1
|
Mugan D, Vuong QC, Dietz BE, Obara I. Characterization of preclinical models to investigate spinal cord stimulation for neuropathic pain: a systematic review and meta-analysis. Pain Rep 2025; 10:e1228. [PMID: 39816902 PMCID: PMC11732658 DOI: 10.1097/pr9.0000000000001228] [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] [Received: 09/11/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 01/18/2025] Open
Abstract
Despite advancements in preclinical and clinical spinal cord stimulation (SCS) research, the mechanisms of SCS action remain unclear. This may result from challenges in translatability of findings between species. Our systematic review (PROSPERO: CRD42023457443) aimed to comprehensively characterize the important translational components of preclinical SCS models, including stimulating elements and stimulation specifications. Databases (Embase, PubMed, Web of Science, and WikiStim) were searched on October 5, 2023, identifying 78 studies meeting the search criteria. We conducted a post hoc meta-analysis, including subgroup analyses and meta-regression, to assess SCS efficacy on mechanical hypersensitivity in rats subjected to neuropathic pain. Although monopolar electrodes were predominantly used as stimulating elements until 2013, quadripolar paddle and cylindrical leads gained recent popularity. Most research was conducted using 50 Hz and 200 µs stimulation. Motor threshold (MT) estimation was the predominant strategy to determine SCS intensity, which was set to 71.9% of MT on average. Our analysis revealed a large effect size for SCS (Hedge g = 1.13, 95% CI: [0.93, 1.32]) with similar magnitudes of effect between conventional (≤100 Hz) and nonconventional SCS paradigms while sham SCS had nonsignificant effect size. In addition, different stimulation intensity, frequency, and electrode design did not affect effect size. The risk of bias was assessed using Systematic Review Centre for Laboratory animal Experimentation criteria and was unclear, and only the frequency subgroup analysis showed publication bias. In summary, our review characterizes the critical components of preclinical SCS models and provides recommendations to improve reproducibility and translatability, thereby advancing the scientific foundation for SCS research.
Collapse
Affiliation(s)
- Dave Mugan
- School of Pharmacy, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Saluda Medical Europe Ltd, Harrogate, United Kingdom
| | - Quoc C. Vuong
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- School of Psychology, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Birte E. Dietz
- School of Pharmacy, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Saluda Medical Europe Ltd, Harrogate, United Kingdom
| | - Ilona Obara
- School of Pharmacy, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| |
Collapse
|
2
|
Krsek A, Ostojic L, Zivalj D, Baticic L. Navigating the Neuroimmunomodulation Frontier: Pioneering Approaches and Promising Horizons-A Comprehensive Review. Int J Mol Sci 2024; 25:9695. [PMID: 39273641 PMCID: PMC11396210 DOI: 10.3390/ijms25179695] [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: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The research in neuroimmunomodulation aims to shed light on the complex relationships that exist between the immune and neurological systems and how they affect the human body. This multidisciplinary field focuses on the way immune responses are influenced by brain activity and how neural function is impacted by immunological signaling. This provides important insights into a range of medical disorders. Targeting both brain and immunological pathways, neuroimmunomodulatory approaches are used in clinical pain management to address chronic pain. Pharmacological therapies aim to modulate neuroimmune interactions and reduce inflammation. Furthermore, bioelectronic techniques like vagus nerve stimulation offer non-invasive control of these systems, while neuromodulation techniques like transcranial magnetic stimulation modify immunological and neuronal responses to reduce pain. Within the context of aging, neuroimmunomodulation analyzes the ways in which immunological and neurological alterations brought on by aging contribute to cognitive decline and neurodegenerative illnesses. Restoring neuroimmune homeostasis through strategies shows promise in reducing age-related cognitive decline. Research into mood disorders focuses on how immunological dysregulation relates to illnesses including anxiety and depression. Immune system fluctuations are increasingly recognized for their impact on brain function, leading to novel treatments that target these interactions. This review emphasizes how interdisciplinary cooperation and continuous research are necessary to better understand the complex relationship between the neurological and immune systems.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Leona Ostojic
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Dorotea Zivalj
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
3
|
Okazaki Y, Sasaki T, Hosomoto K, Tanimoto S, Kawai K, Nagase T, Sugahara C, Yabuno S, Kin K, Sasada S, Yasuhara T, Tanaka S, Date I. Cervical spinal cord stimulation exerts anti-epileptic effects in a rat model of epileptic seizure through the suppression of CCL2-mediated cascades. Sci Rep 2024; 14:14543. [PMID: 38914629 PMCID: PMC11196670 DOI: 10.1038/s41598-024-64972-y] [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: 02/15/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024] Open
Abstract
Epidural spinal cord stimulation (SCS) is indicated for the treatment of intractable pain and is widely used in clinical practice. In previous basic research, the therapeutic effects of SCS have been demonstrated for epileptic seizure. However, the mechanism has not yet been elucidated. In this study, we investigated the therapeutic effect of SCS and the influence of epileptic seizure. First, SCS in the cervical spine was performed. The rats were divided into four groups: control group and treatment groups with SCS conducted at 2, 50, and 300 Hz frequency. Two days later, convulsions were induced by the intraperitoneal administration of kainic acid, followed by video monitoring to assess seizures. We also evaluated glial cells in the hippocampus by fluorescent immunostaining, electroencephalogram measurements, and inflammatory cytokines such as C-C motif chemokine ligand 2 (CCL2) by quantitative real-time polymerase chain reaction. Seizure frequency and the number of glial cells were significantly lower in the 300 Hz group than in the control group. SCS at 300 Hz decreased gene expression level of CCL2, which induces monocyte migration. SCS has anti-seizure effects by inhibiting CCL2-mediated cascades. The suppression of CCL2 and glial cells may be associated with the suppression of epileptic seizure.
Collapse
Grants
- 22K16659 Japan Ministry of Education, Culture, Sports, Science, and Technology
- 22K16688 Japan Ministry of Education, Culture, Sports, Science, and Technology
- 22K09207 Japan Ministry of Education, Culture, Sports, Science, and Technology
Collapse
Affiliation(s)
- Yosuke Okazaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Kakeru Hosomoto
- Department of Neurosurgery, Kure Kyosai Hospital, Kure, Japan
| | - Shun Tanimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Koji Kawai
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Takayuki Nagase
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Shota Tanaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Isao Date
- Department of Neurosurgery, Okayama Rosai Hospital, Okayama, Japan
| |
Collapse
|
4
|
Davidson B, Bhattacharya A, Sarica C, Darmani G, Raies N, Chen R, Lozano AM. Neuromodulation techniques - From non-invasive brain stimulation to deep brain stimulation. Neurotherapeutics 2024; 21:e00330. [PMID: 38340524 PMCID: PMC11103220 DOI: 10.1016/j.neurot.2024.e00330] [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: 10/11/2023] [Revised: 01/14/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Over the past 30 years, the field of neuromodulation has witnessed remarkable advancements. These developments encompass a spectrum of techniques, both non-invasive and invasive, that possess the ability to both probe and influence the central nervous system. In many cases neuromodulation therapies have been adopted into standard care treatments. Transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and transcranial ultrasound stimulation (TUS) are the most common non-invasive methods in use today. Deep brain stimulation (DBS), spinal cord stimulation (SCS), and vagus nerve stimulation (VNS), are leading surgical methods for neuromodulation. Ongoing active clinical trials using are uncovering novel applications and paradigms for these interventions.
Collapse
Affiliation(s)
- Benjamin Davidson
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | | | - Can Sarica
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Ghazaleh Darmani
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Nasem Raies
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Robert Chen
- Krembil Research Institute, University Health Network, Toronto, ON, Canada; Edmond J. Safra Program in Parkinson's Disease Morton and Gloria Shulman Movement Disorders Clinic, Division of Neurology, University of Toronto, Toronto, ON, Canada
| | - Andres M Lozano
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada; Krembil Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
5
|
Wang H, Li X, Qiao Y, Wang M, Wang W, McIntosh JM, Zhangsun D, Luo S. αO-Conotoxin GeXIVA[1,2] Reduced Neuropathic Pain and Changed Gene Expression in Chronic Oxaliplatin-Induced Neuropathy Mice Model. Mar Drugs 2024; 22:49. [PMID: 38276651 PMCID: PMC10821445 DOI: 10.3390/md22010049] [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: 12/27/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a dose-limiting painful neuropathy that occurs commonly during cancer management, which often leads to the discontinuation of medication. Previous studies suggest that the α9α10 nicotinic acetylcholine receptor (nAChR)-specific antagonist αO-conotoxin GeXIVA[1,2] is effective in CIPN models; however, the related mechanisms remain unclear. Here, we analyzed the preventive effect of GeXIVA[1,2] on neuropathic pain in the long-term oxaliplatin injection-induced CIPN model. At the end of treatment, lumbar (L4-L6) spinal cord was extracted, and RNA sequencing and bioinformatic analysis were performed to investigate the potential genes and pathways related to CIPN and GeXIVA[1,2]. GeXIVA[1,2] inhibited the development of mechanical allodynia induced by chronic oxaliplatin treatment. Repeated injections of GeXIVA[1,2] for 3 weeks had no effect on the mice's normal pain threshold or locomotor activity and anxiety-like behavior, as evaluated in the open field test (OFT) and elevated plus maze (EPM). Our RNA sequencing results identified 209 differentially expressed genes (DEGs) in the CIPN model, and simultaneously injecting GeXIVA[1,2] with oxaliplatin altered 53 of the identified DEGs. These reverted genes were significantly enriched in immune-related pathways represented by the cytokine-cytokine receptor interaction pathway. Our findings suggest that GeXIVA[1,2] could be a potential therapeutic compound for chronic oxaliplatin-induced CIPN management.
