 03/23/2023]
Abstract
OBJECTIVE Widespread pain hypersensitivity and enhanced temporal summation of pain (TSP) are commonly reported in patients with complex regional pain syndrome (CRPS) and discussed as proxies for central sensitization. This study aimed to directly relate such signs of neuronal hyperexcitability to the pain phenotype of CRPS patients. METHODS Twenty-one CRPS patients and 20 healthy controls (HC) were recruited. The pain phenotype including spatial pain extent (assessed in % body surface) and intensity were assessed and related to widespread pain hypersensitivity, TSP, and psychological factors. Quantitative sensory testing (QST) was performed in the affected, the contralateral and a remote (control) area. RESULTS CRPS patients showed decreased pressure pain thresholds in all tested areas (affected: t(34) = 4.98, P < .001, contralateral: t(35) = 3.19, P = .005, control: t(31) = 2.65, P = .012). Additionally, patients showed increased TSP in the affected area (F(3,111) = 4.57, P = .009) compared to HC. TSP was even more enhanced in patients with a high compared to a low spatial pain extent (F(3,51) = 5.67, P = .008), suggesting pronounced spinal sensitization in patients with extended pain patterns. Furthermore, the spatial pain extent positively correlated with the Bath Body Perception Disturbance Scale (ρ = 0.491; P = .048). CONCLUSIONS Overall, we provide evidence that the pain phenotype in CRPS, that is, spatial pain extent, might be related to sensitization mechanism within the central nociceptive system. This study points towards central neuronal excitability as a potential therapeutic target in patients with more widespread CRPS.
Collapse
Affiliation(s)
- Iara De Schoenmacker
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Anna Mollo
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Paulina Simonne Scheuren
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Laura Sirucek
- Integrative Spinal Research, Department of Chiropractic Medicine, University Hospital Balgrist, University of Zurich, 8008 Zurich, Switzerland
| | - Florian Brunner
- Physical Medicine and Rheumatology, Balgrist University Hospital, 8008 Zurich, Switzerland
| | - Petra Schweinhardt
- Integrative Spinal Research, Department of Chiropractic Medicine, University Hospital Balgrist, University of Zurich, 8008 Zurich, Switzerland
- Alan Edward Center for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - Armin Curt
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Jan Rosner
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Michèle Hubli
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| |
Collapse
|
31
|
McPherson JG, Bandres MF. Neural population dynamics reveal that motor-targeted intraspinal microstimulation preferentially depresses nociceptive transmission in spinal cord injury-related neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.27.550880. [PMID: 37546721 PMCID: PMC10402167 DOI: 10.1101/2023.07.27.550880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The purpose of this study is to determine whether intraspinal microstimulation (ISMS) intended to enhance voluntary motor output after spinal cord injury (SCI) modulates neural population-level spinal responsiveness to nociceptive sensory feedback. The study was conducted in vivo in three cohorts of rats: neurologically intact, chronic SCI without behavioral signs of neuropathic pain, and chronic SCI with SCI-related neuropathic pain (SCI-NP). Nociceptive sensory feedback was induced by application of graded mechanical pressure to the plantar surface of the hindpaw before, during, and after periods of sub-motor threshold ISMS delivered within the motor pools of the L5 spinal segment. Neural population-level responsiveness to nociceptive feedback was recorded throughout the dorso-ventral extent of the L5 spinal segment using dense multi-channel microelectrode arrays. Whereas motor-targeted ISMS reduced nociceptive transmission across electrodes in neurologically intact animals both during and following stimulation, it was not associated with altered nociceptive transmission in rats with SCI that lacked behavioral signs of neuropathic pain. Surprisingly, nociceptive transmission was reduced both during and following motor-targeted ISMS in rats with SCI-NP, and to an extent comparable to that of neurologically intact animals. The mechanisms underlying the differential anti-nociceptive effects of motor-targeted ISMS are unclear, although they may be related to differences in the intrinsic active membrane properties of spinal neurons across the cohorts. Nevertheless, the results of this study support the notion that it may be possible to purposefully engineer spinal stimulation-based therapies that afford multi-modal rehabilitation benefits, and specifically that it may be possible to do so for the individuals most in need - i.e., those with SCI-related movement impairments and SCI-NP.
