1
|
Barroso J, Branco P, Apkarian AV. The causal role of brain circuits in osteoarthritis pain. Nat Rev Rheumatol 2025; 21:261-274. [PMID: 40164779 DOI: 10.1038/s41584-025-01234-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
Osteoarthritis (OA) is a leading cause of chronic pain worldwide, resulting in substantial disability and placing a substantial burden on patients and society. The hallmark symptom of OA is joint pain. Despite extensive research, new treatments for OA pain remain limited, partly owing to a lack of understanding of underlying pain mechanisms. For a long time, OA pain was seen as a reflection of nociceptive activity at the joint level, and the brain has been viewed as a passive recipient of such information. In this Review, we challenge these concepts and discuss how, over time, the activation of peripheral nociceptors leads to adaptations in the brain that dictate the properties and experience of OA pain. These adaptations are further influenced by the inherent properties of the brain. We review general concepts that distinguish pain from nociception, present evidence on the incongruity between joint injury and experience of OA pain, and review brain circuits that are crucial in the perception of OA pain. Finally, we propose a model that integrates nociception, spinal-cord mechanisms, and central nervous system dynamics, each contributing uniquely to pain perception. This framework has the potential to inform the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Joana Barroso
- Department of Anaesthesiology and Center for Translational Pain Research, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
- Department of Physical Medicine and Rehabilitation, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Paulo Branco
- Department of Anaesthesiology and Center for Translational Pain Research, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
| | - A Vania Apkarian
- Department of Anaesthesiology and Center for Translational Pain Research, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
- Department of Physical Medicine and Rehabilitation, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
2
|
Islam J, Rahman MT, Ali M, Kc E, Lee HJ, Hyun SH, Park YS. CaMKIIα-NpHR-Mediated Optogenetic Inhibition of DRG Glutamatergic Neurons by Flexible Optic Fiber Alleviates Chronic Neuropathic Pain. Neuromolecular Med 2025; 27:26. [PMID: 40227491 DOI: 10.1007/s12017-025-08848-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: 10/15/2024] [Accepted: 03/22/2025] [Indexed: 04/15/2025]
Abstract
Glutamatergic neurons of the dorsal root ganglion (DRGg) exert a significant effect on peripheral nociceptive signal transmission. However, assessing the explicit modulatory effect of DRGg during chronic neuropathic pain (CNP) with neuromodulation techniques remains largely unexplored. Therefore, we inhibited DRGg by optogenetic stimulation and examined whether it could alleviate CNP and associated anxiety-related behaviors in a chronic compressed DRG (CCD) rat model. The CCD pain model was established by inserting an L-shaped rod into the lumbar 5 (L5) intervertebral foramen, and either AAV2-CaMKIIα-eNpHR3.0-mCherry or AAV2-CaMKIIα-mCherry was injected into the L5 DRG. Flexible optic fibers were implanted to direct yellow light into the L5 DRG. Pain and anxiety-related behavioral responses were assessed using mechanical threshold, mechanical latency, thermal latency, and open field tests. In vivo single-unit extracellular recording from the DRG and ventral posterolateral (VPL) thalamus was performed. CNP and anxiety-related behavioral responses along with increased neural firing activity of the DRG and VPL thalamus were observed in CCD animals. Enhanced expression of nociception-influencing molecules was found in the DRG and spinal dorsal horn (SDH). In contrast during optogenetic stimulation, specific DRGg inhibition markedly alleviated the CNP responses and reduced the DRG and VPL thalamic neural hyperactivity in CCD animals. Inhibition of DRGg also reduced the active expression of nociceptive signal mediators in the DRG and SDH. Taken together, our findings suggest that CaMKIIα-NpHR-mediated optogenetic inhibition of DRGg can produce antinociceptive effects in CCD rats during peripheral nerve injury-induced CNP condition by altering peripheral nociceptive signal input in the spinothalamic tract.
Collapse
Affiliation(s)
- Jaisan Islam
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Md Taufiqur Rahman
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Muhammad Ali
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Elina Kc
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Sang Hwan Hyun
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Young Seok Park
- Department of Neuroscience, College of Medicine, Chungbuk National University, Cheongju, Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, 776, 1 Sunhwanro, Seowon-gu, Cheongju, 28644, Chungbuk, Korea.
| |
Collapse
|
3
|
de Geus TJ, Franken G, Joosten EAJ. Spinal Cord Stimulation Paradigms and Alleviation of Neuropathic Pain Behavior in Experimental Painful Diabetic Polyneuropathy. Neuromodulation 2024; 27:1330-1337. [PMID: 39033461 DOI: 10.1016/j.neurom.2024.06.007] [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: 04/09/2024] [Revised: 06/01/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) is an alternative treatment option for painful diabetic polyneuropathy (PDPN). Differential target multiplexed (DTM)-SCS is proposed to be more effective than conventional (Con)-SCS. Animal studies are essential for understanding SCS mechanisms in PDPN pain relief. Although the Von Frey (VF) test is the gold standard for preclinical pain research, it has limitations. Operant testing using the conditioned place preference (CPP) test provides insights into spontaneous neuropathic pain relief and enhances the translatability of findings. This study aims to 1) use the CPP test to evaluate Con- and DTM-SCS effects on spontaneous neuropathic pain relief in PDPN animals and 2) investigate the correlation between mechanical hypersensitivity alleviation and spontaneous neuropathic pain relief. MATERIAL AND METHODS Diabetes was induced through streptozotocin injection in 32 rats; 16 animals developed PDPN and were implanted with a quadripolar lead. Rats were conditioned for Con-SCS (n = 8) or DTM-SCS (n = 7), and a preference score compared with sham was determined. After conditioning, a 30-minute SCS protocol was conducted. Mechanical sensitivity was assessed using VF before, during, and after SCS. RESULTS There were no significant chamber preference changes for DTM-SCS (p = 0.3449) or Con-SCS (p = 0.3632). Subgroups of responders and nonresponders were identified with significant increases in preference score for responders for both DTM-SCS (-266.6 to 119.8; p = 0.0238; n = 4) and Con-SCS (-350.7 to 88.46; p = 0.0148; n = 3). No strong correlation between SCS-induced spontaneous neuropathic pain relief and effects on mechanical hypersensitivity in PDPN animals is noted. CONCLUSIONS The CPP test is a valuable tool to test the efficacy of the pain-relieving potential of various SCS paradigms in PDPN animals. The results of this study show no differences in spontaneous neuropathic pain relief between DTM- and Con-SCS in PDPN animals. Furthermore, there is no correlation between the effect of SCS in spontaneous pain relief and hind paw mechanical hypersensitivity.
Collapse
Affiliation(s)
- Thomas J de Geus
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.
| | - Glenn Franken
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
4
|
Ting JE, Hooper CA, Dalrymple AN, Weber DJ. Tonic Stimulation of Dorsal Root Ganglion Results in Progressive Decline in Recruitment of Aα/β-Fibers in Rats. Neuromodulation 2024; 27:1347-1359. [PMID: 39046395 PMCID: PMC11625011 DOI: 10.1016/j.neurom.2024.06.498] [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: 03/23/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVES In this study, we aimed to characterize the recruitment and maintenance of action potential firing in Aα/β-fibers generated during tonic dorsal root ganglion stimulation (DRGS) applied over a range of clinically relevant stimulation parameters. MATERIALS AND METHODS We delivered electrical stimulation to the L5 dorsal root ganglion and recorded antidromic evoked compound action potentials (ECAPs) in the sciatic nerve during DRGS in Sprague Dawley rats. We measured charge thresholds to elicit ECAPs in Aα/β-fibers during DRGS applied at multiple pulse widths (50, 150, 300, 500 μs) and frequencies (5, 20, 50, 100 Hz). We measured the peak-to-peak amplitudes, latencies, and widths of ECAPs generated during 180 seconds of DRGS, and excitation threshold changes to investigate potential mechanisms of ECAP suppression. RESULTS Tonic DRGS produced ECAPs in Aα/β-fibers at charge thresholds below the motor threshold. Increasing the pulse width of DRGS led to a significant increase in the charge required to elicit ECAPs in Aα/β-fibers, while varying DRGS frequency did not influence ECAP thresholds. Over the course of 180 seconds, ECAP peak-to-peak amplitude decreased progressively in a frequency-dependent manner, where 5- and 100-Hz DRGS resulted in 22% and 87% amplitude reductions, respectively, and ECAP latencies increased from baseline measurements during DRGS at 10, 20, 50, and 100 Hz. Regardless of DRGS frequency, ECAP amplitudes recovered within 120 seconds after turning DRGS off. We determined that ECAP suppression may be attributed to increasing excitation thresholds for individual fibers during DRGS. Following 180 seconds of DRGS, an average of 7.33% increase in stimulation amplitude was required to restore the ECAP to baseline amplitude. CONCLUSIONS DRGS produces a progressive and frequency-dependent reduction in ECAP amplitude that occurs within and above the frequency range used clinically to relieve pain. If DRGS-mediated analgesia relies on Aβ-fiber activation, then the frequency or duty cycle of stimulation should be set to the lowest effective level to maintain sufficient activation of Aβ-fibers.