Collapse
Affiliation(s)
- Huanbai Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - Xiaodan Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
| | - Yamin Qiao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
| | - Meiting Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - Wen Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - J. Michael McIntosh
- Department of Biology and Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA;
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT 84108, USA
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| |
Collapse
|
6
|
Sivanesan E, Sanchez KR, Zhang C, He SQ, Linderoth B, Stephens KE, Raja SN, Guan Y. Spinal Cord Stimulation Increases Chemoefficacy and Prevents Paclitaxel-Induced Pain via CX3CL1. Neuromodulation 2023; 26:938-949. [PMID: 37045646 PMCID: PMC10330336 DOI: 10.1016/j.neurom.2023.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/19/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
INTRODUCTION Despite increasing utilization of spinal cord stimulation (SCS), its effects on chemoefficacy, cancer progression, and chemotherapy-induced peripheral neuropathy (CIPN) pain remain unclear. Up to 30% of adults who are cancer survivors may suffer from CIPN, and there are currently no effective preventative treatments. MATERIALS AND METHODS Through a combination of bioluminescent imaging, behavioral, biochemical, and immunohistochemical approaches, we investigated the role of SCS and paclitaxel (PTX) on tumor growth and PTX-induced peripheral neuropathy (PIPN) pain development in T-cell-deficient male rats (Crl:NIH-Foxn1rnu) with xenograft human non-small cell lung cancer. We hypothesized that SCS can prevent CIPN pain and enhance chemoefficacy partially by modulating macrophages, fractalkine (CX3CL1), and inflammatory cytokines. RESULTS We show that preemptive SCS enhanced the antitumor efficacy of PTX and prevented PIPN pain. Without SCS, rats with and without tumors developed robust PIPN pain-related mechanical hypersensitivity, but only those with tumors developed cold hypersensitivity, suggesting T-cell dependence for different PIPN pain modalities. SCS increased soluble CX3CL1 and macrophages and decreased neuronal and nonneuronal insoluble CX3CL1 expression and inflammation in dorsal root ganglia. CONCLUSION Collectively, our findings suggest that preemptive SCS is a promising strategy to increase chemoefficacy and prevent PIPN pain via CX3CL1-macrophage modulation.
Collapse
Affiliation(s)
- Eellan Sivanesan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Karla R Sanchez
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chi Zhang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Kimberly E Stephens
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
7
|
Dorrian RM, Berryman CF, Lauto A, Leonard AV. Electrical stimulation for the treatment of spinal cord injuries: A review of the cellular and molecular mechanisms that drive functional improvements. Front Cell Neurosci 2023; 17:1095259. [PMID: 36816852 PMCID: PMC9936196 DOI: 10.3389/fncel.2023.1095259] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that causes severe loss of motor, sensory and autonomic functions. Additionally, many individuals experience chronic neuropathic pain that is often refractory to interventions. While treatment options to improve outcomes for individuals with SCI remain limited, significant research efforts in the field of electrical stimulation have made promising advancements. Epidural electrical stimulation, peripheral nerve stimulation, and functional electrical stimulation have shown promising improvements for individuals with SCI, ranging from complete weight-bearing locomotion to the recovery of sexual function. Despite this, there is a paucity of mechanistic understanding, limiting our ability to optimize stimulation devices and parameters, or utilize combinatorial treatments to maximize efficacy. This review provides a background into SCI pathophysiology and electrical stimulation methods, before exploring cellular and molecular mechanisms suggested in the literature. We highlight several key mechanisms that contribute to functional improvements from electrical stimulation, identify gaps in current knowledge and highlight potential research avenues for future studies.
Collapse
Affiliation(s)
- Ryan M. Dorrian
- Spinal Cord Injury Research Group, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia,*Correspondence: Ryan M. Dorrian,
| | | | - Antonio Lauto
- School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Anna V. Leonard
- Spinal Cord Injury Research Group, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
8
|
Rizk E, Madrid A, Koueik J, Sun D, Stewart K, Chen D, Luo S, Hong F, Papale LA, Hariharan N, Alisch RS, Iskandar BJ. Purified regenerating retinal neurons reveal regulatory role of DNA methylation-mediated Na+/K+-ATPase in murine axon regeneration. Commun Biol 2023; 6:120. [PMID: 36717618 PMCID: PMC9886953 DOI: 10.1038/s42003-023-04463-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/11/2023] [Indexed: 01/31/2023] Open
Abstract
While embryonic mammalian central nervous system (CNS) axons readily grow and differentiate, only a minority of fully differentiated mature CNS neurons are able to regenerate injured axons, leading to stunted functional recovery after injury and disease. To delineate DNA methylation changes specifically associated with axon regeneration, we used a Fluorescent-Activated Cell Sorting (FACS)-based methodology in a rat optic nerve transection model to segregate the injured retinal ganglion cells (RGCs) into regenerating and non-regenerating cell populations. Whole-genome DNA methylation profiling of these purified neurons revealed genes and pathways linked to mammalian RGC regeneration. Moreover, whole-methylome sequencing of purified uninjured adult and embryonic RGCs identified embryonic molecular profiles reactivated after injury in mature neurons, and others that correlate specifically with embryonic or adult axon growth, but not both. The results highlight the contribution to both embryonic growth and adult axon regeneration of subunits encoding the Na+/K+-ATPase. In turn, both biochemical and genetic inhibition of the Na+/K+-ATPase pump significantly reduced RGC axon regeneration. These data provide critical molecular insights into mammalian CNS axon regeneration, pinpoint the Na+/K+-ATPase as a key regulator of regeneration of injured mature CNS axons, and suggest that successful regeneration requires, in part, reactivation of embryonic signals.
Collapse
Affiliation(s)
- Elias Rizk
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA ,grid.240473.60000 0004 0543 9901Department of Neurological Surgery, Penn State Milton S. Hershey Medical Center, Hershey, PA 17033 USA
| | - Andy Madrid
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Joyce Koueik
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Dandan Sun
- grid.21925.3d0000 0004 1936 9000Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Krista Stewart
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - David Chen
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Susan Luo
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Felissa Hong
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Ligia A. Papale
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Nithya Hariharan
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Reid S. Alisch
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| | - Bermans J. Iskandar
- grid.14003.360000 0001 2167 3675Department of Neurological Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53792 USA
| |
Collapse
|
9
|
Ghazisaeidi S, Muley MM, Salter MW. Neuropathic Pain: Mechanisms, Sex Differences, and Potential Therapies for a Global Problem. Annu Rev Pharmacol Toxicol 2023; 63:565-583. [PMID: 36662582 DOI: 10.1146/annurev-pharmtox-051421-112259] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The study of chronic pain continues to generate ever-increasing numbers of publications, but safe and efficacious treatments for chronic pain remain elusive. Recognition of sex-specific mechanisms underlying chronic pain has resulted in a surge of studies that include both sexes. A predominant focus has been on identifying sex differences, yet many newly identified cellular mechanisms and alterations in gene expression are conserved between the sexes. Here we review sex differences and similarities in cellular and molecular signals that drive the generation and resolution of neuropathic pain. The mix of differences and similarities reflects degeneracy in peripheral and central signaling processes by which neurons, immune cells, and glia codependently drive pain hypersensitivity. Recent findings identifying critical signaling nodes foreshadow the development of rationally designed, broadly applicable analgesic strategies. However, the paucity of effective, safe pain treatments compels targeted therapies as well to increase therapeutic options that help reduce the global burden of suffering.
Collapse
Affiliation(s)
- Shahrzad Ghazisaeidi
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada;
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- University of Toronto Centre for the Study of Pain, Toronto, Ontario, Canada
| | - Milind M Muley
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada;
- University of Toronto Centre for the Study of Pain, Toronto, Ontario, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada;
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- University of Toronto Centre for the Study of Pain, Toronto, Ontario, Canada
| |
Collapse
|
10
|
de Geus TJ, Franken G, Joosten EA. Conventional, high frequency and differential targeted multiplexed spinal cord stimulation in experimental painful diabetic peripheral neuropathy: Pain behavior and role of the central inflammatory balance. Mol Pain 2023; 19:17448069231193368. [PMID: 37488684 PMCID: PMC10504849 DOI: 10.1177/17448069231193368] [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] [Indexed: 07/26/2023] Open
Abstract
Spinal cord stimulation (SCS) is a last resort treatment for pain relief in painful diabetic peripheral neuropathy (PDPN) patients. However, the effectivity of SCS in PDPN is limited. New SCS paradigms such as high frequency (HF) and differential target multiplexed (DTM) might improve responder rates and efficacy of SCS-induced analgesia in PDPN patients, and are suggested to modulate the inflammatory balance and glial response in the spinal dorsal horn. The aim of this study was to research the effects of Con-, HF- and DTM-SCS on pain behavior and the spinal inflammatory balance in an animal model of PDPN. Streptozotocin-induced PDPN animals were stimulated for 48 hours with either Con-SCS (50Hz), HF-SCS (1200Hz) or DTM-SCS (combination of Con- and HF-SCS). Mechanical hypersensitivity was assessed using Von Frey (VF) test and the motivational aspects of pain were assessed using the mechanical conflict avoidance system (MCAS). The inflammatory balance and glial response were analyzed in the dorsal spinal cord based on RNA expression of pro- and anti-inflammatory cytokines (Tnf-α, Il-1ß, Il-4, Il-10), a microglia marker (Itgam), an astrocyte marker (Gfap), a T-cell marker (Cd3d), microglia proliferation markers (Irf8, Adgre1) and P2X4, p13-MAPK, BDNF signaling markers (P2x4, Mapk14, Bdnf). The results show that Con-, HF-, and DTM-SCS significantly decreased hypersensitivity after 48 hours of stimulation compared to Sham-SCS in PDPN animals, but at the same time did not affect escape latency in the MCAS. At the molecular level, Con-SCS resulted in a significant increase in spinal pro-inflammatory cytokine Tnf-α after 48 hours compared to DTM-SCS and Sham-SCS. In summary, Con-SCS showed a shift of the inflammatory balance towards a pro-inflammatory state whilst HF- and DTM-SCS shifted the balance towards an anti-inflammatory state. These findings suggest that the underlying mechanism of Con-SCS induced pain relief in PDPN differs from that induced by HF- and DTM-SCS.