Collapse
Affiliation(s)
- Jacob G. McPherson
- Program in Physical Therapy, Washington University School of Medicine
- Department of Anesthesiology, Washington University School of Medicine
- Washington University Pain Center, Washington University School of Medicine
- Program in Neurosciences; Washington University School of Medicine
- Department of Biomedical Engineering; Washington University in St. Louis
| | - Maria F. Bandres
- Program in Physical Therapy, Washington University School of Medicine
- Department of Biomedical Engineering; Washington University in St. Louis
| |
Collapse
|
32
|
Gevers-Montoro C, Puente-Tobares M, Monréal A, Conesa-Buendía FM, Piché M, Ortega-De Mues A. Urinary TNF-α as a potential biomarker for chronic primary low back pain. Front Integr Neurosci 2023; 17:1207666. [PMID: 37449008 PMCID: PMC10336221 DOI: 10.3389/fnint.2023.1207666] [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: 04/19/2023] [Accepted: 06/06/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Over two thirds of individuals with low back pain (LBP) may experience recurrent or persistent symptoms in the long term. Yet, current data do not allow to predict who will develop chronic low back pain and who will recover from an acute episode. Elevated serum levels of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) have been associated with poor recovery and persistent pain following an acute episode of LBP. Inflammatory cytokines may also mediate mechanisms involved in nociplastic pain, and thus, have significant implications in chronic primary low back pain (CPLBP). Methods This study aimed to investigate the potential of urinary TNF-α levels for predicting outcomes and characterizing clinical features of CPLBP patients. Twenty-four patients with CPLBP and 24 sex- and age-matched asymptomatic controls were recruited. Urinary TNF-α concentrations were measured at baseline and after 4 weeks, during which CPLBP patients underwent spinal manipulative therapy (SMT). Results Concentrations of TNF-α were found to be elevated in baseline urine samples of CPLBP patients compared to asymptomatic controls. Moreover, these values differed among patients depending on their pain trajectory. Patients with persistent pain showed higher levels of TNF-α, when compared to those with episodic CPLBP. Furthermore, baseline TNF-α concentrations and their changes after 4 weeks predicted alterations in pain intensity and disability following SMT in patients with CPLBP. Discussion These findings warrant further research on the potential use of urinary TNF-α concentrations as a prognostic biomarker for CPLBP.
Collapse
Affiliation(s)
- Carlos Gevers-Montoro
- Madrid College of Chiropractic – RCU María Cristina, Madrid, Spain
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
- CogNAC Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | | | - Aléxiane Monréal
- Madrid College of Chiropractic – RCU María Cristina, Madrid, Spain
| | | | - Mathieu Piché
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
- CogNAC Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | | |
Collapse
|
33
|
Wang C, Chen R, Zhu X, Zhang X. Suberoylanilide Hydroxamic Acid Ameliorates Pain Sensitization in Central Neuropathic Pain After Spinal Cord Injury via the HDAC5/NEDD4/SCN9A Axis. Neurochem Res 2023:10.1007/s11064-023-03913-z. [DOI: 10.1007/s11064-023-03913-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 04/03/2023]
|
34
|
Zhang T, Liang W, Zhang M, Cui S, Huang X, Ou W, Huang R, Gao J, Jia Z, Zhang S. Daphnetin Improves Neuropathic Pain by Inhibiting the Expression of Chemokines and Inflammatory Factors in the Spinal Cord and Interfering with Glial Cell Polarization. Pharmaceuticals (Basel) 2023; 16:243. [PMID: 37259390 PMCID: PMC9964401 DOI: 10.3390/ph16020243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 08/13/2023] Open
Abstract
Neuropathic pain (NP) is a common pain disease that seriously affects the quality of life and physical and mental health of patients. Daphnetin is extracted from the Daphne giraldii Nitsche and has the structure of 7,8-dihydroxy coumarin. As a natural product, daphnetin displays a wide range of pharmacological activities, such as analgesia and anti-inflammatory activities, but whether it is able to improve NP through anti-inflammatory effects is unknown. Therefore, this paper intends to investigate the mechanism of daphnetin in improving NP rats affected by the intrathecal injection of tumor necrosis factor-α (TNF-α) from the perspective of anti-inflammation. Our results showed that daphnetin significantly improved hyperalgesia in NP rats. Daphnetin inhibited the activation and polarization of glial cells and neurons in the spinal cord of NP rats and reduced the expression of mRNA and protein of inflammatory factors and chemokine pairs in the spinal cord. Daphnetin inhibited the polarization of human microglia cell 3 (HMC3) cells and human glioma cells (U251) cells toward M1 microglia and A1 astrocytes, respectively, and induced the conversion of M1 microglia and A1 astrocytes to M2 microglia and A2 astrocytes, respectively. In conclusion, daphnetin ameliorates NP by inhibiting the expression of inflammatory factors and chemokines and the polarization of glial cells in the spinal cord of NP rats. This study provides a theoretical basis for the treatment of NP with daphnetin to expand the clinical application of daphnetin.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Shuofeng Zhang
- Department of Pharmacology of Traditional Chinese Medicine, College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
35
|
Jergova S, Dugan EA, Sagen J. Attenuation of SCI-Induced Hypersensitivity by Intensive Locomotor Training and Recombinant GABAergic Cells. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010084. [PMID: 36671656 PMCID: PMC9854592 DOI: 10.3390/bioengineering10010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
The underlying mechanisms of spinal cord injury (SCI)-induced chronic pain involve dysfunctional GABAergic signaling and enhanced NMDA signaling. Our previous studies showed that SCI hypersensitivity in rats can be attenuated by recombinant rat GABAergic cells releasing NMDA blocker serine-histogranin (SHG) and by intensive locomotor training (ILT). The current study combines these approaches and evaluates their analgesic effects on a model of SCI pain in rats. Cells were grafted into the spinal cord at 4 weeks post-SCI to target the chronic pain, and ILT was initiated 5 weeks post-SCI. The hypersensitivity was evaluated weekly, which was followed by histological and biochemical assays. Prolonged effects of the treatment were evaluated in subgroups of animals after we discontinued ILT. The results show attenuation of tactile, heat and cold hypersensitivity in all of the treated animals and reduced levels of proinflammatory cytokines IL1β and TNFα in the spinal tissue and CSF. Animals with recombinant grafts and ILT showed the preservation of analgesic effects even during sedentary periods when the ILT was discontinued. Retraining helped to re-establish the effect of long-term training in all of the groups, with the greatest impact being in animals with recombinant grafts. These findings suggest that intermittent training in combination with cell therapy might be an efficient approach to manage chronic pain in SCI patients.