Collapse
Affiliation(s)
- Jordyn E Ting
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charli Ann Hooper
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ashley N Dalrymple
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Department of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, UT, USA
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Geus TD, Franken G, Zuidema X, van Zundert J, Joosten EAJ. Structural changes in the nociceptive system induced by long-term conventional spinal cord stimulation in experimental painful diabetic polyneuropathy. Reg Anesth Pain Med 2024:rapm-2024-105919. [PMID: 39542640 DOI: 10.1136/rapm-2024-105919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Clinical studies suggest that long-term conventional spinal cord stimulation (LT-SCS) for painful diabetic peripheral neuropathy (PDPN) is initially effective but may decline in efficacy over time. Preclinical studies indicate that LT-SCS alleviates mechanical hypersensitivity and enhances hind paw blood flow in PDPN rats, suggesting nociceptive system plasticity. This study hypothesized that LT-SCS induces peripheral hind paw small-fiber sprouting and reduces central protein expression of glial and P2X4 brain-derived neurotrophic factor (BDNF) pathway markers. METHODS Diabetes was induced via Streptozotocin injection in 32 rats, with 16 developing PDPN and receiving a quadrupolar lead implant. LT-SCS was applied for 4 weeks, 12 hours per day. Pain behavior was assessed using the Von Frey test for mechanical hypersensitivity and the mechanical conflict avoidance system for motivational aspects of pain. Fiber sprouting was assessed via immunohistochemical analysis of nerve fibers in the hind paw skin. Protein expression in the spinal cord was assessed using western blotting. RESULTS LT-SCS increased the baseline threshold of mechanical hypersensitivity in PDPN animals, consistent with previous findings, but showed no effects on motivational aspects of pain. Hind paw tissue analysis revealed significantly increased intraepidermal nerve fiber density of PGP9.5 fibers in LT-SCS animals compared with Sham-SCS animals. Protein analysis showed significantly decreased pro-BDNF expression in LT-SCS animals compared with Sham-SCS animals. CONCLUSION LT-SCS induces structural changes in both peripheral and central components of the nociceptive system in PDPN animals. These changes may contribute to observed behavioral modifications, elucidating mechanisms underlying LT-SCS efficacy in PDPN management.
Collapse
Affiliation(s)
- Thomas de Geus
- Department of Anesthesiology and Pain Management, MUMC+, Maastricht, The Netherlands
- Maastricht University School for Mental Health and Neuroscience, Maastricht, The Netherlands
| | - Glenn Franken
- Department of Anesthesiology and Pain Management, MUMC+, Maastricht, The Netherlands
- Maastricht University School for Mental Health and Neuroscience, Maastricht, The Netherlands
| | - Xander Zuidema
- Department of Anesthesiology and Pain Management, MUMC+, Maastricht, The Netherlands
- Department of Anesthesiology and Pain Management, Diakonessenhuis Utrecht Zeist Doorn, Utrecht, The Netherlands
| | - Jan van Zundert
- Department of Anesthesiology and Pain Management, MUMC+, Maastricht, The Netherlands
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Multidisciplinary Pain Center, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, MUMC+, Maastricht, The Netherlands
- Maastricht University School for Mental Health and Neuroscience, Maastricht, The Netherlands
| |
Collapse
|
6
|
Ladez SR, Liu J, Chen L, Feng B. Computational Modeling of Dorsal Root Ganglion Stimulation: Understanding Pain Suppression Mechanisms. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039770 DOI: 10.1109/embc53108.2024.10782923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
This study aims to advance our mechanistic understanding of electrical stimulation of dorsal root ganglia (DRG) for treating chronic pain. While DRG stimulation has shown moderate clinical success in managing certain types of chronic pain, the underlying neural mechanism remains inconclusive, hindering the further development of the technology to treat a broader range of chronic pain symptoms and benefit a larger patient population. In this study, we conducted computational simulations in the NEURON simulation environment to assess the neuromodulatory effect of DRG stimulation on action potential transmission in Aδ-fiber and C-fiber sensory afferents. Our simulation incorporates Markov-type state models to capture the subtle gating characteristics of voltage-gated sodium channel subtypes, especially NaV1.6, the anatomical distribution of which was revealed by our immunohistological staining on sparsely labeled afferents. Our simulation results indicate that DRG stimulation causes a significant increase in intra-axonal Na+ concentration and a reduction in K+ concentration, collectively disrupting the transaxonal ionic gradients. This disruption resulted in activitydependent conduction slowing, leading to the eventual conduction block in both Aδ- and C-fiber afferents. This research marks a crucial step forward in unraveling the intricate mechanisms underlying DRG stimulation, presenting a framework for the further development of innovative pain modulation strategies that target the DRG.
Collapse
|
7
|
Chapman KB, Amireh A, van Helmond N, Yousef TA. Evaluation of Washout Periods After Dorsal Root Ganglion Stimulation Trial. Neuromodulation 2024; 27:881-886. [PMID: 38551547 DOI: 10.1016/j.neurom.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVE Dorsal root ganglion stimulation (DRG-S) is a novel therapy to treat chronic pain. It has shown efficacy when delivered intermittently, suggesting a delayed washout effect exists. To measure the washout period, and to determine whether there are differences in washout times among different types of treated pain, we measured the time for pain to return at the end of the patients' one-week DRG stimulation trials. MATERIALS AND METHODS Patients who completed a successful DRG-S trial were included. The times until 25% (t25) and 90% (t90) of baseline pain level returned were recorded. The patients were divided into neuropathic, nociceptive, and mixed pain groups for subgroup comparison. t25 and t90 were plotted in the entire cohort and subgroups using reverse Kaplan-Meier plots (failure curves) and compared using a log-rank test. RESULTS In total, 29 consecutive patients were included. Median t25 and t90 times were 7.1 and 19.5 hours, respectively. Median (interquartile range) times were longest for the nociceptive pain group (n = 17) and shortest for the neuropathic pain group (n = 6), with the mixed-pain group (n = 6) in between (t25: 7.1 [1.7-19.4], 3.40 [1.4-8.4], and 5.7 [0.8-17.6]; t90, 22.0 [10.7-71.0], 7.6 [3.6-19.8], and 20.9 [14.2-31.2], respectively). t90 times differed significantly by pain type (p = 0.040). CONCLUSIONS This study showed a prolonged washout period after cessation of DRG-S therapy. Washout times vary according to pain type. The observed effects are possibly due to long-term depression of pain signaling and could allow the implementation of alternative stimulation strategies with DRG-S. Further investigations evaluating DRG-S washout times are warranted.