Collapse
Affiliation(s)
- Thomas J. de Geus
- Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Anesthesiology and Pain Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Glenn Franken
- Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Anesthesiology and Pain Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Elbert A Joosten
- Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Anesthesiology and Pain Medicine, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
11
|
de Geus TJ, Franken G, Joosten EAJ. Spinal Cord Stimulation Paradigms and Pain Relief: A Preclinical Systematic Review on Modulation of the Central Inflammatory Response in Neuropathic Pain. Neuromodulation 2023; 26:25-34. [PMID: 35931643 DOI: 10.1016/j.neurom.2022.04.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) is a last-resort treatment for patients with chronic neuropathic pain. The mechanism underlying SCS and pain relief is not yet fully understood. Because the inflammatory balance between pro- and anti-inflammatory molecules in the spinal nociceptive network is pivotal in the development and maintenance of neuropathic pain, the working mechanism of SCS is suggested to be related to the modulation of this balance. The aim of this systematic review is to summarize and understand the effects of different SCS paradigms on the central inflammatory balance in the spinal cord. MATERIALS AND METHODS A systematic literature search was conducted using MEDLINE, Embase, and PubMed. All articles studying the effects of SCS on inflammatory or glial markers in neuropathic pain models were included. A quality assessment was performed on predetermined entities of bias. RESULTS A total of 11 articles were eligible for this systematic review. In general, induction of neuropathic pain in rats results in a proinflammatory state and at the same time an increased activity/expression of microglial and astroglial cells in the spinal cord dorsal horn. Conventional SCS seems to further enhance this proinflammatory state and increase the messenger RNA expression of microglial markers, but it also results in a decrease in microglial protein marker levels. High-frequency and especially differential targeted multiplexed SCS can not only restore the balance between pro- and anti-inflammatory molecules but also minimize the overexpression/activation of glial cells. Quality assessment and risk of bias analysis of the studies included make it clear that the results of these preclinical studies must be interpreted with caution. CONCLUSIONS In summary, the preclinical findings tend to indicate that there is a distinct SCS paradigm-related effect in the modulation of the central inflammatory balance of the spinal dorsal horn.
Collapse
Affiliation(s)
- Thomas J de Geus
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| | - Glenn Franken
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Department of Translational Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
12
|
Chu XL, Song XZ, Li Q, Li YR, He F, Gu XS, Ming D. Basic mechanisms of peripheral nerve injury and treatment via electrical stimulation. Neural Regen Res 2022; 17:2185-2193. [PMID: 35259827 PMCID: PMC9083151 DOI: 10.4103/1673-5374.335823] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Previous studies on the mechanisms of peripheral nerve injury (PNI) have mainly focused on the pathophysiological changes within a single injury site. However, recent studies have indicated that within the central nervous system, PNI can lead to changes in both injury sites and target organs at the cellular and molecular levels. Therefore, the basic mechanisms of PNI have not been comprehensively understood. Although electrical stimulation was found to promote axonal regeneration and functional rehabilitation after PNI, as well as to alleviate neuropathic pain, the specific mechanisms of successful PNI treatment are unclear. We summarize and discuss the basic mechanisms of PNI and of treatment via electrical stimulation. After PNI, activity in the central nervous system (spinal cord) is altered, which can limit regeneration of the damaged nerve. For example, cell apoptosis and synaptic stripping in the anterior horn of the spinal cord can reduce the speed of nerve regeneration. The pathological changes in the posterior horn of the spinal cord can modulate sensory abnormalities after PNI. This can be observed in cases of ectopic discharge of the dorsal root ganglion leading to increased pain signal transmission. The injured site of the peripheral nerve is also an important factor affecting post-PNI repair. After PNI, the proximal end of the injured site sends out axial buds to innervate both the skin and muscle at the injury site. A slow speed of axon regeneration leads to low nerve regeneration. Therefore, it can take a long time for the proximal nerve to reinnervate the skin and muscle at the injured site. From the perspective of target organs, long-term denervation can cause atrophy of the corresponding skeletal muscle, which leads to abnormal sensory perception and hyperalgesia, and finally, the loss of target organ function. The mechanisms underlying the use of electrical stimulation to treat PNI include the inhibition of synaptic stripping, addressing the excessive excitability of the dorsal root ganglion, alleviating neuropathic pain, improving neurological function, and accelerating nerve regeneration. Electrical stimulation of target organs can reduce the atrophy of denervated skeletal muscle and promote the recovery of sensory function. Findings from the included studies confirm that after PNI, a series of physiological and pathological changes occur in the spinal cord, injury site, and target organs, leading to dysfunction. Electrical stimulation may address the pathophysiological changes mentioned above, thus promoting nerve regeneration and ameliorating dysfunction.
Collapse
Affiliation(s)
- Xiao-Lei Chu
- Academy of Medical Engineering and Translational Medicine, Tianjin University; Department of Rehabilitation, Tianjin Hospital, Tianjin, China
| | - Xi-Zi Song
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Qi Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University; Department of Rehabilitation, Tianjin Hospital, Tianjin, China
| | - Yu-Ru Li
- College of Exercise & Health Sciences, Tianjin University of Sport, Tianjin, China
| | - Feng He
- College of Precision Instruments & Optoelectronics Engineering, Tianjin University, Tianjin, China
| | - Xiao-Song Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine; College of Precision Instruments & Optoelectronics Engineering, Tianjin University, Tianjin, China
| |
Collapse
|
13
|
Su PYP, Zhang L, He L, Zhao N, Guan Z. The Role of Neuro-Immune Interactions in Chronic Pain: Implications for Clinical Practice. J Pain Res 2022; 15:2223-2248. [PMID: 35957964 PMCID: PMC9359791 DOI: 10.2147/jpr.s246883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic pain remains a public health problem and contributes to the ongoing opioid epidemic. Current pain management therapies still leave many patients with poorly controlled pain, thus new or improved treatments are desperately needed. One major challenge in pain research is the translation of preclinical findings into effective clinical practice. The local neuroimmune interface plays an important role in the initiation and maintenance of chronic pain and is therefore a promising target for novel therapeutic development. Neurons interface with immune and immunocompetent cells in many distinct microenvironments along the nociceptive circuitry. The local neuroimmune interface can modulate the activity and property of the neurons to affect peripheral and central sensitization. In this review, we highlight a specific subset of many neuroimmune interfaces. In the central nervous system, we examine the interface between neurons and microglia, astrocytes, and T lymphocytes. In the periphery, we profile the interface between neurons in the dorsal root ganglion with T lymphocytes, satellite glial cells, and macrophages. To bridge the gap between preclinical research and clinical practice, we review the preclinical studies of each neuroimmune interface, discuss current clinical treatments in pain medicine that may exert its action at the neuroimmune interface, and highlight opportunities for future clinical research efforts.
Collapse
Affiliation(s)
- Po-Yi Paul Su
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Liangliang He
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Na Zhao
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Cedeño DL, Tilley DM, Vetri F, Platt DC, Vallejo R. Proteomic and Phosphoproteomic Changes of MAPK-Related Inflammatory Response in an Animal Model of Neuropathic Pain by Differential Target Multiplexed SCS and Low-Rate SCS. J Pain Res 2022; 15:895-907. [PMID: 35392631 PMCID: PMC8983055 DOI: 10.2147/jpr.s348738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/24/2022] [Indexed: 12/30/2022] Open
Abstract
Introduction Neuropathic pain initiates an interplay of pathways, involving MAP kinases and NFκB-signaling, leading to expression of immune response factors and activation and inactivation of proteins via phosphorylation. Neuropathic pain models demonstrated that spinal cord stimulation (SCS) may provide analgesia by modulating gene and protein expression in neuroinflammatory processes. A differential target multiplexed programming (DTMP) approach was more effective than conventional SCS treatments at modulating these. This work investigated the effect of DTMP and low rate SCS (LR-SCS) on proteins associated with MAP kinases and NFκB-signaling relevant to neuroinflammation. Methods Animals subjected to the spared nerve injury model (SNI) of neuropathic pain were treated continuously (48h) with either DTMP or LR-SCS. No-SNI and No-SCS groups were included as controls. Proteomics and phosphoproteomics of stimulated spinal cord tissues were performed via liquid chromatography/tandem mass spectrometry. Proteins were identified from mass spectra using bioinformatics. Expression levels and fold changes (No-SCS/No-SNI and SCS/No-SCS) were obtained from spectral intensities. Results Analyses identified 7192 proteins, with 1451 and 705 significantly changed by DTMP and LR-SCS, respectively. Eighty-one proteins, including MAP kinases, facilitating NFκB-signaling as part of inflammatory processes were identified. The pain model significantly increased expression levels of complement pathway-related proteins (LBP, NRG1, APP, CFH, C3, C5), which were significantly reversed by DTMP. Expression levels of other complement pathway-related proteins (HMGB1, S100A8, S100A9, CRP, C4) were decreased by DTMP, although not significantly affected by SNI. Other proteins (ORM1, APOE, NG2, CNTF) involved in NFκB-signaling were increased by SNI and decreased by DTMP. Expression levels of phosphorylated protein kinases involved in NFκB-signaling (including MAP kinases, PKC, MARK1) were affected by the pain model and reverse modulated by DTMP. LR-SCS modulated inflammatory-related proteins although to a lesser extent than DTMP. Conclusion Proteomic analyses support the profound effect of the DTMP approach on neuroinflammation via MAP kinases and NFκB-mediated signaling to alleviate neuropathic pain.
Collapse
Affiliation(s)
- David L Cedeño
- Neuroscience, Illinois Wesleyan University, Bloomington, IL, USA
- Research and Development, SGX Medical, Bloomington, IL, USA
| | - Dana M Tilley
- Research and Development, SGX Medical, Bloomington, IL, USA
| | - Francesco Vetri
- Research Department, National Spine and Pain Centers, Bloomington, IL, USA
| | - David C Platt
- Neuroscience, Illinois Wesleyan University, Bloomington, IL, USA
- Research and Development, SGX Medical, Bloomington, IL, USA
| | - Ricardo Vallejo
- Neuroscience, Illinois Wesleyan University, Bloomington, IL, USA
- Research and Development, SGX Medical, Bloomington, IL, USA
- Research Department, National Spine and Pain Centers, Bloomington, IL, USA
- Correspondence: Ricardo Vallejo; David L Cedeño, Email ;
| |
Collapse
|
15
|
Tilley DM, Cedeño DL, Vetri F, Platt DC, Vallejo R. Differential target multiplexed spinal cord stimulation programming modulates proteins involved in ion regulation in an animal model of neuropathic pain. Mol Pain 2022; 18:17448069211060181. [PMID: 35048719 PMCID: PMC8785327 DOI: 10.1177/17448069211060181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The effect of spinal cord stimulation (SCS) using differential target multiplexed programming (DTMP) on proteins involved in the regulation of ion transport in spinal cord (SC) tissue of an animal model of neuropathic pain was evaluated in comparison to low rate (LR) SCS. Rats subjected to the spared nerve injury model (SNI) and implanted with a SCS lead were assigned to DTMP or LR and stimulated for 48 h. A No-SCS group received no stimulation, and a Sham group received no SNI or stimulation. Proteins in the dorsal ipsilateral quadrant of the stimulated SC were identified and quantified using mass spectrometry. Proteins significantly modulated by DTMP or LR relative to No-SCS were identified. Bioinformatic tools were used to identify proteins related to ion transport regulation. DTMP modulated a larger number of proteins than LR. More than 40 proteins significantly involved in the regulation of chloride (Cl-), potassium (K+), sodium (Na+), or calcium (Ca2+) ions were identified. SNI affected proteins that promote the increase of intracellular Ca2+, Na+, and K+ and decrease of intracellular Cl-. DTMP modulated proteins involved in glial response to neural injury that affect Ca2+ signaling. DTMP decreased levels of proteins related to Ca2+ transport that may result in the reduction of intracellular Ca2+. Presynaptic proteins involved in GABA vesicle formation and release were upregulated by DTMP. DTMP also upregulated postsynaptic proteins involved with elevated intracellular Cl-, while modulating proteins, expressed by astrocytes, that regulate postsynaptic Cl- inhibition. DTMP downregulated K+ regulatory proteins affected by SNI that affect neuronal depolarization, and upregulated proteins that are associated with a decrease of intracellular neuronal K+ and astrocyte uptake of extracellular K+. DTMP treatment modulated the expression of proteins with the potential to facilitate a reversal of dysregulation of ion transport and signaling associated with a model of neuropathic pain.
