1
|
Cocea AC, Stoica CI. Interactions and Trends of Interleukins, PAI-1, CRP, and TNF-α in Inflammatory Responses during the Perioperative Period of Joint Arthroplasty: Implications for Pain Management-A Narrative Review. J Pers Med 2024; 14:537. [PMID: 38793119 PMCID: PMC11122505 DOI: 10.3390/jpm14050537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Inflammation during the perioperative period of joint arthroplasty is a critical aspect of patient outcomes, influencing both the pathophysiology of pain and the healing process. This narrative review comprehensively evaluates the roles of specific cytokines and inflammatory biomarkers in this context and their implications for pain management. Inflammatory responses are initiated and propagated by cytokines, which are pivotal in the development of both acute and chronic postoperative pain. Pro-inflammatory cytokines play essential roles in up-regulating the inflammatory response, which, if not adequately controlled, leads to sustained pain and impaired tissue healing. Anti-inflammatory cytokines work to dampen inflammatory responses and promote resolution. Our discussion extends to the genetic and molecular influences on cytokine production, which influence pain perception and recovery rates post-surgery. Furthermore, the role of PAI-1 in modulating inflammation through its impact on the fibrinolytic system highlights its potential as a therapeutic target. The perioperative modulation of these cytokines through various analgesic and anesthetic techniques, including the fascia iliac compartment block, demonstrates a significant reduction in pain and inflammatory markers, thus underscoring the importance of targeted therapeutic strategies. Our analysis suggests that a nuanced understanding of the interplay between pro-inflammatory and anti-inflammatory cytokines is required. Future research should focus on individualized pain management strategies.
Collapse
Affiliation(s)
- Arabela-Codruta Cocea
- Faculty of Medicine, Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Ioan Stoica
- Orthopedics, Anaesthesia Intensive Care Unit, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
2
|
Lesnak JB, Berardi G, Sluka KA. Influence of routine exercise on the peripheral immune system to prevent and alleviate pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100126. [PMID: 37179769 PMCID: PMC10173010 DOI: 10.1016/j.ynpai.2023.100126] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 05/15/2023]
Abstract
Routine physical activity reduces the onset of pain and exercise is a first line treatment for individuals who develop chronic pain. In both preclinical and clinical research regular exercise (routine exercise sessions) produces pain relief through multiple mechanisms such as alterations in the central and peripheral nervous system. More recently, it has been appreciated that exercise can also alter the peripheral immune system to prevent or reduce pain. In animal models, exercise can alter the immune system at the site of injury or pain model induction, in the dorsal root ganglia, and systemically throughout the body to produce analgesia. Most notably exercise shows the ability to dampen the presence of pro-inflammatory immune cells and cytokines at these locations. Exercise decreases M1 macrophages and the cytokines IL-6, IL-1β, and TFNα, while increasing M2 macrophages and the cytokines IL-10, IL-4, and IL-1ra. In clinical research, a single bout of exercise produces an acute inflammatory response, however repeated training can lead to an anti-inflammatory immune profile leading to symptom relief. Despite the clinical and immune benefits of routine exercise, the direct effect of exercise on immune function in clinical pain populations remains unexplored. This review will discuss in more detail the preclinical and clinical research which demonstrates the numerous ways through which multiple types of exercise alter the peripheral immune system. This review closes with the clinical implications of these findings along with suggestions for future research directions.
Collapse
Affiliation(s)
- Joseph B. Lesnak
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Giovanni Berardi
- Department of Physical Therapy & Rehabilitation Sciences, University of Iowa, Iowa City, IA, USA
| | - Kathleen A. Sluka
- Department of Physical Therapy & Rehabilitation Sciences, University of Iowa, Iowa City, IA, USA
- Corresponding author.