Collapse
Affiliation(s)
- Kenneth B Chapman
- The Spine & Pain Institute of New York, New York City, NY, USA; Department of Anesthesiology, New York University Langone Medical Center, New York City, NY, USA; The Zucker School of Medicine at Hofstra/Northwell, New York City, NY, USA; Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Nijmegen, Nijmegen, The Netherlands.
| | - Ahmad Amireh
- The Spine & Pain Institute of New York, New York City, NY, USA
| | - Noud van Helmond
- The Spine & Pain Institute of New York, New York City, NY, USA; Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Tariq A Yousef
- The Spine & Pain Institute of New York, New York City, NY, USA
| |
Collapse
|
8
|
Sivanesan E, North RB, Russo MA, Levy RM, Linderoth B, Hayek SM, Eldabe S, Lempka SF. A Definition of Neuromodulation and Classification of Implantable Electrical Modulation for Chronic Pain. Neuromodulation 2024; 27:1-12. [PMID: 37952135 DOI: 10.1016/j.neurom.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/24/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVES Neuromodulation therapies use a variety of treatment modalities (eg, electrical stimulation) to treat chronic pain. These therapies have experienced rapid growth that has coincided with escalating confusion regarding the nomenclature surrounding these neuromodulation technologies. Furthermore, studies are often published without a complete description of the effective stimulation dose, making it impossible to replicate the findings. To improve clinical care and facilitate dissemination among the public, payors, research groups, and regulatory bodies, there is a clear need for a standardization of terms. APPROACH We formed an international group of authors comprising basic scientists, anesthesiologists, neurosurgeons, and engineers with expertise in neuromodulation. Because the field of neuromodulation is extensive, we chose to focus on creating a taxonomy and standardized definitions for implantable electrical modulation of chronic pain. RESULTS We first present a consensus definition of neuromodulation. We then describe a classification scheme based on the 1) intended use (the site of modulation and its indications) and 2) physical properties (waveforms and dose) of a neuromodulation therapy. CONCLUSIONS This framework will help guide future high-quality studies of implantable neuromodulatory treatments and improve reporting of their findings. Standardization with this classification scheme and clear definitions will help physicians, researchers, payors, and patients better understand the applications of implantable electrical modulation for pain and guide informed treatment decisions.
Collapse
Affiliation(s)
- Eellan Sivanesan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Richard B North
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
| | - Marc A Russo
- Hunter Pain Specialists, Broadmeadow, New South Wales, Australia
| | - Robert M Levy
- Neurosurgical Services, Clinical Research, Anesthesia Pain Care Consultants, Tamarac, FL, USA
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Salim M Hayek
- Division of Pain Medicine, University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
| | - Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Wahezi SE, Caparo MA, Malhotra R, Sundaram L, Batti K, Ejindu P, Veeramachaneni R, Anitescu M, Hunter CW, Naeimi T, Farah F, Kohan L. Current Waveforms in Spinal Cord Stimulation and Their Impact on the Future of Neuromodulation: A Scoping Review. Neuromodulation 2024; 27:47-58. [PMID: 38184341 DOI: 10.1016/j.neurom.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/16/2023] [Accepted: 11/13/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND Neuromodulation is a standard and well-accepted treatment for chronic refractory neuropathic pain. There has been progressive innovation in the field over the last decade, particularly in areas of spinal cord stimulation (SCS) and dorsal root ganglion stimulation. Improved outcomes using proprietary waveforms have become customary in the field, leading to an unprecedented expansion of these products and a plethora of options for the management of pain. Although advances in waveform technology have improved our fundamental understanding of neuromodulation, a scoping review describing new energy platforms and their associated clinical effects and outcomes is needed. The authors submit that understanding electrophysiological neuromodulation may be important for clinical decision-making and programming selection for personalized patient care. OBJECTIVE This review aims to characterize ways differences in mechanism of action and clinical outcomes of current spinal neuromodulation products may affect contemporary clinical decision-making while outlining a possible path for the future SCS. STUDY DESIGN The study is a scoping review of the literature about newer generation SCS waveforms. MATERIALS AND METHODS A literature report was performed on PubMed and chapters to include articles on spine neuromodulation mechanism of action and efficacy. RESULTS A total of 8469 studies were identified, 75 of which were included for the scoping review after keywords defining recent waveform technology were added. CONCLUSIONS Clinical data suggest that neuromodulation remains a promising tool in the treatment of chronic pain. The evidence for SCS for treating chronic pain seems compelling; however, more long-term and comparative data are needed for a comparison of waveforms when it comes to the etiology of pain. In addition, an exploration into combination waveform therapy and waveform cycling may be paramount for future clinical studies and the development of new technologies.
Collapse
Affiliation(s)
- Sayed E Wahezi
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA.
| | - Moorice A Caparo
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Ria Malhotra
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Lakshman Sundaram
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Kevin Batti
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Prince Ejindu
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | | | - Magdalena Anitescu
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Corey W Hunter
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Tahereh Naeimi
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Fadi Farah
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| | - Lynn Kohan
- Multidisciplinary Pain Program, Montefiore Medical Center, Bronx, New York, USA
| |
Collapse
|
10
|
Modi AD, Parekh A, Pancholi YN. Evaluating Pain Behaviours: Widely Used Mechanical and Thermal Methods in Rodents. Behav Brain Res 2023; 446:114417. [PMID: 37003494 DOI: 10.1016/j.bbr.2023.114417] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/13/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Globally, over 300 million surgical procedures are performed annually, with pain being one of the most common post-operative side effects. During the onset of injury, acute pain plays a protective role in alerting the individual to remove noxious stimuli, while long-lasting chronic pain without any physiological reason is detrimental to the recovery process. Hence, it created an urgent need to better understand the pain mechanism and explore therapeutic targets. Despite the hardship in performing human pain studies due to ethical considerations, clinically relevant rodent pain models provide an excellent opportunity to perform pain studies. Several neurobehavioural tests are used to assess the drug efficacy in rodents to determine avoidance behaviour latency and threshold. This review article provides a methodological overview of mechanical (i.e. von Frey, Mechanical Conflict System) and thermal (i.e. Hargreaves Assay, Hot and Cold Plate, Temperature Place Preference) tests to assess pain in clinically relevant pain rodent models. We further discussed the current modifications of those tests along with their use in literature, the impact of confounding variables, advantages and disadvantages.
Collapse
Affiliation(s)
- Akshat D Modi
- Department of Biological Sciences, University of Toronto, Scarborough, Ontario M1C 1A4, Canada; Department of Genetics and Development, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada.
| | - Anavi Parekh
- Department of Neuroscience, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Yajan N Pancholi
- Department of Neuroscience, University of Toronto, Scarborough, Ontario M1C 1A4, Canada
| |
Collapse
|
11
|
Chapman KB, Sayed D, Lamer T, Hunter C, Weisbein J, Patel KV, Dickerson D, Hagedorn JM, Lee DW, Amirdelfan K, Deer T, Chakravarthy K. Best Practices for Dorsal Root Ganglion Stimulation for Chronic Pain: Guidelines from the American Society of Pain and Neuroscience. J Pain Res 2023; 16:839-879. [PMID: 36942306 PMCID: PMC10024474 DOI: 10.2147/jpr.s364370] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 01/17/2023] [Indexed: 03/14/2023] Open
Abstract
With continued innovations in neuromodulation comes the need for evolving reviews of best practices. Dorsal root ganglion stimulation (DRG-S) has significantly improved the treatment of complex regional pain syndrome (CRPS), and it has broad applicability across a wide range of other conditions. Through funding and organizational leadership by the American Society for Pain and Neuroscience (ASPN), this best practices consensus document has been developed for the selection, implantation, and use of DRG stimulation for the treatment of chronic pain syndromes. This document is composed of a comprehensive narrative literature review that has been performed regarding the role of the DRG in chronic pain and the clinical evidence for DRG-S as a treatment for multiple pain etiologies. Best practice recommendations encompass safety management, implantation techniques, and mitigation of the potential complications reported in the literature. Looking to the future of neuromodulation, DRG-S holds promise as a robust intervention for otherwise intractable pain.