Collapse
Affiliation(s)
| | - David L Cedeño
- SGX Medical LLC, Bloomington, IL, USA.,Department of Psychology, 1770Illinois Wesleyan University, Bloomington, IL, USA
| | | | - David C Platt
- SGX Medical LLC, Bloomington, IL, USA.,Department of Psychology, 1770Illinois Wesleyan University, Bloomington, IL, USA
| | - Ricardo Vallejo
- SGX Medical LLC, Bloomington, IL, USA.,Department of Psychology, 1770Illinois Wesleyan University, Bloomington, IL, USA.,National Spine and Pain Centers, Bloomington, IL, USA
| |
Collapse
|
16
|
Zhang H, Yang T. FBXW7alpha Promotes the Recovery of Traumatic Spinal Cord. Curr Mol Med 2021; 20:494-504. [PMID: 31870261 DOI: 10.2174/1566524020666191223164916] [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: 08/15/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND White matter damage and neuronal cell death are incurred by spinal cord injury (SCI). FBXW7α, an important mediator of cell division and growth was investigated to explore its role in repairing the traumatic spinal cord in rats. Underlying mechanisms such as oxidative stress and inflammasomes signaling were also studied. METHODS Spinal cord injury in rats was established by longitudinal surgical incision from the lower to mid-thoracic vertebrae on the backside, followed by 20-g weight placed on the exposed Th12 surface for 30 min. AAV-delivered FBXW7α and -sh-FBXW7α were intrathecally injected into the rat spinal cord. Indices of oxidation, neurotrophic factors, and pyroptosis were measured by Western blot, Elisa, and RT-PCR. RESULTS We found the overexpression of FBXW7α in spinal cord rescue neuronal death triggered by the injury. Specifically, the nutritional condition, oxidative stress, and pyroptosis were improved. A synchronization of BNDF and GDNF expression patterns in various groups indicated the secretion of neurotrophic factors affect the outcome of SCI. The SOD1, CAT, and GSH-px were suppressed after trauma but all restored in response to FBXW7α overexpression. Inflammasomes-activated pyroptosis was incurred after the injury, and relevant biomarkers such as GSDMD, caspase-1, caspase- 11, IL-1β, and IL-18 were down-regulated after the introduction of FBXW7α into the injured cord. Additionally, up-regulating FBXW7α also repaired the mitochondria dysfunction. CONCLUSION Our data indicate FBXW7α probably serves as an important molecular target for the therapy of spinal cord injury.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Trauma Center, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, 222061, China
| | - Tao Yang
- Department of Orthopedics, 4th (Xing Yuan) Hospital of Yulin, Yulin City, Shaanxi Province, 719000, China
| |
Collapse
|
17
|
Malone IG, Nosacka RL, Nash MA, Otto KJ, Dale EA. Electrical epidural stimulation of the cervical spinal cord: implications for spinal respiratory neuroplasticity after spinal cord injury. J Neurophysiol 2021; 126:607-626. [PMID: 34232771 PMCID: PMC8409953 DOI: 10.1152/jn.00625.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/07/2021] [Accepted: 06/27/2021] [Indexed: 01/15/2023] Open
Abstract
Traumatic cervical spinal cord injury (cSCI) can lead to damage of bulbospinal pathways to the respiratory motor nuclei and consequent life-threatening respiratory insufficiency due to respiratory muscle paralysis/paresis. Reports of electrical epidural stimulation (EES) of the lumbosacral spinal cord to enable locomotor function after SCI are encouraging, with some evidence of facilitating neural plasticity. Here, we detail the development and success of EES in recovering locomotor function, with consideration of stimulation parameters and safety measures to develop effective EES protocols. EES is just beginning to be applied in other motor, sensory, and autonomic systems; however, there has only been moderate success in preclinical studies aimed at improving breathing function after cSCI. Thus, we explore the rationale for applying EES to the cervical spinal cord, targeting the phrenic motor nucleus for the restoration of breathing. We also suggest cellular/molecular mechanisms by which EES may induce respiratory plasticity, including a brief examination of sex-related differences in these mechanisms. Finally, we suggest that more attention be paid to the effects of specific electrical parameters that have been used in the development of EES protocols and how that can impact the safety and efficacy for those receiving this therapy. Ultimately, we aim to inform readers about the potential benefits of EES in the phrenic motor system and encourage future studies in this area.
Collapse
Affiliation(s)
- Ian G Malone
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center (BREATHE), University of Florida, Gainesville, Florida
| | - Rachel L Nosacka
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Marissa A Nash
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Kevin J Otto
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center (BREATHE), University of Florida, Gainesville, Florida
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
- Department of Neuroscience, University of Florida, Gainesville, Florida
- Department of Neurology, University of Florida, Gainesville, Florida
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Erica A Dale
- Breathing Research and Therapeutics Center (BREATHE), University of Florida, Gainesville, Florida
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Department of Neuroscience, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| |
Collapse
|
18
|
Echeverria-Villalobos M, Mitchell J, Fiorda-Diaz J, Weaver T. Effects of Dorsal Column Spinal Cord Stimulation on Neuroinflammation: Revisiting Molecular Mechanisms and Clinical Outcomes on Chronic Lumbar/Leg Pain and Failed Back Surgery Syndrome. J Pain Res 2021; 14:2337-2345. [PMID: 34354373 PMCID: PMC8331196 DOI: 10.2147/jpr.s309872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/26/2021] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE In this narrative review, we reviewed and discussed current literature describing the molecular mechanisms leading to neuroinflammation and its role in the onset and progression of chronic neuropathic lumbar and leg pain in patients with persistent spinal pain syndrome. In addition, we reviewed the proposed mechanisms and impact of spinal cord stimulation (SCS) on neuroinflammation. METHODS A broad search of current literature in PubMed, Embase, Scopus, Cochrane library, Medline/Ovid, and Web of Science was performed using the following terms and their combinations: "biomarkers", "chronic back and leg pain", "cytokines", "neuroinflammation", "spinal cord stimulation (scs)," and "spinal cord modulation". We selected: 1) articles published in the English language between January 2000 and July 2020 2) preclinical and clinical data 3) case reports 4) meta-analysis and systematic reviews and 5) conference abstracts. Manuscripts not disclosing methodology or without full-text availability were excluded. DISCUSSION SCS techniques have gradually evolved since inception to include novel methods such as burst-SCS, high frequency SCS, and differential targeted multiplexed SCS. The incidence of chronic pain after spine surgery is highly variable, with at least one third of patients developing persistent spinal pain syndrome. Novel SCS techniques have been associated with improved clinical and functional outcomes thus increasing patient quality of life. CONCLUSION Currently, health care providers rely on different options and methods for SCS when treating patients with refractory chronic lumbar pain and persistent spinal pain syndrome. Nevertheless, compelling clinical trials remain necessary to elucidate the long-term benefits and mechanisms of neuromodulation of all different types of SCS.
Collapse
Affiliation(s)
| | - Justin Mitchell
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Juan Fiorda-Diaz
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tristan Weaver
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
19
|
Cedeño DL, Kelley CA, Chakravarthy K, Vallejo R. Modulation of Glia-Mediated Processes by Spinal Cord Stimulation in Animal Models of Neuropathic Pain. FRONTIERS IN PAIN RESEARCH 2021; 2:702906. [PMID: 35295479 PMCID: PMC8915735 DOI: 10.3389/fpain.2021.702906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/31/2021] [Indexed: 12/23/2022] Open
Abstract
Glial cells play an essential role in maintaining the proper functioning of the nervous system. They are more abundant than neurons in most neural tissues and provide metabolic and catabolic regulation, maintaining the homeostatic balance at the synapse. Chronic pain is generated and sustained by the disruption of glia-mediated processes in the central nervous system resulting in unbalanced neuron–glial interactions. Animal models of neuropathic pain have been used to demonstrate that changes in immune and neuroinflammatory processes occur in the course of pain chronification. Spinal cord stimulation (SCS) is an electrical neuromodulation therapy proven safe and effective for treating intractable chronic pain. Traditional SCS therapies were developed based on the gate control theory of pain and rely on stimulating large Aβ neurons to induce paresthesia in the painful dermatome intended to mask nociceptive input carried out by small sensory neurons. A paradigm shift was introduced with SCS treatments that do not require paresthesia to provide effective pain relief. Efforts to understand the mechanism of action of SCS have considered the role of glial cells and the effect of electrical parameters on neuron–glial interactions. Recent work has provided evidence that SCS affects expression levels of glia-related genes and proteins. This inspired the development of a differential target multiplexed programming (DTMP) approach using electrical signals that can rebalance neuroglial interactions by targeting neurons and glial cells differentially. Our group pioneered the utilization of transcriptomic and proteomic analyses to identify the mechanism of action by which SCS works, emphasizing the DTMP approach. This is an account of evidence demonstrating the effect of SCS on glia-mediated processes using neuropathic pain models, emphasizing studies that rely on the evaluation of large sets of genes and proteins. We show that SCS using a DTMP approach strongly affects the expression of neuron and glia-specific transcriptomes while modulating them toward expression levels of healthy animals. The ability of DTMP to modulate key genes and proteins involved in glia-mediated processes affected by pain toward levels found in uninjured animals demonstrates a shift in the neuron–glial environment promoting analgesia.