Collapse
|
36
|
Yeo JH, Roh DH. The mTOR inhibitor rapamycin suppresses trigeminal neuropathic pain and p-MKK4/p-p38 mitogen-activated protein kinase-mediated microglial activation in the trigeminal nucleus caudalis of mice with infraorbital nerve injury. Front Mol Neurosci 2023; 16:1172366. [PMID: 37122619 PMCID: PMC10140572 DOI: 10.3389/fnmol.2023.1172366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Neuropathic pain caused by trigeminal nerve injury is a typical refractory orofacial chronic pain accompanied by the development of hyperalgesia and allodynia. We previously demonstrated that the mammalian target of rapamycin (mTOR) inhibitor rapamycin suppressed orofacial formalin injection-induced nociception; however, the underlying mechanism is unclear, and it is unknown whether it can reduce trigeminal neuropathic pain. In mice, left infraorbital nerve and partial nerve ligation (ION-pNL) was performed using a silk suture (8-0). Fourteen days after surgery, neuropathic pain behavior was examined on a whisker pad and rapamycin (0.1, 0.3, and 1.0 mg/kg) was administered intraperitoneally. Mechanical and cold sensitivities in the orofacial region were quantified using von Frey filaments and acetone solution, respectively. Changes in mTOR and related proteins, such as p-MKK3/6, p-MKK4, p-JNK, p-ERK, p-p38 MAPK, GFAP, and Iba-1, in the trigeminal nucleus caudalis (TNC) or the trigeminal ganglia (TG) tissues were examined via western blot analysis or immunohistochemistry. Mice demonstrated significant mechanical and cold allodynia 2 weeks following ION-pNL injury, both of which were significantly reduced 1 h after the administration of high-dose rapamycin (1.0 mg/kg). In the TG tissue, ION-pNL surgery or rapamycin treatment did not change p-mTOR and p-4EBP1, but rapamycin reduced the increase of p-S6 and S6 induced by ION-pNL. In the TNC tissue, neither ION-pNL surgery nor rapamycin treatment altered p-mTOR, p-S6, and p-4EBP1 expressions, whereas rapamycin significantly decreased the ION-pNL-induced increase in Iba-1 expression. In addition, rapamycin suppressed the increase in p-p38 MAPK and p-MKK4 expressions but not p-MKK3/6 expression. Moreover, p-p38 MAPK-positive cells were colocalized with increased Iba-1 in the TNC. Our findings indicate that rapamycin treatment reduces both mechanical and cold orofacial allodynia in mice with trigeminal neuropathic pain, which is closely associated with the modulation of p-MKK4/p-p38 MAPK-mediated microglial activation in the TNC.
Collapse
|
37
|
Zhang K, Li P, Jia Y, Liu M, Jiang J. Non-coding RNA and n6-methyladenosine modification play crucial roles in neuropathic pain. Front Mol Neurosci 2022; 15:1002018. [PMID: 36466810 PMCID: PMC9716653 DOI: 10.3389/fnmol.2022.1002018] [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] [Received: 07/24/2022] [Accepted: 10/28/2022] [Indexed: 07/30/2023] Open
Abstract
After peripheral nerve injury, pain signals are transmitted from primary sensory neurons in the dorsal root ganglion (DRG) to the central nervous system. Epigenetic modification affects neuropathic pain through alterations in the gene expression in pain-related areas and glial cell activation. Recent studies have shown that non-coding RNA and n6-methyladenosine (m6A) methylation modification play pivotal regulatory roles in the occurrence and maintenance of neuropathic pain. Dysregulation of the RNA m6A level via dynamic changes in methyltransferase and demethylase after central or peripheral nerve injury commonly regulates pain-associated genes, contributing to the induction and maintenance of neuropathic pain. The dynamic process has significant implications for the development and maintenance of neuropathic pain. However, the underlying mechanisms by which non-coding RNA and m6A RNA modification regulate neuropathic pain are not well-characterized. This article elucidates the multiple mechanisms of non-coding RNA and m6A methylation in the context of neuropathic pain, and summarizes its potential functions as well as recent advances.