Collapse
Affiliation(s)
- Kenneth B Chapman
- The Spine & Pain Institute of New York, New York, NY, USA
- Department of Anesthesiology, Zucker School of Medicine at Hofstra Northwell, Manhasset, NY, USA
- Department of Anesthesiology, NYU Langone Medical Center, New York, NY, USA
| | - Dawood Sayed
- Department of Anesthesiology, The University of Kansas Medical Center (KUMC), Kansas City, KS, USA
| | - Tim Lamer
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, Mayo Clinic, Rochester, MN, USA
| | - Corey Hunter
- Ainsworth Institute of Pain Management, New York, NY, USA
| | | | - Kiran V Patel
- The Spine & Pain Institute of New York, New York, NY, USA
- Department of Anesthesiology, Zucker School of Medicine at Hofstra Northwell, Manhasset, NY, USA
- Department of Anesthesiology, NYU Langone Medical Center, New York, NY, USA
| | - David Dickerson
- Department of Anesthesiology, Critical Care and Pain Medicine, NorthShore University Health System, Evanston, IL, USA
- Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, USA
| | | | - David W Lee
- Fullerton Orthopedic Surgery Medical Group, Fullerton, CA, USA
| | | | - Timothy Deer
- The Spine and Nerve Center of the Virginias, Charleston, WV, USA
| | - Krishnan Chakravarthy
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
12
|
Chapman KB, Tupper C, Vissers KC, van Helmond N, Yousef T. Dorsal root ganglion stimulation for the treatment of joint pain with predominantly nociceptive characteristics: A case series. Pain Pract 2023; 23:317-324. [PMID: 36334041 DOI: 10.1111/papr.13180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/28/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Dorsal root ganglion stimulation (DRG-S) has recently emerged as a novel therapy in neuromodulation that demonstrated a higher rate of success than spinal cord stimulation (SCS) in a prospective, head-to-head randomized comparative trial to treat complex regional pain syndrome (CRPS) and causalgia. In contrast to SCS, DRG-S also shows promise in treating conditions that are not purely neuropathic such as axial low back pain, which has a prominent nociplastic pain component. It is not known to what extent the effectiveness of DRG-S for such indications is due to effective treatment of the neuropathic pain component versus the effects of DRG-S on mechanical pain. Although rarely studied, reporting outcomes of DRG-S to treat predominantly mechanical/nociceptive pain may help point toward expanding the utility of this therapy. Here, we present five cases of refractory mechanical pain treated with DRG-S. METHODS A retrospective analysis of all patients who underwent a successful DRG-S trial and implant between September 2017 and September 2021 at our institute was performed. Patients who had intractable joint pain without strong evidence of neuropathic pain were included in this case series. The Budapest criteria for CRPS, the Douleur Neuropathique 4 Questions (DN4) survey, or a definable nerve injury were used to determine the presence of neuropathic pain. Baseline assessments for pain (Numeric Rating Scale [NRS]), function (Oswestry Disability Index [ODI]), quality of life (EuroQol-5 Dimension [EQ-5D]), and other applicable joint surveys were extracted from pre-trial baseline and follow-up appointments. RESULTS Five patients were identified and included. Patient diagnoses consisted of refractory joint pain of the hip, knee, or ankle. Mean NRS pain scores improved by 74% from 9.2 at baseline to 2.4 at the last follow-up (mean = 28 months post-implant). From baseline to the last follow-up, mean ODI scores improved by 65% from 66 to 23 and EQ-5D scores more than doubled from an average of 0.371 to 0.797. CONCLUSION This clinical report illustrates the potential utility DRG-S has in treating pain that clinically presents as predominantly refractory mechanical joint pain without a significant neuropathic component. The physiological reasons for our observations may be that DRG-S is able to directly influence the conduction of nociceptive signaling at the DRG and within the spinal cord. Further investigations are warranted to determine if DRG-S is a potential treatment option for chronic mechanical pain.
Collapse
Affiliation(s)
- Kenneth B Chapman
- Spine & Pain Institute of New York, New York, New York, USA.,Department of Anesthesiology, New York University Langone Medical Center, New York, New York, USA.,Departement of Anesthesiology, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Connor Tupper
- Creighton University Medical College, Omaha, Nebraska, USA
| | - Kris C Vissers
- Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Noud van Helmond
- Spine & Pain Institute of New York, New York, New York, USA.,Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Tariq Yousef
- Spine & Pain Institute of New York, New York, New York, USA
| |
Collapse
|
13
|
Kuwabara Y, Howard-Quijano K, Salavatian S, Yamaguchi T, Saba S, Mahajan A. Thoracic dorsal root ganglion stimulation reduces acute myocardial ischemia induced ventricular arrhythmias. Front Neurosci 2023; 17:1091230. [PMID: 36793544 PMCID: PMC9922704 DOI: 10.3389/fnins.2023.1091230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Background Dorsal root ganglion stimulation (DRGS) may serve as a novel neuromodulation strategy to reduce cardiac sympathoexcitation and ventricular excitability. Objective In this pre-clinical study, we investigated the effectiveness of DRGS on reducing ventricular arrhythmias and modulating cardiac sympathetic hyperactivity caused by myocardial ischemia. Methods Twenty-three Yorkshire pigs were randomized to two groups, which was control LAD ischemia-reperfusion (CONTROL) or LAD ischemia-reperfusion + DRGS (DRGS) group. In the DRGS group (n = 10), high-frequency stimulation (1 kHz) at the second thoracic level (T2) was initiated 30 min before ischemia and continued throughout 1 h of ischemia and 2 h of reperfusion. Cardiac electrophysiological mapping and Ventricular Arrhythmia Score (VAS) were assessed, along with evaluation of cFos expression and apoptosis in the T2 spinal cord and DRG. Results DRGS decreased the magnitude of activation recovery interval (ARI) shortening in the ischemic region (CONTROL: -201 ± 9.8 ms, DRGS: -170 ± 9.4 ms, p = 0.0373) and decreased global dispersion of repolarization (DOR) at 30 min of myocardial ischemia (CONTROL: 9546 ± 763 ms2, DRGS: 6491 ± 636 ms2, p = 0.0076). DRGS also decreased ventricular arrhythmias (VAS-CONTROL: 8.9 ± 1.1, DRGS: 6.3 ± 1.0, p = 0.038). Immunohistochemistry studies showed that DRGS decreased % cFos with NeuN expression in the T2 spinal cord (p = 0.048) and the number of apoptotic cells in the DRG (p = 0.0084). Conclusion DRGS reduced the burden of myocardial ischemia-induced cardiac sympathoexcitation and has a potential to be a novel treatment option to reduce arrhythmogenesis.
Collapse
Affiliation(s)
- Yuki Kuwabara
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kimberly Howard-Quijano
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Siamak Salavatian
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Tomoki Yamaguchi
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Samir Saba
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Aman Mahajan
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Cruz CJ, Dewberry LS, Otto KJ, Allen KD. Neuromodulation as a Potential Disease-Modifying Therapy for Osteoarthritis. Curr Rheumatol Rep 2023; 25:1-11. [PMID: 36435890 PMCID: PMC11438129 DOI: 10.1007/s11926-022-01094-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW The following review discusses the therapeutic potential of targeting the autonomic nervous system (ANS) for osteoarthritis (OA) treatment and encourages the field to consider the candidacy of bioelectronic medicine as a novel OA treatment strategy. RECENT FINDINGS The study of OA pathogenesis has focused on changes occurring at the joint level. As such, treatments for OA have been aimed at the local joint environment, intending to resolve local inflammation and decrease pain. However, OA pathogenesis has shown to be more than joint wear and tear. Specifically, OA-related peripheral and central sensitization can prompt neuroplastic changes in the nervous system beyond the articular joint. These neuroplastic changes may alter physiologic systems, like the neuroimmune axis. In this way, OA and related comorbidities may share roots in the form of altered neuroimmune communication and autonomic dysfunction. ANS modulation may be able to modify OA pathogenesis or reduce the impact of OA comorbidities. Moreover, blocking chronic nociceptive drive from the joint may help to prevent maladaptive nervous system plasticity in OA.