Collapse
Affiliation(s)
- David L. Cedeño
- Research and Development, Lumbrera LLC, Bloomington, IL, United States
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, United States
- *Correspondence: David L. Cedeño
| | - Courtney A. Kelley
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, United States
| | - Krishnan Chakravarthy
- Deparment of Anesthesiology and Pain Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Ricardo Vallejo
- Research and Development, Lumbrera LLC, Bloomington, IL, United States
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, United States
- Research Department, National Spine and Pain Center, Bloomington, IL, United States
| |
Collapse
|
20
|
Albik S, Tao YX. Emerging role of RNA m6A modification in chronic pain. Pain 2021; 162:1897-1898. [PMID: 33675633 PMCID: PMC8205953 DOI: 10.1097/j.pain.0000000000002219] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/04/2021] [Indexed: 01/25/2023]
Affiliation(s)
- Sfian Albik
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Physiology, Pharmacology & Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
21
|
Smith WJ, Cedeño DL, Thomas SM, Kelley CA, Vetri F, Vallejo R. Modulation of microglial activation states by spinal cord stimulation in an animal model of neuropathic pain: Comparing high rate, low rate, and differential target multiplexed programming. Mol Pain 2021; 17:1744806921999013. [PMID: 33626981 PMCID: PMC7925954 DOI: 10.1177/1744806921999013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While numerous studies and patient experiences have demonstrated the efficacy of spinal cord stimulation as a treatment for chronic neuropathic pain, the exact mechanism underlying this therapy is still uncertain. Recent studies highlighting the importance of microglial cells in chronic pain and characterizing microglial activation transcriptomes have created a focus on microglia in pain research. Our group has investigated the modulation of gene expression in neurons and glial cells after spinal cord stimulation (SCS), specifically focusing on transcriptomic changes induced by varying SCS stimulation parameters. Previous work showed that, in rodents subjected to the spared nerve injury (SNI) model of neuropathic pain, a differential target multiplexed programming (DTMP) approach provided significantly better relief of pain-like behavior compared to high rate (HRP) and low rate programming (LRP). While these studies demonstrated the importance of transcriptomic changes in SCS mechanism of action, they did not specifically address the role of SCS in microglial activation. The data presented herein utilizes microglia-specific activation transcriptomes to further understand how an SNI model of chronic pain and subsequent continuous SCS treatment with either DTMP, HRP, or LRP affects microglial activation. Genes for each activation transcriptome were identified within our dataset and gene expression levels were compared with that of healthy animals, naïve to injury and interventional procedures. Pearson correlations indicated that DTMP yields the highest significant correlations to expression levels found in the healthy animals across all microglial activation transcriptomes. In contrast, HRP or LRP yielded weak or very weak correlations for these transcriptomes. This work demonstrates that chronic pain and subsequent SCS treatments can modulate microglial activation transcriptomes, supporting previous research on microglia in chronic pain. Furthermore, this study provides evidence that DTMP is more effective than HRP and LRP at modulating microglial transcriptomes, offering potential insight into the therapeutic efficacy of DTMP.
Collapse
Affiliation(s)
- William J Smith
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - David L Cedeño
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Samuel M Thomas
- College of Osteopathic Medicine, Des Moines University, Des Moines, IA, USA
| | - Courtney A Kelley
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | | | - Ricardo Vallejo
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA.,National Spine and Pain Centers, Bloomington, IL, USA
| |
Collapse
|
22
|
Wang J, Zheng X, Liu B, Yin C, Chen R, Li X, Li Y, Nie H, Zeng D, He X, Jiang Y, Fang J, Liu B. Electroacupuncture Alleviates Mechanical Allodynia of a Rat Model of CRPS-I and Modulates Gene Expression Profiles in Dorsal Root Ganglia. Front Neurol 2020; 11:580997. [PMID: 33193035 PMCID: PMC7661737 DOI: 10.3389/fneur.2020.580997] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Complex regional pain syndrome type-I (CRPS-I) is chronic neurological disorder accompanied with devastating pain. Most conventional medical treatments lack effectiveness, making CRPS-I a challenging clinical condition. Electroacupuncture (EA) showed effectiveness in alleviating the pain symptoms of CRPS-I patients. However, the molecular mechanisms underlying EA's therapeutic effect are still not well-understood. Here, we established the rat chronic post-ischemic pain (CPIP) model to mimic CRPS-I and performed repetitive EA on bilateral hind limbs of the CPIP model rats. We then performed RNA-sequencing (RNA-Seq) to study the differences in gene expression, gene networks, and molecular pathways in ipsilateral DRGs innervating the hind limb of the CPIP model rats with and without repetitive EA treatment. Our results found that repetitive EA treatment significantly alleviated mechanical allodynia in bilateral hind limbs of CPIP model rats. RNA-Seq analysis indicated that EA modulated the expression of multiple genes and gene networks in the DRGs of CPIP model rats. Further bioinformatics analysis identified the up-regulation of an array of genes involved in biological process such as neutrophil chemotaxis and immune response in the DRGs of CPIP model rats after EA treatment. Thus, these results suggest that EA may alleviate pain response in CPIP model rats via regulating multiple genes. Our work may help to further advance the understandings of the molecular mechanisms underlying EA's therapeutic effects on CRPS-I and help to identify novel targets for CRPS-I treatment.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoli Zheng
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Boyu Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengyu Yin
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruixiang Chen
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaojie Li
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuanyuan Li
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huimin Nie
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Danyi Zeng
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Boyi Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
Vallejo R, Gupta A, Cedeno DL, Vallejo A, Smith WJ, Thomas SM, Benyamin R, Kaye AD, Manchikanti L. Clinical Effectiveness and Mechanism of Action of Spinal Cord Stimulation for Treating Chronic Low Back and Lower Extremity Pain: a Systematic Review. Curr Pain Headache Rep 2020; 24:70. [PMID: 32997170 DOI: 10.1007/s11916-020-00907-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW The purpose of the present systematic review is to provide a current understanding of the mechanism of action and the evidence available to support clinical decision-making. The focus is to summarize randomized controlled trials (RCTs) and nonrandomized or observational studies of spinal cord stimulation in chronic pain to understand clinical effectiveness and the mechanism of action. RECENT FINDINGS Several recent studies have demonstrated the benefit of spinal cord stimulation in managing chronic pain. Until recently, the mechanism of action was founded on a central paradigm derived from gate control theory, which is the need to stimulate the dorsal column of the spinal cord to generate paresthesia. The recent development of new therapies that do not rely on paresthesia has left the field without a clear mechanism of action that could serve as a strong foundation to further improve clinical outcomes. Consequently, multiple theories have emerged to explain how electrical pulse applied to the spinal cord could alleviate pain, including activation of specific supraspinal pathways, and segmental modulation of the neurological interaction. Recent systematic reviews also have shown the clinical effectiveness of spinal cord stimulation in managing chronic spinal pain, phantom limb pain, complex regional pain syndrome, and other chronic painful conditions. Spinal cord stimulation for the treatment of chronic pain is rapidly evolving with technology at its forefront. This comprehensive focused review evaluated 11 RCTs and 7 nonrandomized/observational studies which provided levels of evidence ranging from I to II.
Collapse
Affiliation(s)
- Ricardo Vallejo
- Millennium Pain Center - National Spine and Pain Centers, 2406 E Empire, Bloomington, IL, 61704, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
- SGX Medical LLC, Bloomington, IL, USA
| | - Ashim Gupta
- South Texas Orthopaedic Research Institute, Laredo, TX, USA
- BioIntegrate, 2505 Newpoint Pkwy Suite 100-A, Lawrenceville, GA, USA
- Future Biologics, Lawrenceville, GA, USA
| | - David L Cedeno
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA.
- SGX Medical LLC, Bloomington, IL, USA.
- Lumbrera LLC, 2406 E Empire, Bloomington, IL, 61704, USA.
| | - Alejandro Vallejo
- Lumbrera LLC, 2406 E Empire, Bloomington, IL, 61704, USA
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, USA
| | - William J Smith
- Lumbrera LLC, 2406 E Empire, Bloomington, IL, 61704, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Samuel M Thomas
- College of Osteopathic Medicine, Des Moines University, Des Moines, IA, USA
| | - Ramsin Benyamin
- Millennium Pain Center - National Spine and Pain Centers, 2406 E Empire, Bloomington, IL, 61704, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
- College of Medicine, University of Illinois, Urbana-Champaign, IL, USA
| | - Alan D Kaye
- Department of Anesthesiology, LSU Health Science Center, 1542 Tulane Ave Room 659, New Orleans, LA, 70112, USA
- Department of Pharmacology, LSU Health Science Center, 1542 Tulane Ave Room 659, New Orleans, LA, 70112, USA
| | - Laxmaiah Manchikanti
- Pain Management Centers of America, Management Center of Paducah, 67 Lakeview Dr., Paducah, KY, 42001, USA
- Anesthesiology and Perioperative Medicine, University of Louisville, Louisville, KY, USA
- Department of Anesthesiology, School of Medicine, LSU Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
24
|
Shu B, He SQ, Guan Y. Spinal Cord Stimulation Enhances Microglial Activation in the Spinal Cord of Nerve-Injured Rats. Neurosci Bull 2020; 36:1441-1453. [PMID: 32889636 DOI: 10.1007/s12264-020-00568-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022] Open
Abstract
Microglia can modulate spinal nociceptive transmission. Yet, their role in spinal cord stimulation (SCS)-induced pain inhibition is unclear. Here, we examined how SCS affects microglial activation in the lumbar cord of rats with chronic constriction injury (CCI) of the sciatic nerve. Male rats received conventional SCS (50 Hz, 80% motor threshold, 180 min, 2 sessions/day) or sham stimulation on days 18-20 post-CCI. SCS transiently attenuated the mechanical hypersensitivity in the ipsilateral hind paw and increased OX-42 immunoreactivity in the bilateral dorsal horns. SCS also upregulated the mRNAs of M1-like markers, but not M2-like markers. Inducible NOS protein expression was increased, but brain-derived neurotrophic factor was decreased after SCS. Intrathecal minocycline (1 μg-100 μg), which inhibits microglial activation, dose-dependently attenuated the mechanical hypersensitivity. Pretreatment with low-dose minocycline (1 μg, 30 min) prolonged the SCS-induced pain inhibition. These findings suggest that conventional SCS may paradoxically increase spinal M1-like microglial activity and thereby compromise its own ability to inhibit pain.