Collapse
|
38
|
Lütolf R, Rosner J, Curt A, Hubli M. Indicators of central sensitization in chronic neuropathic pain after spinal cord injury. Eur J Pain 2022; 26:2162-2175. [PMID: 36008094 PMCID: PMC9826442 DOI: 10.1002/ejp.2028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 08/01/2022] [Accepted: 08/20/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Central sensitization is considered a key mechanism underlying neuropathic pain (NP) after spinal cord injury (SCI). METHODS Two novel proxies for central sensitization were investigated in thoracic SCI subjects with (SCI-NP) and without NP (SCI-nonNP) compared to healthy controls (HC). Specifically, temporal summation of pain (TSP) was investigated by examining pain ratings during a 2-min tonic heat application to the volar forearm. Additionally, palmar heat-induced sympathetic skin responses (SSR) were recorded in order to reveal changes in pain-autonomic interaction above the lesion level. Pain extent was assessed as the percentage of the body area and the number of body regions being affected by NP. RESULTS Enhanced TSP was observed in SCI-NP (+66%) compared to SCI-nonNP (-75%, p = 0.009) and HC (-59%, p = 0.021). In contrast, no group differences were found (p = 0.685) for SSR habituation. However, pain extent in SCI-NP was positively correlated with deficient SSR habituation (body area: r = 0.561, p = 0.024; body regions: r = 0.564, p = 0.023). CONCLUSIONS These results support the value of TSP and heat-induced SSRs as proxies for central sensitization in widespread neuropathic pain syndromes after SCI. Measures of pain-autonomic interaction emerged as a promising tool for the objective investigation of sensitized neuronal states in chronic pain conditions. SIGNIFICANCE We present two surrogate readouts for central sensitization in neuropathic pain following SCI. On the one hand, temporal summation of tonic heat pain is enhanced in subjects with neuropathic pain. On the other hand, pain-autonomic interaction reveals potential advanced measures in chronic pain, as subjects with a high extent of neuropathic pain showed diminished habituation of pain-induced sympathetic measures. A possible implication for clinical practice is constituted by an improved assessment of neuronal hyperexcitability potentially enabling mechanism-based treatment.
Collapse
Affiliation(s)
- Robin Lütolf
- Spinal Cord Injury CenterBalgrist University Hospital, University of ZurichZurichSwitzerland
| | - Jan Rosner
- Spinal Cord Injury CenterBalgrist University Hospital, University of ZurichZurichSwitzerland,Department of NeurologyUniversity Hospital Bern, Inselspital, University of BernBernSwitzerland
| | - Armin Curt
- Spinal Cord Injury CenterBalgrist University Hospital, University of ZurichZurichSwitzerland
| | - Michèle Hubli
- Spinal Cord Injury CenterBalgrist University Hospital, University of ZurichZurichSwitzerland
| |
Collapse
|
39
|
Lütolf R, De Schoenmacker I, Rosner J, Sirucek L, Schweinhardt P, Curt A, Hubli M. Anti- and Pro-Nociceptive mechanisms in neuropathic pain after human spinal cord injury. Eur J Pain 2022; 26:2176-2187. [PMID: 36000307 PMCID: PMC9826499 DOI: 10.1002/ejp.2029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/01/2022] [Accepted: 08/20/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Deficient endogenous pain modulation and increased nociceptive excitability are key features of central sensitization and can be assessed in humans by conditioned pain modulation (CPM, anti-nociceptive) and temporal summation of pain (TSP, pro-nociceptive), respectively. This study aimed to investigate these measures as proxies for central sensitization in subjects with chronic neuropathic pain (NP) after spinal cord injury (SCI). METHODS In paraplegic subjects with NP (SCI-NP; n = 17) and healthy controls (HC; n = 17), parallel and sequential sham-controlled CPM paradigms were performed using pressure pain threshold at the hand, that is, above lesion level, as test stimulus. The conditioning stimulus was a noxious cold (verum) or lukewarm water bath (sham) applied contralaterally. Regarding pro-nociceptive mechanisms, a TSP protocol with individually-adjusted pressure pain stimuli at the thenar eminence was used. CPM and TSP magnitudes were related to intensity and spatial extent of spontaneous NP. RESULTS Neither the parallel nor sequential sham-controlled CPM paradigm showed any significant inhibition of above-level pressure pain thresholds for SCI-NP or HC. Accordingly, no group difference in CPM capacity was found, however, subjects with more intense spontaneous NP showed lower inhibitory CPM capacity. TSP was observed for both groups but was not enhanced in SCI-NP. CONCLUSIONS Our results do not support altered above-level anti- or pro-nociceptive mechanisms in SCI-NP compared with HC; however, they also highlight the relevance of spontaneous NP intensity with regards to the capacity of endogenous pain modulation in SCI subjects. SIGNIFICANCE Central sensitization encompasses deficient endogenous pain modulation and increased nociceptive excitability. These two mechanisms can be assessed in humans by conditioned pain modulation and temporal summation of pain, respectively. Our data demonstrates a lack of descending pain inhibition only in subjects with severe neuropathic pain which may hint towards central sensitization at spinal and/or supra-spinal levels. Disentangling the mechanisms of endogenous pain modulation and neuronal hyperexcitability might improve mechanism-based treatment of neuropathic pain in subjects with spinal cord injury.