Collapse
Affiliation(s)
- Carlos J Cruz
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
- Pain Research and Intervention Center of Excellence, Gainesville, FL, USA
| | - L Savannah Dewberry
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
| | - Kevin J Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
- Department of Neurology, University of Florida, Gainesville, FL, USA
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA.
- Pain Research and Intervention Center of Excellence, Gainesville, FL, USA.
- Department of Orthopaedics and Rehabilitation, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
15
|
Chao D, Tran H, Hogan QH, Pan B. Analgesic dorsal root ganglion field stimulation blocks both afferent and efferent spontaneous activity in sensory neurons of rats with monosodium iodoacetate-induced osteoarthritis. Osteoarthritis Cartilage 2022; 30:1468-1481. [PMID: 36030058 PMCID: PMC9588581 DOI: 10.1016/j.joca.2022.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Chronic joint pain is common in patients with osteoarthritis (OA). Non-steroidal anti-inflammatory drugs and opioids are used to relieve OA pain, but they are often inadequately effective. Dorsal root ganglion field stimulation (GFS) is a clinically used neuromodulation approach, although it is not commonly employed for patients with OA pain. GFS showed analgesic effectiveness in our previous study using the monosodium iodoacetate (MIA) - induced OA rat pain model. This study was to evaluate the mechanism of GFS analgesia in this model. METHODS After osteoarthritis was induced by intra-articular injection of MIA, pain behavioral tests were performed. Effects of GFS on the spontaneous activity (SA) were tested with in vivo single-unit recordings from teased fiber saphenous nerve, sural nerve, and dorsal root. RESULTS Two weeks after intra-articular MIA injection, rats developed pain-like behaviors. In vivo single unit recordings from bundles teased from the saphenous nerve and third lumbar (L3) dorsal root of MIA-OA rats showed a higher incidence of SA than those from saline-injected control rats. GFS at the L3 level blocked L3 dorsal root SA. MIA-OA reduced the punctate mechanical force threshold for inducing AP firing in bundles teased from the L4 dorsal root, which reversed to normal with GFS. After MIA-OA, there was increased retrograde SA (dorsal root reflex), which can be blocked by GFS. CONCLUSIONS These results indicate that GFS produces analgesia in MIA-OA rats at least in part by producing blockade of afferent inputs, possibly also by blocking efferent activity from the dorsal horn.
Collapse
Affiliation(s)
- D Chao
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA
| | - H Tran
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA
| | - Q H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA
| | - B Pan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|
16
|
Itson-Zoske B, Shin SM, Xu H, Qiu C, Fan F, Hogan QH, Yu H. Selective block of sensory neuronal T-type/Cav3.2 activity mitigates neuropathic pain behavior in a rat model of osteoarthritis pain. Arthritis Res Ther 2022; 24:168. [PMID: 35842727 PMCID: PMC9287929 DOI: 10.1186/s13075-022-02856-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Peripheral and central nociceptive sensitization is a critical pathogenetic component in osteoarthritis (OA) chronic pain. T-type calcium channel 3.2 (CaV3.2) regulates neuronal excitability and plays important roles in pain processing. We previously identified that enhanced T-type/CaV3.2 activity in the primary sensory neurons (PSNs) of dorsal root ganglia (DRG) is associated with neuropathic pain behavior in a rat model of monosodium iodoacetate (MIA)-induced knee OA. PSN-specific T-type/CaV3.2 may therefore represent an important mediator in OA painful neuropathy. Here, we test the hypothesis that the T-type/CaV3.2 channels in PSNs can be rationally targeted for pain relief in MIA-OA. METHODS MIA model of knee OA was induced in male and female rats by a single injection of 2 mg MIA into intra-knee articular cavity. Two weeks after induction of knee MIA-OA pain, recombinant adeno-associated viruses (AAV)-encoding potent CaV3.2 inhibitory peptide aptamer 2 (CaV3.2iPA2) that have been characterized in our previous study were delivered into the ipsilateral lumbar 4/5 DRG. Effectiveness of DRG-CaV3.2iPA2 treatment on evoked (mechanical and thermal) and spontaneous (conditioned place preference) pain behavior, as well as weight-bearing asymmetry measured by Incapacitance tester, in the arthritic limbs of MIA rats were evaluated. AAV-mediated transgene expression in DRG was determined by immunohistochemistry. RESULTS AAV-mediated expression of CaV3.2iPA2 selective in the DRG-PSNs produced significant and comparable mitigations of evoked and spontaneous pain behavior, as well as normalization of weight-bearing asymmetry in both male and female MIA-OA rats. Analgesia of DRG-AAV-CaV3.2iPA1, another potent CaV3.2 inhibitory peptide, was also observed. Whole-cell current-clamp recordings showed that AAV-mediated CaV3.2iPA2 expression normalized hyperexcitability of the PSNs dissociated from the DRG of MIA animals, suggesting that CaV3.2iPA2 attenuated pain behavior by reversing MIA-induced neuronal hyperexcitability. CONCLUSIONS Together, our results add therapeutic support that T-type/CaV3.2 in primary sensory pathways contributes to MIA-OA pain pathogenesis and that CaV3.2iPAs are promising analgesic leads that, combined with AAV-targeted delivery in anatomically segmental sensory ganglia, have the potential for further development as a peripheral selective T-type/CaV3.2-targeting strategy in mitigating chronic MIA-OA pain behavior. Validation of the therapeutic potential of this strategy in other OA models may be valuable in future study.
Collapse
Affiliation(s)
- Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hao Xu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chensheng Qiu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fan Fan
- Department of Pharmacology and Toxicology, Mississippi University Medical Center, Jackson, MS, 39216, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
17
|
Chapman KB, Yang A, Mogilner AY, Mandelberg N, Patel KV, Lubenow T, Deer T, Kallewaard JW, van Helmond N. Dorsal Root Ganglion Stimulation Device Explantation: A Multicenter Pooled Data Analysis. Pain Pract 2022; 22:522-531. [DOI: 10.1111/papr.13113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Kenneth B. Chapman
- The Spine & Pain Institute of New York, NY NY USA
- Department of Anesthesiology, NYU Langone Medical Center, NY NY USA
- Northwell Health New York City NY USA
| | - Ajax Yang
- The Spine & Pain Institute of New York, NY NY USA
- Northwell Health New York City NY USA
| | - Alon Y. Mogilner
- Department of Neurosurgery, NYU Langone Medical Center, NY NY USA
| | | | - Kiran V. Patel
- The Spine & Pain Institute of New York, NY NY USA
- Northwell Health New York City NY USA
| | - Timothy Lubenow
- Department of Anesthesiology Rush University Medical Center Chicago IL
| | - Timothy Deer
- The Spine and Nerve Center of the Virginias Charleston WV
| | | | - Noud van Helmond
- The Spine & Pain Institute of New York, NY NY USA
- Cooper Medical School of Rowan University Cooper University Hospital Camden NJ USA
| |
Collapse
|
18
|
Chen L, Guo T, Zhang S, Smith PP, Feng B. Blocking peripheral drive from colorectal afferents by subkilohertz dorsal root ganglion stimulation. Pain 2022; 163:665-681. [PMID: 34232925 PMCID: PMC8720331 DOI: 10.1097/j.pain.0000000000002395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 06/16/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Clinical evidence indicates dorsal root ganglion (DRG) stimulation effectively reduces pain without the need to evoke paresthesia. This paresthesia-free anesthesia by DRG stimulation can be promising to treat pain from the viscera, where paresthesia usually cannot be produced. Here, we explored the mechanisms and parameters for DRG stimulation using an ex vivo preparation with mouse distal colon and rectum (colorectum), pelvic nerve, L6 DRG, and dorsal root in continuity. We conducted single-fiber recordings from split dorsal root filaments and assessed the effect of DRG stimulation on afferent neural transmission. We determined the optimal stimulus pulse width by measuring the chronaxies of DRG stimulation to be below 216 µs, indicating spike initiation likely at attached axons rather than somata. Subkilohertz DRG stimulation significantly attenuates colorectal afferent transmission (10, 50, 100, 500, and 1000 Hz), of which 50 and 100 Hz show superior blocking effects. Synchronized spinal nerve and DRG stimulation reveals a progressive increase in conduction delay by DRG stimulation, suggesting activity-dependent slowing in blocked fibers. Afferents blocked by DRG stimulation show a greater increase in conduction delay than the unblocked counterparts. Midrange frequencies (50-500 Hz) are more efficient at blocking transmission than lower or higher frequencies. In addition, DRG stimulation at 50 and 100 Hz significantly attenuates in vivo visceromotor responses to noxious colorectal balloon distension. This reversible conduction block in C-type and Aδ-type afferents by subkilohertz DRG stimulation likely underlies the paresthesia-free anesthesia by DRG stimulation, thereby offering a promising new approach for managing chronic visceral pain.