Collapse
Affiliation(s)
- Bin Shu
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.,Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA. .,Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
25
|
Nagel SJ, Hsieh J, Machado AG, Frizon LA, Howard MA, Gillies GT, Wilson S. Biomarker Optimization of Spinal Cord Stimulation Therapies. Neuromodulation 2020; 24:3-12. [PMID: 32881257 DOI: 10.1111/ner.13252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/18/2020] [Accepted: 06/29/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES We are in the process of designing and testing an intradural stimulation device that will shorten the distance between the location of the electrode array and the targeted neural tissue, thus improving the efficacy of electrical current delivery. Identifying a biomarker that accurately reflects the response to this intervention is highly valued because of the potential to optimize interventional parameters or predict a response before it is clinically measurable. In this report, we summarize the findings pertaining to the study of biomarkers so that we and others will have an up-to-date reference that critically evaluates the current approaches and select one or several for testing during the development of our device. MATERIALS AND METHODS We have conducted a broad survey of the existing literature to catalogue the biomarkers that could be coupled to intradural spinal cord stimulation. We describe in detail some of the most promising biomarkers, existing limitations, and suitability to managing chronic pain. RESULTS Chronic, intractable pain is an all-encompassing condition that is incurable. Many treatments for managing chronic pain are nonspecific in action and intermittently administered; therefore, patients are particularly susceptible to large fluctuations in pain control over the course of a day. The absence of a reliable biomarker challenges assessment of therapeutic efficacy and contributes to either incomplete and inconsistent pain relief or, alternatively, intolerable side effects. Fluctuations in metabolites or inflammatory markers, signals captured during dynamic imaging, and genomics will likely have a role in governing how a device is modulated. CONCLUSIONS Efforts to identify one or more biomarkers are well underway with some preliminary evidence supporting their efficacy. This has far-reaching implications, including improved outcomes, fewer adverse events, harmonization of treatment and individuals, performance gains, and cost savings. We anticipate that novel biomarkers will be used widely to manage chronic pain.
Collapse
Affiliation(s)
- Sean J Nagel
- Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH, USA
| | - Jason Hsieh
- Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH, USA
| | - Andre G Machado
- Center for Neurological Restoration, Cleveland Clinic, Cleveland, OH, USA
| | - Leonardo A Frizon
- Department of Neurosurgery, Hospital Marcelino Champagnat, Curitiba, PR, Brazil
| | - Matthew A Howard
- Department of Neurosurgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - George T Gillies
- Department of Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, VA, USA
| | - Saul Wilson
- Department of Neurosurgery, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
26
|
Chernov AV, Hullugundi SK, Eddinger KA, Dolkas J, Remacle AG, Angert M, James BP, Yaksh TL, Strongin AY, Shubayev VI. A myelin basic protein fragment induces sexually dimorphic transcriptome signatures of neuropathic pain in mice. J Biol Chem 2020; 295:10807-10821. [PMID: 32532796 DOI: 10.1074/jbc.ra120.013696] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
In the peripheral nerve, mechanosensitive axons are insulated by myelin, a multilamellar membrane formed by Schwann cells. Here, we offer first evidence that a myelin degradation product induces mechanical hypersensitivity and global transcriptomics changes in a sex-specific manner. Focusing on downstream signaling events of the functionally active 84-104 myelin basic protein (MBP(84-104)) fragment released after nerve injury, we demonstrate that exposing the sciatic nerve to MBP(84-104) via endoneurial injection produces robust mechanical hypersensitivity in female, but not in male, mice. RNA-seq and systems biology analysis revealed a striking sexual dimorphism in molecular signatures of the dorsal root ganglia (DRG) and spinal cord response, not observed at the nerve injection site. Mechanistically, intra-sciatic MBP(84-104) induced phospholipase C (PLC)-driven (females) and phosphoinositide 3-kinase-driven (males) phospholipid metabolism (tier 1). PLC/inositol trisphosphate receptor (IP3R) and estrogen receptor co-regulation in spinal cord yielded Ca2+-dependent nociceptive signaling induction in females that was suppressed in males (tier 2). IP3R inactivation by intrathecal xestospongin C attenuated the female-specific hypersensitivity induced by MBP(84-104). According to sustained sensitization in tiers 1 and 2, T cell-related signaling spreads to the DRG and spinal cord in females, but remains localized to the sciatic nerve in males (tier 3). These results are consistent with our previous finding that MBP(84-104)-induced pain is T cell-dependent. In summary, an autoantigenic peptide endogenously released in nerve injury triggers multisite, sex-specific transcriptome changes, leading to neuropathic pain only in female mice. MBP(84-104) acts through sustained co-activation of metabolic, estrogen receptor-mediated nociceptive, and autoimmune signaling programs.
Collapse
Affiliation(s)
- Andrei V Chernov
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA .,Infectious & Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Swathi K Hullugundi
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA.,Veterans Affairs San Diego Healthcare System, La Jolla, California, USA
| | - Kelly A Eddinger
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
| | - Jennifer Dolkas
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA.,Veterans Affairs San Diego Healthcare System, La Jolla, California, USA
| | - Albert G Remacle
- Infectious & Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Mila Angert
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA.,Veterans Affairs San Diego Healthcare System, La Jolla, California, USA
| | - Brian P James
- Infectious & Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Tony L Yaksh
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
| | - Alex Y Strongin
- Infectious & Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Veronica I Shubayev
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA .,Veterans Affairs San Diego Healthcare System, La Jolla, California, USA
| |
Collapse
|
27
|
Royds J, Conroy MJ, Dunne MR, Cassidy H, Matallanas D, Lysaght J, McCrory C. Examination and characterisation of burst spinal cord stimulation on cerebrospinal fluid cellular and protein constituents in patient responders with chronic neuropathic pain - A Pilot Study. J Neuroimmunol 2020; 344:577249. [PMID: 32361148 DOI: 10.1016/j.jneuroim.2020.577249] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/11/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Patients with neuropathic pain have altered proteomic and neuropeptide constituents in cerebrospinal fluid (CSF) compared to controls. Tonic spinal cord stimulation (SCS) has demonstrated differential expression of neuropeptides in CSF before and after treatment suggesting potential mechanisms of action. Burst-SCS is an evidence-based paraesthesia free waveform utilised for neuropathic pain with a potentially different mechanistic action to tonic SCS. This study examines the dynamic biological changes of CSF at a cellular and proteome level after Burst-SCS. METHODS Patients with neuropathic pain selected for SCS had CSF sampled prior to implant of SCS and following 8 weeks of continuous Burst-SCS. Baseline and 8-week pain scores with demographics were recorded. T cell frequencies were analysed by flow cytometry, proteome analysis was performed using mass spectrometry and secreted cytokines, chemokines and neurotrophins were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS 4 patients (2 females, 2 males) with a mean age of 51 years (+/-SEM 2.74, SD 5.48) achieved a reduction in pain of >50% following 8 weeks of Burst-SCS. Analysis of the CSF proteome indicated a significant alteration in protein expression most related to synapse assembly and immune regulators. There was significantly lower expression of the proteins: growth hormone A1 (PRL), somatostatin (SST), nucleobindin-2 (NUCB2), Calbindin (CALB1), acyl-CoA binding protein (DBI), proSAAS (PCSK1N), endothelin-3 (END3) and cholecystokinin (CCK) after Burst-SCS. The concentrations of secreted chemokines and cytokines and the frequencies of T cells were not significantly changed following Burst-SCS. CONCLUSION This study characterised the alteration in the CSF proteome in response to burst SCS in vivo. Functional analysis indicated that the alterations in the CSF proteome is predominately linked to synapse assembly and immune effectors. Individual protein analysis also suggests potential supraspinal mechanisms.
Collapse
Affiliation(s)
- Jonathan Royds
- Department of Pain Medicine, St. James Hospital, Dublin and School of Medicine, Trinity College Dublin, Ireland.
| | - Melissa J Conroy
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland
| | - Margaret R Dunne
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland
| | - Hilary Cassidy
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland
| | - Connail McCrory
- Department of Pain Medicine, St. James Hospital, Dublin and School of Medicine, Trinity College Dublin, Ireland
| |
Collapse
|
28
|
Suppression of Superficial Microglial Activation by Spinal Cord Stimulation Attenuates Neuropathic Pain Following Sciatic Nerve Injury in Rats. Int J Mol Sci 2020; 21:ijms21072390. [PMID: 32235682 PMCID: PMC7177766 DOI: 10.3390/ijms21072390] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 12/29/2022] Open
Abstract
We evaluated the mechanisms underlying the spinal cord stimulation (SCS)-induced analgesic effect on neuropathic pain following spared nerve injury (SNI). On day 3 after SNI, SCS was performed for 6 h by using electrodes paraspinally placed on the L4-S1 spinal cord. The effects of SCS and intraperitoneal minocycline administration on plantar mechanical sensitivity, microglial activation, and neuronal excitability in the L4 dorsal horn were assessed on day 3 after SNI. The somatosensory cortical responses to electrical stimulation of the hind paw on day 3 following SNI were examined by using in vivo optical imaging with a voltage-sensitive dye. On day 3 after SNI, plantar mechanical hypersensitivity and enhanced microglial activation were suppressed by minocycline or SCS, and L4 dorsal horn nociceptive neuronal hyperexcitability was suppressed by SCS. In vivo optical imaging also revealed that electrical stimulation of the hind paw-activated areas in the somatosensory cortex was decreased by SCS. The present findings suggest that SCS could suppress plantar SNI-induced neuropathic pain via inhibition of microglial activation in the L4 dorsal horn, which is involved in spinal neuronal hyperexcitability. SCS is likely to be a potential alternative and complementary medicine therapy to alleviate neuropathic pain following nerve injury.