Collapse
Affiliation(s)
- Robin Lütolf
- Spinal Cord Injury Center, Balgrist University HospitalUniversity of ZurichZurichSwitzerland
| | - Iara De Schoenmacker
- Spinal Cord Injury Center, Balgrist University HospitalUniversity of ZurichZurichSwitzerland
| | - Jan Rosner
- Spinal Cord Injury Center, Balgrist University HospitalUniversity of ZurichZurichSwitzerland,Department of NeurologyUniversity Hospital Bern, Inselspital, University of BernBernSwitzerland
| | - Laura Sirucek
- Integrative Spinal Research, Department of Chiropractic MedicineBalgrist University Hospital, University of ZurichZurichSwitzerland
| | - Petra Schweinhardt
- Integrative Spinal Research, Department of Chiropractic MedicineBalgrist University Hospital, University of ZurichZurichSwitzerland
| | - Armin Curt
- Spinal Cord Injury Center, Balgrist University HospitalUniversity of ZurichZurichSwitzerland
| | - Michèle Hubli
- Spinal Cord Injury Center, Balgrist University HospitalUniversity of ZurichZurichSwitzerland
| |
Collapse
|
40
|
The Impact of Activity-Based Interventions on Neuropathic Pain in Experimental Spinal Cord Injury. Cells 2022; 11:cells11193087. [PMID: 36231048 PMCID: PMC9563089 DOI: 10.3390/cells11193087] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Physical activity-based rehabilitative interventions represent the main treatment concept for people suffering from spinal cord injury (SCI). The role such interventions play in the relief of neuropathic pain (NP) states is emerging, along with underlying mechanisms resulting in SCI-induced NP (SCI-NP). Animal models have been used to investigate the benefits of activity-based interventions (ABI), such as treadmill training, wheel running, walking, swimming, and bipedal standing. These activity-based paradigms have been shown to modulate inflammatory-related alterations as well as induce functional and structural changes in the spinal cord gray matter circuitry correlated with pain behaviors. Thus far, the research available provides an incomplete picture of the cellular and molecular pathways involved in this beneficial effect. Continued research is essential for understanding how such interventions benefit SCI patients suffering from NP and allow the development of individualized rehabilitative therapies. This article reviews preclinical studies on this specific topic, goes over mechanisms involved in SCI-NP in relation to ABI, and then discusses the effectiveness of different activity-based paradigms as they relate to different forms, intensity, initiation times, and duration of ABI. This article also summarizes the mechanisms of respective interventions to ameliorate NP after SCI and provides suggestions for future research directions.
Collapse
|
41
|
7,8-Dihydroxyflavone accelerates recovery of Brown-Sequard syndrome in adult female rats with spinal cord lateral hemisection. Biomed Pharmacother 2022; 153:113397. [DOI: 10.1016/j.biopha.2022.113397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
|
42
|
Baron-Flores V, Diaz-Ruiz A, Manzanares J, Rios C, Burelo M, Jardon-Guadarrama G, Martínez-Cárdenas MDLÁ, Mata-Bermudez A. Cannabidiol attenuates hypersensitivity and oxidative stress after traumatic spinal cord injury in rats. Neurosci Lett 2022; 788:136855. [PMID: 36028005 DOI: 10.1016/j.neulet.2022.136855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/08/2022] [Accepted: 08/19/2022] [Indexed: 10/15/2022]
Abstract
Neuropathic pain (NP) arises as a direct consequence of traumatic spinal cord injury (SCI), which leads to devastating consequences for people suffering from this condition since no specific treatment has been defined. One relevant mechanism in generating painful stimuli involves the direct participation of reactive oxygen species (ROS) at the cellular and subcellular levels. Cannabidiol (CBD) is one of the two most crucial cannabinoid components of the cannabis plant and has been proposed as a potential treatment for NP. Its antioxidant, neuroprotective and anti-inflammatory properties have been documented. However, there is insufficient evidence regarding CBD as treatment of NP induced by SCI or the mechanisms that underlie this effect. In this study, we evaluated the antinociceptive effect of CBD as an acute treatment after the nociceptive behaviors characteristic of NP were established (hypersensitivity threshold and hypersensitivity response). Furthermore, the participation of oxidative stress was determined by lipid peroxidation (LP) and glutathione concentration (GSH) in female Wistar rats with SCI. Acute treatment with CBD (2.5-20 mg/kg, i.p.) decreased nociceptive behaviors in a dose-dependent manner, decreased LP, and increased GSH concentration in injured tissue 15 days after injury. The findings of this study suggest that the antinociceptive effect induced by CBD is regulated by reducing oxidative stress by decreasing the LP and increasing the concentration of antioxidant (GSH) defenses.
Collapse
Affiliation(s)
- Verónica Baron-Flores
- Laboratorio de Neurofarmacología Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, Mexico
| | - Araceli Diaz-Ruiz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain; Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Camilo Rios
- Laboratorio de Neurofarmacología Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, Mexico; Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Masha Burelo
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Gustavo Jardon-Guadarrama
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, Mexico
| | | | - Alfonso Mata-Bermudez
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, Mexico.
| |
Collapse
|
43
|
The Role of Inflammation, Hypoxia, and Opioid Receptor Expression in Pain Modulation in Patients Suffering from Obstructive Sleep Apnea. Int J Mol Sci 2022; 23:ijms23169080. [PMID: 36012341 PMCID: PMC9409023 DOI: 10.3390/ijms23169080] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 12/18/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a relatively common disease in the general population. Besides its interaction with many comorbidities, it can also interact with potentially painful conditions and modulate its course. The association between OSA and pain modulation has recently been a topic of concern for many scientists. The mechanism underlying OSA-related pain connection has been linked with different pathophysiological changes in OSA and various pain mechanisms. Furthermore, it may cause both chronic and acute pain aggravation as well as potentially influencing the antinociceptive mechanism. Characteristic changes in OSA such as nocturnal hypoxemia, sleep fragmentation, and systemic inflammation are considered to have a curtailing impact on pain perception. Hypoxemia in OSA has been proven to have a significant impact on increased expression of proinflammatory cytokines influencing the hyperalgesic priming of nociceptors. Moreover, hypoxia markers by themselves are hypothesized to modulate intracellular signal transduction in neurons and have an impact on nociceptive sensitization. Pain management in patients with OSA may create problems arousing from alterations in neuropeptide systems and overexpression of opioid receptors in hypoxia conditions, leading to intensification of side effects, e.g., respiratory depression and increased opioid sensitivity for analgesic effects. In this paper, we summarize the current knowledge regarding pain and pain treatment in OSA with a focus on molecular mechanisms leading to nociceptive modulation.