Collapse
Affiliation(s)
- Longtu Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269
| | - Tiantian Guo
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269
| | - Shaopeng Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269
| | - Phillip P. Smith
- School of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269
| |
Collapse
|
19
|
Graham RD, Sankarasubramanian V, Lempka SF. Dorsal Root Ganglion Stimulation for Chronic Pain: Hypothesized Mechanisms of Action. THE JOURNAL OF PAIN 2022; 23:196-211. [PMID: 34425252 PMCID: PMC8943693 DOI: 10.1016/j.jpain.2021.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/28/2021] [Accepted: 07/20/2021] [Indexed: 02/03/2023]
Abstract
Dorsal root ganglion stimulation (DRGS) is a neuromodulation therapy for chronic pain that is refractory to conventional medical management. Currently, the mechanisms of action of DRGS-induced pain relief are unknown, precluding both our understanding of why DRGS fails to provide pain relief to some patients and the design of neurostimulation technologies that directly target these mechanisms to maximize pain relief in all patients. Due to the heterogeneity of sensory neurons in the dorsal root ganglion (DRG), the analgesic mechanisms could be attributed to the modulation of one or many cell types within the DRG and the numerous brain regions that process sensory information. Here, we summarize the leading hypotheses of the mechanisms of DRGS-induced analgesia, and propose areas of future study that will be vital to improving the clinical implementation of DRGS. PERSPECTIVE: This article synthesizes the evidence supporting the current hypotheses of the mechanisms of action of DRGS for chronic pain and suggests avenues for future interdisciplinary research which will be critical to fully elucidate the analgesic mechanisms of the therapy.
Collapse
Affiliation(s)
- Robert D. Graham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Vishwanath Sankarasubramanian
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Scott F. Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States,Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, United States,Corresponding author: Scott F. Lempka, PhD, Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Road, NCRC 14-184, Ann Arbor, MI 48109-2800,
| |
Collapse
|
20
|
Graham RD, Jhand AS, Lempka SF. Dorsal root ganglion stimulation produces differential effects on action potential propagation across a population of biophysically distinct C-neurons. FRONTIERS IN PAIN RESEARCH 2022; 3:1017344. [PMID: 36387415 PMCID: PMC9643723 DOI: 10.3389/fpain.2022.1017344] [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: 08/11/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022] Open
Abstract
Dorsal root ganglion stimulation (DRGS) is a neurostimulation therapy used to manage chronic pain that does not respond to conventional therapies. Unfortunately, not all patients receive sufficient pain relief from DRGS, leaving them with few other treatment options. Presently, our understanding of the mechanisms of action of DRGS is incomplete, preventing us from determining why some patients do not receive analgesia from the therapy. One hypothesis suggests that DRGS augments the filtering of action potentials (APs) at the T-junction of nociceptive C-neurons. To test this hypothesis, we utilized a computational modeling approach in which we developed a population of one thousand biophysically distinct C-neuron models which each produced electrophysiological characteristics (e.g., AP height, AP duration) reported in previous experimental studies. We used this population of model C-neurons to study how morphological and electrophysiological characteristics affected the propagation of APs through the T-junction. We found that trains of APs can propagate through the T-junction in the orthodromic direction at a higher frequency than in the antidromic direction due to the decrease in axonal diameter from the peripheral to spinal axon. Including slow outward conductances in the axonal compartments near the T-junction reduced following frequencies to ranges measured experimentally. We next used the population of C-neuron models to investigate how DRGS affected the orthodromic propagation of APs through the T-junction. Our data suggest that suprathreshold DRGS augmented the filtering of APs at the T-junction of some model C-neurons while increasing the activity of other model C-neurons. However, the stimulus pulse amplitudes required to induce activity in C-neurons (i.e., several mA) fell outside the range of stimulation pulse amplitudes used clinically (i.e., typically ≤1 mA). Furthermore, our data suggest that somatic GABA currents activated directly or indirectly by the DRGS pulse may produce diverse effects on orthodromic AP propagation in C-neurons. These data suggest DRGS may produce differential effects across a population of C-neurons and indicate that understanding how inherent biological variability affects a neuron's response to therapeutic electrical stimulation may be helpful in understanding its mechanisms of action.
Collapse
Affiliation(s)
- Robert D Graham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Amolak S Jhand
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States.,Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
21
|
Vuka I, Marciuš T, Kovačić D, Šarolić A, Puljak L, Sapunar D. Implantable, Programmable, and Wireless Device for Electrical Stimulation of the Dorsal Root Ganglion in Freely-Moving Rats: A Proof of Concept Study. J Pain Res 2021; 14:3759-3772. [PMID: 34916842 PMCID: PMC8668248 DOI: 10.2147/jpr.s332438] [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: 08/03/2021] [Accepted: 11/23/2021] [Indexed: 11/23/2022] Open
Abstract
Objective This was a proof of concept study, based on systematic reviews of the efficacy and safety of the dorsal root ganglion (DRG) stimulation. The main objective was to develop an implantable, programmable, and wireless device for electrical stimulation of DRG and a methodology that can be used in translational research, especially to understand the mechanism of neuromodulation and to test new treatment modalities in animal models of pain. Methods We developed and tested a stimulator that uses a battery-powered microelectronic circuit, to generate constant current square biphasic or monophasic pulsed waveform of variable amplitudes and duration. It is controlled by software and an external controller that allows radio frequency communication with the stimulator. The stimulator was implanted in Sprague–Dawley (SD) rats. The lead was positioned at the L5 DRG level, while the stimulator was placed in the skin pocket at the ipsilateral side. Forty-five animals were used and divided into six groups: spinal nerve ligation (SNL), chronic compression injury of the DRG (CCD), SNL + active DRG stimulation, intact control group, group with the implanted sham stimulator, and sham lead. Behavioral testing was performed on the day preceding surgery and three times postoperatively (1st, 3rd, and 7th day). Results In animals with SNL, neurostimulation reduced pain-related behavior, tested with pinprick hyperalgesia, pinprick withdrawal test, and cold test, while the leads per se did not cause DRG compression. The rats well tolerated the stimulator. It did not hinder animal movement, and it enabled the animals to be housed under regular conditions. Conclusion A proof-of-concept experiment with our stimulator verified the usability of the device. The stimulator enables a wide range of research applications from adjusting stimulation parameters for different pain conditions, studying new stimulation methods with different frequencies and waveforms to obtain knowledge about analgesic mechanisms of DRG stimulation.