| |
Collapse
|
3
|
Green-Fulgham SM, Harland ME, Ball JB, Li J, Lacagnina MJ, D’Angelo H, Dreher RA, Willcox KF, Lorca SA, Kwilasz AJ, Maier SF, Watkins LR, Grace PM. Preconditioning by voluntary wheel running attenuates later neuropathic pain via nuclear factor E2-related factor 2 antioxidant signaling in rats. Pain 2022; 163:1939-1951. [PMID: 35486864 PMCID: PMC9308835 DOI: 10.1097/j.pain.0000000000002589] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/19/2022] [Indexed: 02/04/2023]
Abstract
ABSTRACT Animal and human studies have shown that exercise prior to nerve injury prevents later chronic pain, but the mechanisms of such preconditioning remain elusive. Given that exercise acutely increases the formation of free radicals, triggering antioxidant compensation, we hypothesized that voluntary running preconditioning would attenuate neuropathic pain by supporting redox homeostasis after sciatic nerve injury in male and female rats. We show that 6 weeks of voluntary wheel running suppresses neuropathic pain development induced by chronic constriction injury across both sexes. This attenuation was associated with reduced nitrotyrosine immunoreactivity-a marker for peroxynitrite-at the sciatic nerve injury site. Our data suggest that prior voluntary wheel running does not reduce the production of peroxynitrite precursors, as expression levels of inducible nitric oxide synthase and NADPH oxidase 2 were unchanged. Instead, voluntary wheel running increased superoxide scavenging by elevating expression of superoxide dismutases 1 and 2. Prevention of neuropathic pain was further associated with the activation of the master transcriptional regulator of the antioxidant response, nuclear factor E2-related factor 2 (Nrf2). Six weeks of prior voluntary wheel running increased Nrf2 nuclear translocation at the sciatic nerve injury site; in contrast, 3 weeks of prior wheel running, which failed to prevent neuropathic pain, had no effect on Nrf2 nuclear translocation. The protective effects of prior voluntary wheel running were mediated by Nrf2, as suppression was abolished across both sexes when Nrf2 activation was blocked during the 6-week running phase. This study provides insight into the mechanisms by which physical activity may prevent neuropathic pain. Preconditioning by voluntary wheel running, terminated prior to nerve injury, suppresses later neuropathic pain in both sexes, and it is modulated through the activation of Nrf2-antioxidant signaling.
Collapse
Affiliation(s)
- Suzanne M. Green-Fulgham
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Michael E. Harland
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Jayson B. Ball
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Jiahe Li
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| | - Michael J. Lacagnina
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| | - Heather D’Angelo
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Renee A. Dreher
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Kendal F. Willcox
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| | - Sabina A. Lorca
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew J. Kwilasz
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Steven F. Maier
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Peter M. Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| |
Collapse
|
4
|
Liu XG. Normalization of Neuroinflammation: A New Strategy for Treatment of Persistent Pain and Memory/Emotional Deficits in Chronic Pain. J Inflamm Res 2022; 15:5201-5233. [PMID: 36110505 PMCID: PMC9469940 DOI: 10.2147/jir.s379093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic pain, which affects around 1/3 of the world population and is often comorbid with memory deficit and mood depression, is a leading source of suffering and disability. Studies in past decades have shown that hyperexcitability of primary sensory neurons resulting from abnormal expression of ion channels and central sensitization mediated pathological synaptic plasticity, such as long-term potentiation in spinal dorsal horn, underlie the persistent pain. The memory/emotional deficits are associated with impaired synaptic connectivity in hippocampus. Dysregulation of numerous endogenous proteins including receptors and intracellular signaling molecules is involved in the pathological processes. However, increasing knowledge contributes little to clinical treatment. Emerging evidence has demonstrated that the neuroinflammation, characterized by overproduction of pro-inflammatory cytokines and glial activation, is reliably detected in humans and animals with chronic pain, and is sufficient to induce persistent pain and memory/emotional deficits. The abnormal expression of ion channels and pathological synaptic plasticity in spinal dorsal horn and in hippocampus are resulting from neuroinflammation. The neuroinflammation is initiated and maintained by the interactions of circulating monocytes, glial cells and neurons. Obviously, unlike infectious diseases and cancer, which are caused by pathogens or malignant cells, chronic pain is resulting from alterations of cells and molecules which have numerous physiological functions. Therefore, normalization (counterbalance) but not simple inhibition of the neuroinflammation is the right strategy for treating neuronal disorders. Currently, no such agent is available in clinic. While experimental studies have demonstrated that intracellular Mg2+ deficiency is a common feature of chronic pain in animal models and supplement Mg2+ are capable of normalizing the neuroinflammation, activation of upregulated proteins that promote recovery, such as translocator protein (18k Da) or liver X receptors, has a similar effect. In this article, relevant experimental and clinical evidence is reviewed and discussed.