Collapse
|
44
|
Eller OC, Stair RN, Neal C, Rowe PS, Nelson-Brantley J, Young EE, Baumbauer KM. Comprehensive phenotyping of cutaneous afferents reveals early-onset alterations in nociceptor response properties, release of CGRP, and hindpaw edema following spinal cord injury. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 12:100097. [PMID: 35756343 PMCID: PMC9218836 DOI: 10.1016/j.ynpai.2022.100097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) is a complex syndrome that has profound effects on patient well-being, including the development of medically-resistant chronic pain. The mechanisms underlying SCI pain have been the subject of thorough investigation but remain poorly understood. While the majority of the research has focused on changes occurring within and surrounding the site of injury in the spinal cord, there is now a consensus that alterations within the peripheral nervous system, namely sensitization of nociceptors, contribute to the development and maintenance of chronic SCI pain. Using an ex vivo skin/nerve/DRG/spinal cord preparation to characterize afferent response properties following SCI, we found that SCI increased mechanical and thermal responding, as well as the incidence of spontaneous activity (SA) and afterdischarge (AD), in below-level C-fiber nociceptors 24 hr following injury relative to naïve controls. Interestingly, the distribution of nociceptors that exhibit SA and AD are not identical, and the development of SA was observed more frequently in nociceptors with low heat thresholds, while AD was found more frequently in nociceptors with high heat thresholds. We also found that SCI resulted in hindpaw edema and elevated cutaneous calcitonin gene-related peptide (CGRP) concentration that were not observed in naïve mice. These results suggest that SCI causes a rapidly developing nociceptor sensitization and peripheral inflammation that may contribute to the early emergence and persistence of chronic SCI pain.
Collapse
Affiliation(s)
- Olivia C. Eller
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Rena N. Stair
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Christopher Neal
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Peter S.N. Rowe
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, United States
- The Kidney Institute & Division of Nephrology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jennifer Nelson-Brantley
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Erin E. Young
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
- Center for Advancement in Managing Pain, School of Nursing, University of Connecticut, Storrs, CT, United States
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT, United States
- Department of Neuroscience, UConn Health, Farmington, CT, United States
| | - Kyle M. Baumbauer
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
- Center for Advancement in Managing Pain, School of Nursing, University of Connecticut, Storrs, CT, United States
- Department of Neuroscience, UConn Health, Farmington, CT, United States
| |
Collapse
|
45
|
North RY, Odem MA, Li Y, Tatsui CE, Cassidy RM, Dougherty PM, Walters ET. Electrophysiological Alterations Driving Pain-Associated Spontaneous Activity in Human Sensory Neuron Somata Parallel Alterations Described in Spontaneously Active Rodent Nociceptors. THE JOURNAL OF PAIN 2022; 23:1343-1357. [PMID: 35292377 PMCID: PMC9357108 DOI: 10.1016/j.jpain.2022.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 06/10/2023]
Abstract
Neuropathic pain in rodents can be driven by ectopic spontaneous activity (SA) generated by sensory neurons in dorsal root ganglia (DRG). The recent demonstration that SA in dissociated human DRG neurons is associated with reported neuropathic pain in patients enables a detailed comparison of pain-linked electrophysiological alterations driving SA in human DRG neurons to alterations that distinguish SA in nociceptors from SA in low-threshold mechanoreceptors (LTMRs) in rodent neuropathy models. Analysis of recordings from dissociated somata of patient-derived DRG neurons showed that SA and corresponding pain in both sexes were significantly associated with the three functional electrophysiological alterations sufficient to generate SA in the absence of extrinsic depolarizing inputs. These include enhancement of depolarizing spontaneous fluctuations of membrane potential (DSFs), which were analyzed quantitatively for the first time in human DRG neurons. The functional alterations were indistinguishable from SA-driving alterations reported for nociceptors in rodent chronic pain models. Irregular, low-frequency DSFs in human DRG neurons closely resemble DSFs described in rodent nociceptors while differing substantially from the high-frequency sinusoidal oscillations described in rodent LTMRs. These findings suggest that conserved physiological mechanisms of SA in human nociceptor somata can drive neuropathic pain despite documented cellular differences between human and rodent DRG neurons. PERSPECTIVE: Electrophysiological alterations in human sensory neurons associated with patient-reported neuropathic pain include all three of the functional alterations that logically can promote spontaneous activity. The similarity of distinctively altered spontaneous depolarizations in human DRG neurons and rodent nociceptors suggests that spontaneously active human nociceptors can persistently promote neuropathic pain in patients.