Collapse
Affiliation(s)
- Ivana Vuka
- Laboratory for Pain Research, University of Split School of Medicine, Split, Croatia
| | - Tihana Marciuš
- Laboratory for Pain Research, University of Split School of Medicine, Split, Croatia
| | - Damir Kovačić
- Laboratory for Biophysics and Medical Neuroelectronics, University of Split Faculty of Science, Split, Croatia
| | - Antonio Šarolić
- Laboratory for Applied Electromagnetics (EMLab), FESB, University of Split, Split, Croatia
| | - Livia Puljak
- Centre for Evidence-Based Medicine and Health Care, Catholic University of Croatia, Zagreb, Croatia
| | - Damir Sapunar
- Laboratory for Pain Research, University of Split School of Medicine, Split, Croatia
| |
Collapse
|
22
|
Chao D, Mecca CM, Yu G, Segel I, Gold MS, Hogan QH, Pan B. Dorsal root ganglion stimulation of injured sensory neurons in rats rapidly eliminates their spontaneous activity and relieves spontaneous pain. Pain 2021; 162:2917-2932. [PMID: 33990112 PMCID: PMC8486885 DOI: 10.1097/j.pain.0000000000002284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/23/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Dorsal root ganglion field stimulation (GFS) relieves evoked and spontaneous neuropathic pain by use-dependent blockade of impulse trains through the sensory neuron T-junction, which becomes complete within less than 1 minute for C-type units, also with partial blockade of Aδ units. We used this tool in the spinal nerve ligation (SNL) rat model to selectively block sensory neuron spontaneous activity (SA) of axotomized neurons at the fifth lumbar (L5) level vs blockade of units at the L4 level that remain uninjured but exposed to inflammation. In vivo dorsal root single-unit recordings after SNL showed increased SA in L5 units but not L4 units. Ganglion field stimulation blocked this SA. Ganglion field stimulation delivered at the L5 dorsal root ganglion blocked mechanical hyperalgesia behavior, mechanical allodynia, and ongoing spontaneous pain indicated by conditioned place preference, whereas GFS at L4 blocked evoked pain behavior but not spontaneous pain. In vivo single-unit recordings of spinal cord dorsal horn (DH) wide-dynamic-range neurons showed elevated SA after SNL, which was reduced by GFS at the L5 level but not by GFS at the L4 level. In addition, L5 GFS, but not L4 GFS, increased mechanical threshold of DH units during cutaneous mechanical stimulation, while L5 GFS exceeded L4 GFS in reducing evoked firing rates. Our results indicate that SA in injured neurons supports increased firing of DH wide-dynamic-range neurons, contributing to hyperalgesia, allodynia, and ongoing pain. Ganglion field stimulation analgesic effects after nerve injury are at least partly attributable to blocking propagation of this SA.
Collapse
Affiliation(s)
- Dongman Chao
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Christina M. Mecca
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Guoliang Yu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Ian Segel
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Michael S. Gold
- Department of Neurobiology, University of Pittsburgh, 3500 Terrace Street Rm E1440 BST, Pittsburgh, PA 15213
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| |
Collapse
|
23
|
Yu G, Segel I, Tran H, Park HJ, Ross E, Hogan QH, Pan B. Analgesic Effects of Tonic and Burst Dorsal Root Ganglion Stimulation in Rats With Painful Tibial Nerve Injury. Neuromodulation 2021; 25:970-979. [PMID: 34096146 DOI: 10.1111/ner.13472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/06/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Dorsal root ganglion (DRG) stimulation is effective in treating chronic pain. While burst stimulation has been proven to enhance the therapeutic efficacy in spinal cord stimulation, currently only a tonic stimulation waveform is clinically used in DRG stimulation. We hypothesized that burst DRG stimulation might also produce analgesic effect in a preclinical neuropathic pain model. We evaluated both the therapeutic effects of burst DRG stimulation and the possible effects of DRG stimulation upon inflammation within the DRG in a preclinical neuropathic pain model. MATERIALS AND METHODS Rats received either a painful tibial nerve injury or sham surgery. Analgesic effects of DRG stimulation were evaluated by testing a battery of evoked pain-related behaviors as well as measuring the positive affective state associated with relief of spontaneous pain using conditioned place preference. Histological evidence for neuronal trauma or neuroinflammation was evaluated. RESULTS All of the waveforms tested (20 Hz-tonic, 20 Hz-burst, and 40 Hz-burst) have similar analgesic effects in sensory tests and conditioned place preference. Long-term DRG stimulation for two weeks does not change DRG expression of markers for nerve injury and neuroinflammation. CONCLUSIONS DRG stimulation using burst waveform might be also suitable for treating neuropathic pain.
Collapse
Affiliation(s)
- Guoliang Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ian Segel
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hai Tran
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Erika Ross
- Abbott Neuromodulation, Plano, TX, 75024, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| |
Collapse
|
24
|
Knotkova H, Hamani C, Sivanesan E, Le Beuffe MFE, Moon JY, Cohen SP, Huntoon MA. Neuromodulation for chronic pain. Lancet 2021; 397:2111-2124. [PMID: 34062145 DOI: 10.1016/s0140-6736(21)00794-7] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 03/24/2021] [Accepted: 03/31/2021] [Indexed: 12/23/2022]
Abstract
Neuromodulation is an expanding area of pain medicine that incorporates an array of non-invasive, minimally invasive, and surgical electrical therapies. In this Series paper, we focus on spinal cord stimulation (SCS) therapies discussed within the framework of other invasive, minimally invasive, and non-invasive neuromodulation therapies. These therapies include deep brain and motor cortex stimulation, peripheral nerve stimulation, and the non-invasive treatments of repetitive transcranial magnetic stimulation, transcranial direct current stimulation, and transcutaneous electrical nerve stimulation. SCS methods with electrical variables that differ from traditional SCS have been approved. Although methods devoid of paraesthesias (eg, high frequency) should theoretically allow for placebo-controlled trials, few have been done. There is low-to-moderate quality evidence that SCS is superior to reoperation or conventional medical management for failed back surgery syndrome, and conflicting evidence as to the superiority of traditional SCS over sham stimulation or between different SCS modalities. Peripheral nerve stimulation technologies have also undergone rapid development and become less invasive, including many that are placed percutaneously. There is low-to-moderate quality evidence that peripheral nerve stimulation is effective for neuropathic pain in an extremity, low quality evidence that it is effective for back pain with or without leg pain, and conflicting evidence that it can prevent migraines. In the USA and many areas in Europe, deep brain and motor cortex stimulation are not approved for chronic pain, but are used off-label for refractory cases. Overall, there is mixed evidence supporting brain stimulation, with most sham-controlled trials yielding negative findings. Regarding non-invasive modalities, there is moderate quality evidence that repetitive transcranial magnetic stimulation does not provide meaningful benefit for chronic pain in general, but conflicting evidence regarding pain relief for neuropathic pain and headaches. For transcranial direct current stimulation, there is low-quality evidence supporting its benefit for chronic pain, but conflicting evidence regarding a small treatment effect for neuropathic pain and headaches. For transcutaneous electrical nerve stimulation, there is low-quality evidence that it is superior to sham or no treatment for neuropathic pain, but conflicting evidence for non-neuropathic pain. Future research should focus on better evaluating the short-term and long-term effectiveness of all neuromodulation modalities and whether they decrease health-care use, and on refining selection criteria and treatment variables.