Collapse
|
29
|
Tilley DM, Lietz CB, Cedeno DL, Kelley CA, Li L, Vallejo R. Proteomic Modulation in the Dorsal Spinal Cord Following Spinal Cord Stimulation Therapy in an In Vivo Neuropathic Pain Model. Neuromodulation 2020; 24:22-32. [PMID: 32157770 DOI: 10.1111/ner.13103] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 11/12/2019] [Accepted: 12/18/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) provides relief for patients suffering from chronic neuropathic pain although its mechanism may not be as dependent on electrical interference as classically considered. Recent evidence has been growing regarding molecular changes that are induced by SCS as being a key player in reversing the pain process. Here, we observed the effect of SCS on altering protein expression in spinal cord tissue using a proteomic analysis approach. METHODS A microlead was epidurally implanted following induction of an animal neuropathic pain model. After the model was established, stimulation was applied for 72 hours continuously followed by tissue collection and proteomic analysis via tandem mass spectroscopy. Identified proteins were run through online data bases for protein identification and classification of biological processes. RESULTS A significant improvement in mechanical sensitivity was observed following 48 hours of SCS therapy. Proteomic analysis identified 5840 proteins, of which 155 were significantly affected by SCS. Gene ontology data bases indicated that a significant number of proteins were associated to stress response, oxidation/reduction, or extracellular matrix pathways. Additionally, many of the proteins identified also play a role in neuron-glial interactions and are involved in nociception. CONCLUSIONS The development of an injury unbalances the proteome of the local neural tissue, neurons, and glial cells, and shifts the proteomic profile to a pain producing state. This study demonstrates the reversal of the injury-induced proteomic state by applying conventional SCS therapy. Additional studies looking at variations in electrical parameters are needed to optimize SCS.
Collapse
Affiliation(s)
| | | | - David L Cedeno
- Lumbrera LLC, Bloomington, IL, USA.,Stimgenics LLC, Bloomington, IL, USA
| | | | - Lingjun Li
- University of Wisconsin-Madison, Madison, WI, USA
| | - Ricardo Vallejo
- Lumbrera LLC, Bloomington, IL, USA.,Stimgenics LLC, Bloomington, IL, USA.,National Spine and Pain Center, Bloomington, IL, USA
| |
Collapse
|
30
|
Vallejo R, Gupta A, Kelley CA, Vallejo A, Rink J, Williams JM, Cass CL, Smith WJ, Benyamin R, Cedeño DL. Effects of Phase Polarity and Charge Balance Spinal Cord Stimulation on Behavior and Gene Expression in a Rat Model of Neuropathic Pain. Neuromodulation 2020; 23:26-35. [PMID: 31070863 DOI: 10.1111/ner.12964] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/12/2019] [Accepted: 04/03/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the effect of phase polarity and charge balance of spinal cord stimulation (SCS) waveforms on pain behavior and gene expression in a neuropathic pain rodent model. We hypothesized that differing waveforms will result in diverse behavioral and transcriptomics expression due to unique mechanisms of action. MATERIALS AND METHODS Rats were implanted with a four-contact cylindrical mini-lead and randomly assigned to two control (no-pain and pain model) and five test groups featuring monophasic, as well as charge-unbalanced and charge-balanced biphasic SCS waveforms. Mechanical and cold allodynia were assessed to measure efficacy. The ipsilateral dorsal quadrant of spinal cord adjacent to the lead was harvested post-stimulation and processed to determine gene expression via real-time reverse-transcriptase polymerase chain reaction (RT-PCR). Gene expression, SCS intensity (mA), and behavioral score as percent of baseline (BSPB) were statistically analyzed and used to generate correlograms using R-Studio. Statistical analysis was performed using SPSS22.0, and p < 0.05 was considered significant. RESULTS As expected, BSPB was significantly lower for the pain model group compared to the no-pain group. BSPB was significantly improved post-stim compared to pre-stim using cathodic, anodic, symmetric biphasic, or asymmetric biphasic 1:2 waveforms; however, BSPB was not restored to Sham levels. RT-PCR analysis showed that eight genes demonstrated a significant difference between the pain model and SCS waveforms and between waveforms. Correlograms reveal a linear correlation between regulation of expression of a given gene in relation to mA, BSPB, or other genes. CONCLUSIONS Our results exhibit that specific SCS waveforms differentially modulate several key transcriptional pathways that are relevant in chronic pain conditions. These results have significant implications for SCS: whether to move beyond traditional paradigm of neuronal activation to focus also on modulating immune-driven processes.
Collapse
Affiliation(s)
- Ricardo Vallejo
- Millennium Pain Center, Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Ashim Gupta
- Millennium Pain Center, Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
- South Texas Orthopaedic Research Institute, Laredo, TX, USA
| | - Courtney A Kelley
- Millennium Pain Center, Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | | | - Jonathan Rink
- Department of Biology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Joseph M Williams
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Cynthia L Cass
- Millennium Pain Center, Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - William J Smith
- Millennium Pain Center, Bloomington, IL, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Ramsin Benyamin
- Millennium Pain Center, Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
- College of Medicine, University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, USA
| | - David L Cedeño
- Millennium Pain Center, Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| |
Collapse
|
31
|
Vallejo R, Kelley CA, Gupta A, Smith WJ, Vallejo A, Cedeño DL. Modulation of neuroglial interactions using differential target multiplexed spinal cord stimulation in an animal model of neuropathic pain. Mol Pain 2020; 16:1744806920918057. [PMID: 32290778 PMCID: PMC7160773 DOI: 10.1177/1744806920918057] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
The development and maintenance of chronic neuropathic pain involves distorted neuroglial interactions, which result in prolonged perturbations of immune and inflammatory response, as well as disrupted synapses and cellular interactions. Spinal cord stimulation (SCS) has proven effective and safe for more than 40 years, but comprehensive understanding of its mode of action remains elusive. Previous work in our laboratory provided evidence that conventional SCS parameters modulate biological processes associated with neuropathic pain in animals. This inspired the development of differential target multiplexed programming (DTMP) in which multiple electrical signals are used for modulating glial cells and neurons in order to rebalance their interactions. This work compares DTMP with both low rate and high rate programming using an animal model of neuropathic pain. The spared nerve injury model was implemented in 48 rats equally randomized into four experimental groups: No-SCS, DTMP, low rate, and high rate. Naive animals (N = 7) served as a reference control. SCS was applied continuously for 48 h and pain-related behavior assessed before and after SCS. RNA from the spinal cord exposed to SCS was sequenced to determine changes in gene expression as a result of injury (No-SCS vs. naïve) and as a result of SCS (SCS vs. No-SCS). Bioinformatics tools (Weighted Gene Co-expression Network Analysis and Gene Ontology Enrichment Analysis) were used to evaluate the significance of the results. All three therapies significantly reduced mechanical hypersensitivity, although DTMP provided statistically better results overall. DTMP also reduced thermal hypersensitivity significantly. RNA-sequencing corroborated the complex effects of nerve injury on the transcriptome. In addition, DTMP provided significantly more effective modulation of genes associated with pain-related processes in returning their expression toward levels observed in naïve, noninjured animals. DTMP provides a more effective way of modulating the expression of genes involved in pain-relevant biological processes associated with neuroglial interactions.
Collapse
Affiliation(s)
- Ricardo Vallejo
- Department of Basic Science, Millennium Pain Center,
Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University,
Bloomington, IL, USA
| | - Courtney A Kelley
- Department of Basic Science, Millennium Pain Center,
Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University,
Bloomington, IL, USA
| | - Ashim Gupta
- Department of Basic Science, Millennium Pain Center,
Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University,
Bloomington, IL, USA
- Department of Research, South Texas Orthopaedic Research
Institute, Laredo, TX, USA
| | - William J Smith
- Department of Basic Science, Millennium Pain Center,
Bloomington, IL, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH,
USA
| | - Alejandro Vallejo
- Department of Basic Science, Millennium Pain Center,
Bloomington, IL, USA
| | - David L Cedeño
- Department of Basic Science, Millennium Pain Center,
Bloomington, IL, USA
- Department of Psychology, Illinois Wesleyan University,
Bloomington, IL, USA
| |
Collapse
|
32
|
Vallejo R, Platt DC, Rink JA, Jones MA, Kelley CA, Gupta A, Cass CL, Eichenberg K, Vallejo A, Smith WJ, Benyamin R, Cedeño DL. Electrical Stimulation of C6 Glia-Precursor Cells In Vitro Differentially Modulates Gene Expression Related to Chronic Pain Pathways. Brain Sci 2019; 9:303. [PMID: 31683631 PMCID: PMC6896182 DOI: 10.3390/brainsci9110303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/27/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
Glial cells comprise the majority of cells in the central nervous system and exhibit diverse functions including the development of persistent neuropathic pain. While earlier theories have proposed that the applied electric field specifically affects neurons, it has been demonstrated that electrical stimulation (ES) of neural tissue modulates gene expression of the glial cells. This study examines the effect of ES on the expression of eight genes related to oxidative stress and neuroprotection in cultured rodent glioma cells. Concentric bipolar electrodes under seven different ES types were used to stimulate cells for 30 min in the presence and absence of extracellular glutamate. ES consisted of rectangular pulses at 50 Hz in varying proportions of anodic and cathodic phases. Real-time reverse-transcribed quantitative polymerase chain reaction was used to determine gene expression using the ∆∆Cq method. The results demonstrate that glutamate has a significant effect on gene expression in both stimulated and non-stimulated groups. Furthermore, stimulation parameters have differential effects on gene expression, both in the presence and absence of glutamate. ES has an effect on glial cell gene expression that is dependent on waveform composition. Optimization of ES therapy for chronic pain applications can be enhanced by this understanding.
Collapse
Affiliation(s)
- Ricardo Vallejo
- Millennium Pain Center, Bloomington, IL 61704, USA.
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL 61701, USA.
| | - David C Platt
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Jonathan A Rink
- Department of Biology, Illinois Wesleyan University, Bloomington, IL 61701, USA.
| | - Marjorie A Jones
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Courtney A Kelley
- Millennium Pain Center, Bloomington, IL 61704, USA.
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL 61701, USA.
| | - Ashim Gupta
- Millennium Pain Center, Bloomington, IL 61704, USA.
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL 61701, USA.
- South Texas Orthopaedic Research Institute, Laredo, TX 78045, USA.
| | - Cynthia L Cass
- Millennium Pain Center, Bloomington, IL 61704, USA.
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL 61701, USA.
| | - Kirk Eichenberg
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | | | - William J Smith
- Millennium Pain Center, Bloomington, IL 61704, USA.