Collapse
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, Texas
| | - Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Claudio Esteves Tatsui
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ryan M Cassidy
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Patrick M Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas..
| |
Collapse
|
46
|
Bavencoffe A, Spence EA, Zhu MY, Garza-Carbajal A, Chu KE, Bloom OE, Dessauer CW, Walters ET. Macrophage Migration Inhibitory Factor (MIF) Makes Complex Contributions to Pain-Related Hyperactivity of Nociceptors after Spinal Cord Injury. J Neurosci 2022; 42:5463-5480. [PMID: 35610050 PMCID: PMC9270921 DOI: 10.1523/jneurosci.1133-21.2022] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 02/08/2023] Open
Abstract
Neuropathic pain is a major, inadequately treated challenge for people with spinal cord injury (SCI). While SCI pain mechanisms are often assumed to be in the CNS, rodent studies have revealed mechanistic contributions from primary nociceptors. These neurons become chronically hyperexcitable after SCI, generating ongoing electrical activity that promotes ongoing pain. A major question is whether extrinsic chemical signals help to drive ongoing electrical activity after SCI. People living with SCI exhibit acute and chronic elevation of circulating levels of macrophage migration inhibitory factor (MIF), a cytokine implicated in preclinical pain models. Probable nociceptors isolated from male rats and exposed to an MIF concentration reported in human plasma (1 ng/ml) showed hyperactivity similar to that induced by SCI, although, surprisingly, a 10-fold higher concentration failed to increase excitability. Conditioned behavioral aversion to a chamber associated with peripheral MIF injection suggested that MIF stimulates affective pain. A MIF inhibitor, Iso-1, reversed SCI-induced hyperexcitability. Unlike chronic SCI-induced hyperexcitability, acute MIF-induced hyperexcitability was only partially abrogated by inhibiting ERK signaling. Unexpectedly, MIF concentrations that induced hyperactivity in nociceptors from naive animals, after SCI induced a long-lasting conversion from a highly excitable nonaccommodating type to a rapidly accommodating, hypoexcitable type, possibly as a homeostatic response to prolonged depolarization. Treatment with conditioned medium from cultures of DRG cells obtained after SCI was sufficient to induce MIF-dependent hyperactivity in neurons from naive rats. Thus, changes in systemic and DRG levels of MIF may help to maintain SCI-induced nociceptor hyperactivity that persistently promotes pain.SIGNIFICANCE STATEMENT Chronic neuropathic pain is a major challenge for people with spinal cord injury (SCI). Pain can drastically impair quality of life, and produces substantial economic and social burdens. Available treatments, including opioids, remain inadequate. This study shows that the cytokine macrophage migration inhibitory factor (MIF) can induce pain-like behavior and plays an important role in driving persistent ongoing electrical activity in injury-detecting sensory neurons (nociceptors) in a rat SCI model. The results indicate that SCI produces an increase in MIF release within sensory ganglia. Low MIF levels potently excite nociceptors, but higher levels trigger a long-lasting hypoexcitable state. These findings suggest that therapeutic targeting of MIF in neuropathic pain states may reduce pain and sensory dysfunction by curbing nociceptor hyperactivity.
Collapse
Affiliation(s)
- Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas 77030
| | - Emily A Spence
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas 77030
| | - Michael Y Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas 77030
| | - Anibal Garza-Carbajal
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas 77030
| | - Kerry E Chu
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas 77030
| | - Ona E Bloom
- Laboratory of Spinal Cord Injury Research, Feinstein Institutes for Medical Research, Manhasset, New York 11030
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas 77030
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas 77030
| |
Collapse
|
47
|
Miranpuri GS, Bali P, Nguyen J, Kim JJ, Modgil S, Mehra P, Buttar S, Brown G, Yutuc N, Singh H, Wood A, Singh J, Anand A. Role of Microglia and Astrocytes in Spinal Cord Injury Induced Neuropathic Pain. Ann Neurosci 2022; 28:219-228. [PMID: 35341227 PMCID: PMC8948321 DOI: 10.1177/09727531211046367] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/03/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Spinal cord injuries incite varying degrees of symptoms in patients, ranging
from weakness and incoordination to paralysis. Common amongst spinal cord
injury (SCI) patients, neuropathic pain (NP) is a debilitating medical
condition. Unfortunately, there remain many clinical impediments in treating
NP because there is a lack of understanding regarding the mechanisms behind
SCI-induced NP (SCINP). Given that more than 450,000 people in the United
States alone suffer from SCI, it is unsatisfactory that current treatments
yield poor results in alleviating and treating NP. Summary: In this review, we briefly discussed the models of SCINP along with the
mechanisms of NP progression. Further, current treatment modalities are
herein explored for SCINP involving pharmacological interventions targeting
glia cells and astrocytes. Key message: The studies presented in this review provide insight for new directions
regarding SCINP alleviation. Given the severity and incapacitating effects
of SCINP, it is imperative to study the pathways involved and find new
therapeutic targets in coordination with stem cell research, and to develop
a new gold-standard in SCINP treatment.