Collapse
Affiliation(s)
- Helena Knotkova
- MJHS Institute for Innovation in Palliative Care, New York, NY, USA; Department of Family and Social Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Clement Hamani
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Harquail Centre for Neuromodulation, University of Toronto, Toronto, ON, Canada
| | - Eellan Sivanesan
- Department of Anesthesiology & Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Jee Youn Moon
- Department of Anesthesiology, Seoul National University, Seoul, South Korea
| | - Steven P Cohen
- Department of Neurology, Department of Physical Medicine & Rehabilitation, and Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Anesthesiology and Department of Physical Medicine & Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Marc A Huntoon
- Department of Anesthesiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
25
|
Analgesic dorsal root ganglionic field stimulation blocks conduction of afferent impulse trains selectively in nociceptive sensory afferents. Pain 2021; 161:2872-2886. [PMID: 32658148 DOI: 10.1097/j.pain.0000000000001982] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increased excitability of primary sensory neurons after peripheral nerve injury may cause hyperalgesia and allodynia. Dorsal root ganglion field stimulation (GFS) is effective in relieving clinical pain associated with nerve injury and neuropathic pain in animal models. However, its mechanism has not been determined. We examined effects of GFS on transmission of action potentials (APs) from the peripheral to central processes by in vivo single-unit recording from lumbar dorsal roots in sham injured rats and rats with tibial nerve injury (TNI) in fiber types defined by conduction velocity. Transmission of APs directly generated by GFS (20 Hz) in C-type units progressively abated over 20 seconds, whereas GFS-induced Aβ activity persisted unabated, while Aδ showed an intermediate pattern. Activity generated peripherally by electrical stimulation of the sciatic nerve and punctate mechanical stimulation of the receptive field (glabrous skin) was likewise fully blocked by GFS within 20 seconds in C-type units, whereas Aβ units were minimally affected and a subpopulation of Aδ units was blocked. After TNI, the threshold to induce AP firing by punctate mechanical stimulation (von Frey) was reduced, which was reversed to normal during GFS. These results also suggest that C-type fibers, not Aβ, mainly contribute to mechanical and thermal hypersensitivity (von Frey, brush, acetone) after injury. Ganglion field stimulation produces use-dependent blocking of afferent AP trains, consistent with enhanced filtering of APs at the sensory neuron T-junction, particularly in nociceptive units.
Collapse
|
26
|
Franken G, Douven P, Debets J, Joosten EAJ. Conventional Dorsal Root Ganglion Stimulation in an Experimental Model of Painful Diabetic Peripheral Neuropathy: A Quantitative Immunocytochemical Analysis of Intracellular γ-Aminobutyric Acid in Dorsal Root Ganglion Neurons. Neuromodulation 2021; 24:639-645. [PMID: 33942947 PMCID: PMC8360133 DOI: 10.1111/ner.13398] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 11/26/2022]
Abstract
Background and Objective The sensory cell somata in the DRG contain all equipment necessary for extensive GABAergic signaling and are able to release GABA upon depolarization. With this study, we hypothesize that pain relief induced by conventional dorsal root ganglion stimulation (Con‐DRGS) in animals with experimental painful diabetic peripheral neuropathy is related to the release of GABA from DRG neurons. With use of quantitative immunocytochemistry, we hypothesize DRGS to result in a decreased intensity of intracellular GABA‐immunostaining in DRG somata. Materials and Methods Female Sprague‐Dawley rats (n = 31) were injected with streptozotocin (STZ) in order to induce Diabetes Mellitus. Animals that developed neuropathic pain after four weeks (Von Frey) were implanted with a unilateral DRGS device at L4 (n = 14). Animals were then stimulated for 30 min with Con‐DRGS (20 Hz, pulse width = 0.2 msec, amplitude = 67% of motor threshold, n = 8) or Sham‐DRGS (n = 6), while pain behavior (von Frey) was measured. DRGs were then collected and immunostained for GABA, and a relation to size of sensory cell soma diameter (small: 12–26 μm, assumed to be C‐fiber related sensory neurons; medium: 26–40 μm, assumed to be Aδ related sensory neurons; and large: 40–54 μm, assumed to be Aβ related sensory neurons) was made. Results DRGS treated animals showed significant reductions in STZ‐induced mechanical hypersensitivity. No significant differences in GABA immunostaining intensity per sensory neuron cell soma type (small‐, medium‐, or large‐sized) were noted in DRGs of stimulated (Con‐DRGS) animals versus Sham animals. No differences in GABA immunostaining intensity per sensory cell soma type in ipsi‐ as compared to contralateral DRGs were observed. Conclusion Con‐DRGS does not affect the average intracellular GABA immunofluorescence staining intensity in DRG sensory neurons of those animals which showed significant pain reduction. Similarly, no soma size related changes in intracellular GABA immunofluorescence were observed following Con‐DRGS.
Collapse
Affiliation(s)
- Glenn Franken
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Perla Douven
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Urology, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,Department of Surgery, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| | - Jacques Debets
- School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Muroidean Facility, School of Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.,School for Mental Health and Neuroscience (MHeNS), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
27
|
Kuwabara Y, Salavatian S, Howard-Quijano K, Yamaguchi T, Lundquist E, Mahajan A. Neuromodulation With Thoracic Dorsal Root Ganglion Stimulation Reduces Ventricular Arrhythmogenicity. Front Physiol 2021; 12:713717. [PMID: 34690795 PMCID: PMC8528951 DOI: 10.3389/fphys.2021.713717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Sympathetic hyperactivity is strongly associated with ventricular arrhythmias and sudden cardiac death. Neuromodulation provides therapeutic options for ventricular arrhythmias by modulating cardiospinal reflexes and reducing sympathetic output at the level of the spinal cord. Dorsal root ganglion stimulation (DRGS) is a recent neuromodulatory approach; however, its role in reducing ventricular arrhythmias has not been evaluated. The aim of this study was to determine if DRGS can reduce cardiac sympathoexcitation and the indices for ventricular arrhythmogenicity induced by programmed ventricular extrastimulation. We evaluated the efficacy of thoracic DRGS at both low (20 Hz) and high (1 kHz) stimulation frequencies. Methods: Cardiac sympathoexcitation was induced in Yorkshire pigs (n = 8) with ventricular extrastimulation (S1/S2 pacing), before and after DRGS. A DRG-stimulating catheter was placed at the left T2 spinal level, and animals were randomized to receive low-frequency (20 Hz and 0.4 ms) or high-frequency (1 kHz and 0.03 ms) DRGS for 30 min. High-fidelity cardiac electrophysiological recordings were performed with an epicardial electrode array measuring the indices of ventricular arrhythmogenicity-activation recovery intervals (ARIs), electrical restitution curve (Smax), and Tpeak-Tend interval (Tp-Te interval). Results: Dorsal root ganglion stimulation, at both 20 Hz and 1 kHz, decreased S1/S2 pacing-induced ARI shortening (20 Hz DRGS -21±7 ms, Control -50±9 ms, P = 0.007; 1 kHz DRGS -13 ± 2 ms, Control -46 ± 8 ms, P = 0.001). DRGS also reduced arrhythmogenicity as measured by a decrease in Smax (20 Hz DRGS 0.5 ± 0.07, Control 0.7 ± 0.04, P = 0.006; 1 kHz DRGS 0.5 ± 0.04, Control 0.7 ± 0.03, P = 0.007), and a decrease in Tp-Te interval/QTc (20 Hz DRGS 2.7 ± 0.13, Control 3.3 ± 0.12, P = 0.001; 1 kHz DRGS 2.8 ± 0.08, Control; 3.1 ± 0.03, P = 0.007). Conclusions: In a porcine model, we show that thoracic DRGS decreased cardiac sympathoexcitation and indices associated with ventricular arrhythmogenicity during programmed ventricular extrastimulation. In addition, we demonstrate that both low-frequency and high-frequency DRGS can be effective neuromodulatory approaches for reducing cardiac excitability during sympathetic hyperactivity.
Collapse
Affiliation(s)
- Yuki Kuwabara
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Siamak Salavatian
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kimberly Howard-Quijano
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Tomoki Yamaguchi
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Eevanna Lundquist
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Aman Mahajan
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- *Correspondence: Aman Mahajan
| |
Collapse
|
28
|
Sivanesan E, Cohen SP. Neuromodulation for Pain Treatment: Building a Foundation for Future Study. Anesthesiology 2020; 133:262-264. [PMID: 32568851 DOI: 10.1097/aln.0000000000003384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Eellan Sivanesan
- From the Department of Anesthesiology and Critical Care Medicine (E.S., S.P.C.) the Departments of Neurology and Physical Medicine and Rehabilitation (S.P.C.), Johns Hopkins University School of Medicine, Baltimore, Maryland the Departments of Anesthesiology and Physical Medicine and Rehabilitation, Walter Reed National Military Medical Center, Uniformed Services University of the Health Sciences, Bethesda, Maryland (S.P.C.)
| | | |
Collapse
|