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA.
| | - Ramsin Benyamin
- Millennium Pain Center, Bloomington, IL 61704, USA.
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL 61701, USA.
- College of Medicine, Department of Surgery, University of Illinois at Urbana-Champaign, Champaign-Urbana, IL 61801, USA.
| | - David L Cedeño
- Millennium Pain Center, Bloomington, IL 61704, USA.
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL 61701, USA.
| |
Collapse
|
33
|
Spinal cord stimulation prevents paclitaxel-induced mechanical and cold hypersensitivity and modulates spinal gene expression in rats. Pain Rep 2019; 4:e785. [PMID: 31875188 PMCID: PMC6882571 DOI: 10.1097/pr9.0000000000000785] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/06/2019] [Accepted: 08/10/2019] [Indexed: 01/28/2023] Open
Abstract
Supplemental Digital Content is Available in the Text. Introduction: Paclitaxel-induced peripheral neuropathy (PIPN) is a common dose-limiting side effect of this cancer treatment drug. Spinal cord stimulation (SCS) has demonstrated efficacy for attenuating some neuropathic pain conditions. Objective: We aim to examine the inhibitory effect of SCS on the development of PIPN pain and changes of gene expression in the spinal cord in male rats after SCS. Methods: We examined whether traditional SCS (50 Hz, 6–8 h/session daily for 14 consecutive days) administered during paclitaxel treatment (1.5 mg/kg, i.p.) attenuates PIPN-related pain behavior. After SCS treatment, we performed RNA-seq of the lumbar spinal cord to examine which genes are differentially expressed after PIPN with and without SCS. Results: Compared to rats treated with paclitaxel alone (n = 7) or sham SCS (n = 6), SCS treatment (n = 11) significantly inhibited the development of paclitaxel-induced mechanical and cold hypersensitivity, without altering open-field exploratory behavior. RNA-seq showed that SCS induced upregulation of 836 genes and downregulation of 230 genes in the spinal cord of paclitaxel-treated rats (n = 3) as compared to sham SCS (n = 5). Spinal cord stimulation upregulated immune responses in paclitaxel-treated rats, including transcription of astrocyte- and microglial-related genes, but repressed transcription of multiple gene networks associated with synapse transmission, neuron projection development, γ-aminobutyric acid reuptake, and neuronal plasticity. Conclusion: Our findings suggest that traditional SCS may attenuate the development of pain-related behaviors in PIPN rats, possibly by causing aggregate inhibition of synaptic plasticity through upregulation and downregulation of gene networks in the spinal cord.
Collapse
|
34
|
Caylor J, Reddy R, Yin S, Cui C, Huang M, Huang C, Rao R, Baker DG, Simmons A, Souza D, Narouze S, Vallejo R, Lerman I. Spinal cord stimulation in chronic pain: evidence and theory for mechanisms of action. Bioelectron Med 2019; 5:12. [PMID: 31435499 PMCID: PMC6703564 DOI: 10.1186/s42234-019-0023-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/30/2019] [Indexed: 12/30/2022] Open
Abstract
Well-established in the field of bioelectronic medicine, Spinal Cord Stimulation (SCS) offers an implantable, non-pharmacologic treatment for patients with intractable chronic pain conditions. Chronic pain is a widely heterogenous syndrome with regard to both pathophysiology and the resultant phenotype. Despite advances in our understanding of SCS-mediated antinociception, there still exists limited evidence clarifying the pathways recruited when patterned electric pulses are applied to the epidural space. The rapid clinical implementation of novel SCS methods including burst, high frequency and dorsal root ganglion SCS has provided the clinician with multiple options to treat refractory chronic pain. While compelling evidence for safety and efficacy exists in support of these novel paradigms, our understanding of their mechanisms of action (MOA) dramatically lags behind clinical data. In this review, we reconstruct the available basic science and clinical literature that offers support for mechanisms of both paresthesia spinal cord stimulation (P-SCS) and paresthesia-free spinal cord stimulation (PF-SCS). While P-SCS has been heavily examined since its inception, PF-SCS paradigms have recently been clinically approved with the support of limited preclinical research. Thus, wide knowledge gaps exist between their clinical efficacy and MOA. To close this gap, many rich investigative avenues for both P-SCS and PF-SCS are underway, which will further open the door for paradigm optimization, adjunctive therapies and new indications for SCS. As our understanding of these mechanisms evolves, clinicians will be empowered with the possibility of improving patient care using SCS to selectively target specific pathophysiological processes in chronic pain.
Collapse
Affiliation(s)
- Jacob Caylor
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA USA
| | - Rajiv Reddy
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA USA
| | - Sopyda Yin
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA USA
| | - Christina Cui
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA USA
| | - Mingxiong Huang
- Department of Radiology, University of California San Diego School of Medicine, La Jolla, CA USA
- Department of Radiology, VA San Diego Healthcare System, La Jolla, CA USA
| | - Charles Huang
- Department of Radiology, VA San Diego Healthcare System, La Jolla, CA USA
- Department of Bioengineering, Stanford University, Palo Alto, CA USA
| | - Ramesh Rao
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA USA
| | - Dewleen G. Baker
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA USA
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA USA
| | - Alan Simmons
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA USA
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA USA
| | - Dmitri Souza
- Center for Pain Medicine, Western Reserve Hospital. Department of Surgery, Northeast Ohio Medical School (NEOMED), Athens, OH USA
| | - Samer Narouze
- Center for Pain Medicine, Western Reserve Hospital. Department of Surgery, Northeast Ohio Medical School (NEOMED), Athens, OH USA
| | - Ricardo Vallejo
- Basic Science Research, Millennium Pain Center, Bloomington, IL USA
- School of Biological Sciences, Illinois State University, Normal, IL USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL USA
| | - Imanuel Lerman
- Department of Anesthesiology, Center for Pain Medicine, University of California San Diego School of Medicine, La Jolla, CA USA
- VA Center of Excellence for Stress and Mental Health, VA San Diego Healthcare System, La Jolla, CA USA
- Department of Radiology, VA San Diego Healthcare System, La Jolla, CA USA
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA USA
- Present Address: VA San Diego, 3350 La Jolla Village Dr, (MC116A), San Diego, CA 92161 USA
| |
Collapse
|
35
|
Kinfe TM, Asif M, Chakravarthy KV, Deer TR, Kramer JM, Yearwood TL, Hurlemann R, Hussain MS, Motameny S, Wagle P, Nürnberg P, Gravius S, Randau T, Gravius N, Chaudhry SR, Muhammad S. Unilateral L4-dorsal root ganglion stimulation evokes pain relief in chronic neuropathic postsurgical knee pain and changes of inflammatory markers: part II whole transcriptome profiling. J Transl Med 2019; 17:205. [PMID: 31217010 PMCID: PMC6585082 DOI: 10.1186/s12967-019-1952-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/09/2019] [Indexed: 01/08/2023] Open
Abstract
Background In our recent clinical trial, increased peripheral concentrations of pro-inflammatory molecular mediators were determined in complex regional pain syndrome (CRPS) patients. After 3 months adjunctive unilateral, selective L4 dorsal root ganglion stimulation (L4-DRGSTIM), significantly decreased serum IL-10 and increased saliva oxytocin levels were assessed along with an improved pain and functional state. The current study extended molecular profiling towards gene expression analysis of genes known to be involved in the gonadotropin releasing hormone receptor and neuroinflammatory (cytokines/chemokines) signaling pathways. Methods Blood samples were collected from 12 CRPS patients for whole-transcriptome profiling in order to assay 18,845 inflammation-associated genes from frozen blood at baseline and after 3 months L4-DRGSTIM using PANTHER™ pathway enrichment analysis tool. Results Pathway enrichment analyses tools (GOrilla™ and PANTHER™) showed predominant involvement of inflammation mediated by chemokines/cytokines and gonadotropin releasing hormone receptor pathways. Further, screening of differentially regulated genes showed changes in innate immune response related genes. Transcriptomic analysis showed that 21 genes (predominantly immunoinflammatory) were significantly changed after L4-DRGSTIM. Seven genes including TLR1, FFAR2, IL1RAP, ILRN, C5, PKB and IL18 were down regulated and fourteen genes including CXCL2, CCL11, IL36G, CRP, SCGB1A1, IL-17F, TNFRSF4, PLA2G2A, CREB3L3, ADAMTS12, IL1F10, NOX1, CHIA and BDKRB1 were upregulated. Conclusions In our sub-group analysis of L4-DRGSTIM treated CRPS patients, we found either upregulated or downregulated genes involved in immunoinflammatory circuits relevant for the pathophysiology of CRPS indicating a possible relation. However, large biobank-based approaches are recommended to establish genetic phenotyping as a quantitative outcome measure in CRPS patients. Trial registration The study protocol was registered at the 15.11.2016 on German Register for Clinical Trials (DRKS ID 00011267). https://www.drks.de/drks_web/navigate.do?navigationId=trial.HTML&TRIAL_ID=DRKS00011267
Collapse
Affiliation(s)
- Thomas M Kinfe
- Department of Psychiatry, Rheinische Friedrich-Wilhelms University, Sigmund-Freud Street 25, 53105, Bonn, Germany. .,Division of Medical Psychology (NEMO Neuromodulation of Emotions), Rheinische Friedrich-Wilhelms University, Bonn, Germany. .,University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany.
| | - Maria Asif
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany.,Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Krishnan V Chakravarthy
- Department of Anesthesiology and Pain Medicine, University of California, San Diego, CA, USA.,San Diego Health Sciences, VA San Diego Healthcare System, San Diego, CA, USA
| | - Timothy R Deer
- The Spine and Nerve Center of the Virginias, Charleston, WV, USA
| | | | | | - Rene Hurlemann
- Department of Psychiatry, Rheinische Friedrich-Wilhelms University, Sigmund-Freud Street 25, 53105, Bonn, Germany.,Division of Medical Psychology (NEMO Neuromodulation of Emotions), Rheinische Friedrich-Wilhelms University, Bonn, Germany.,University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Muhammad Sajid Hussain
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany.,Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Susanne Motameny
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Prerana Wagle
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Sascha Gravius
- University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Thomas Randau
- University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Nadine Gravius
- University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Shafqat R Chaudhry
- Dept. of Basic Medical Sciences Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Sajjad Muhammad
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|