Collapse
Affiliation(s)
- Gurwattan S Miranpuri
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Parul Bali
- Department of Biological Sciences, Indian Institute of Science Education & Research Mohali, India
| | - Justyn Nguyen
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Jason J Kim
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Shweta Modgil
- Neuroscience research lab, Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Priya Mehra
- Neuroscience research lab, Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.,Department of Biotechnology, Panjab University, Chandigarh, India
| | - Seah Buttar
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Greta Brown
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Noemi Yutuc
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Harpreet Singh
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Aleksandar Wood
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Jagtar Singh
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Akshay Anand
- Neuroscience research lab, Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.,CCRYN- Collaborative Centre for Mind Body Intervention through Yoga.,Centre of Phenomenology and Cognitive Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
48
|
Dietz V, Knox K, Moore S, Roberts N, Corona KK, Dulin JN. Dorsal horn neuronal sparing predicts the development of at-level mechanical allodynia following cervical spinal cord injury in mice. Exp Neurol 2022; 352:114048. [PMID: 35304102 DOI: 10.1016/j.expneurol.2022.114048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/03/2022] [Accepted: 03/13/2022] [Indexed: 11/04/2022]
Abstract
Spinal cord injury (SCI) frequently results in immediate and sustained neurological dysfunction, including intractable neuropathic pain in approximately 60-80% of individuals. SCI induces immediate mechanical damage to spinal cord tissue followed by a period of secondary injury in which tissue damage is further propagated, contributing to the development of anatomically unique lesions. Variability in lesion size and location influences the degree of motor and sensory dysfunction incurred by an individual. We predicted that variability in lesion parameters may also explain why some, but not all, experimental animals develop mechanical sensitivity after SCI. To characterize the relationship of lesion anatomy to mechanical allodynia, we utilized a mouse cervical hemicontusion model of SCI that has been shown to lead to the development and persistence of mechanical allodynia in the ipsilateral forelimb after injury. At four weeks post-SCI, the numbers and locations of surviving neurons were quantified along with total lesion volume and nociceptive fiber sprouting. We found that the subset of animals exhibiting mechanical allodynia had significantly increased neuronal sparing in the ipsilateral dorsal horn around the lesion epicenter compared to animals that did not exhibit mechanical allodynia. Additionally, we failed to observe significant differences between groups in nociceptive fiber density in the dorsal horn around the lesion epicenter. Notably, we found that impactor probe displacement upon administration of the SCI surgery was significantly lower in sensitive animals compared with not-sensitive animals. Together, our data indicate that lesion severity negatively correlates with the manifestation of at-level mechanical hypersensitivity and suggests that sparing of dorsal horn neurons may be required for the development of neuropathic pain.
Collapse
Affiliation(s)
- Valerie Dietz
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Katelyn Knox
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Sherilynne Moore
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Nolan Roberts
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | | | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX 77843, USA; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
49
|
Liu X, Liu BL, Yang Q, Zhou X, Tang SJ. Microglial ablation does not affect opioid-induced hyperalgesia in rodents. Pain 2022; 163:508-517. [PMID: 34433775 PMCID: PMC8678393 DOI: 10.1097/j.pain.0000000000002376] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Opioids are the frontline analgesics in pain management. However, chronic use of opioid analgesics causes paradoxical pain that contributes to the decrease of their efficacy in pain control and the escalation of dose in long-term management of pain. The underling pathogenic mechanism is not well understood. Microglia have been commonly believed to play a critical role in the expression of opioid-induced hyperalgesia in animal models. We performed microglial ablation experiments using either genetic (CD11b-diphtheria toxin receptor transgenic mouse) or pharmacological (colony-stimulating factor-1 receptor inhibitor PLX5622) approaches. Surprisingly, ablating microglia using these specific and effective approaches did not cause detectable impairment in the expression of hyperalgesia induced by morphine. We confirmed this conclusion with a behavioral test of mechanical and thermal hyperalgesia, in male and female mice, and with different species (mouse and rat). These findings raise caution about the widely assumed contribution of microglia to the development of opioid-induced hyperalgesia.
Collapse
Affiliation(s)
- Xin Liu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bo-Long Liu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Urology, The Third Affiliated Hospital of the Sun Yat-Sen University, Guangzhou, China
| | - Qing Yang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xiangfu Zhou
- Department of Urology, The Third Affiliated Hospital of the Sun Yat-Sen University, Guangzhou, China
| | - Shao-Jun Tang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
50
|
Jergova S, Martinez H, Hernandez M, Schachner B, Gross S, Sagen J. Development of a Phantom Limb Pain Model in Rats: Behavioral and Histochemical Evaluation. FRONTIERS IN PAIN RESEARCH 2021; 2:675232. [PMID: 35295448 PMCID: PMC8915728 DOI: 10.3389/fpain.2021.675232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Therapeutic strategies targeting phantom limb pain (PLP) provide inadequate pain relief; therefore, a robust and clinically relevant animal model is necessary. Animal models of PLP are based on a deafferentation injury followed by autotomy behavior. Clinical studies have shown that the presence of pre-amputation pain increases the risk of developing PLP. In the current study, we used Sprague-Dawley male rats with formalin injections or constriction nerve injury at different sites or time points prior to axotomy to mimic clinical scenarios of pre-amputation inflammatory and neuropathic pain. Animals were scored daily for PLP autotomy behaviors, and several pain-related biomarkers were evaluated to discover possible underlying pathological changes. Majority displayed some degree of autotomy behavior following axotomy. Injury prior to axotomy led to more severe PLP behavior compared to animals without preceding injury. Autotomy behaviors were more directed toward the pretreatment insult origin, suggestive of pain memory. Increased levels of IL-1β in cerebrospinal fluid and enhanced microglial responses and the expression of NaV1.7 were observed in animals displaying more severe PLP outcomes. Decreased expression of GAD65/67 was consistent with greater PLP behavior. This study provides a preclinical basis for future understanding and treatment development in the management of PLP.
Collapse
|