Collapse
Affiliation(s)
- Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
5
|
Min C, Ling R, Chen M, Xia D, Chen R, Li X. Enriched environment rescues neonatal pain induced cognitive deficits and the impaired hippocampal synaptic plasticity later in life. Dev Neurobiol 2022; 82:545-561. [PMID: 35945168 DOI: 10.1002/dneu.22898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/04/2022] [Accepted: 07/27/2022] [Indexed: 11/06/2022]
Abstract
Although extensive and untreated pain that occurs during a critical developmental window may impair cognition later in life, environmental interventions at early might promote. However, the underlying mechanism is poorly understood. Our current study utilized a rat model of "repetitive needle pricks" from the day of birth (P0) to postnatal day 7 (P7) to mimic the painful experience of preterm neonates in the Neonatal intensive care unit (NICU). Enriched environment (EE) during development period (from P15 to P70) was implemented as a nonpharmacological intervention approach. Electrophysiological recording, behavioral tests and biochemical analysis were performed after the end of EE (between P71 and P80). Results showed neonatal repetitive pain resulted in a reduction in mechanical withdrawal thresholds by the von Frey test in P70 (P < 0.001). Furthermore, neonatal repetitive pain impaired spatial learning and memory (P < 0.05) and even led to dysfunction in fear memory (P < 0.01). In contrast, EE rescued neonatal pain induced cognitive deficits and normalized hippocampal long-term potentiation in rats exposed to neonatal pain (P < 0.05). The beneficial effect of EE might be the improvements in hippocampal synaptic plasticity via up-regulating neurotrophic factors and N-methyl-D-aspartate (NMDA) receptors in the hippocampus. Our findings provide evidence that early environmental intervention might be a safe strategy to overcome neurodevelopmental abnormalities in preterm infants who experienced multiple procedural painful events during the early critical period. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Cuiting Min
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing, Jiangsu Province, 210008, P.R. China
| | - Ru Ling
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing, Jiangsu Province, 210008, P.R. China
| | - Mengying Chen
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing, Jiangsu Province, 210008, P.R. China
| | - Dongqing Xia
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing, Jiangsu Province, 210008, P.R. China
| | - Ran Chen
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing, Jiangsu Province, 210008, P.R. China
| | - Xiaonan Li
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, No.72 Guangzhou Road, Nanjing, Jiangsu Province, 210008, P.R. China
| |
Collapse
|
6
|
Disruption of Hyaluronic Acid in Skeletal Muscle Induces Decreased Voluntary Activity via Chemosensitive Muscle Afferent Sensitization in Male Mice. eNeuro 2022; 9:ENEURO.0522-21.2022. [PMID: 35387844 PMCID: PMC9014980 DOI: 10.1523/eneuro.0522-21.2022] [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: 11/16/2021] [Revised: 02/25/2022] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
PEGPH20, a human recombinant hyaluronidase, has been proposed as a coadjutant to pancreatic cancer chemotherapy. In early trials, patients reported increased widespread muscle pain as the main adverse reaction to PEGPH20. To understand how PEGPH20 caused musculoskeletal pain, we systemically administered PEGPH20 to male mice and measured voluntary wheel activity and pain-related behaviors. These were paired with ex vivo electrophysiology of primary sensory neurons, whole DRG real-time PCR, and immunohistochemistry of hindpaw muscle. PEGPH20 induced significantly lower wheel running, compared with vehicle-treated animals, and decreased mechanical withdrawal thresholds 5 d after PEGPH20 injections. Chemo-sensory muscle afferents showed increased responses to noxious chemical stimulation of their receptive fields (RFs) in the PEGPH20-treated group. This was correlated with upregulation of the NGF receptor TrkA, the transient receptor potential vanilloid type 1 (TRPV1) channel and ATP-sensitive channel P2X3 in the DRG. Immunohistochemistry of hindpaw muscles revealed damage to the muscle architecture and extensive infiltration of the tissue by cells of the myelomonocytic lineage 3 d after PEGPH20 injection. Peripheral macrophage ablation in macrophage Fas-induced apoptosis (MaFIA) mice, however, did not prevent the decreased voluntary activity and instead caused even lower levels of running. These results suggest that disruption of hyaluronic acid (HA) within the muscle extracellular matrix (ECM) sensitizes chemo-nociceptive muscle afferents possibly leading to altered pain-like behaviors. Ablation experiments suggest macrophages are necessary for adequate recovery of voluntary activity after HA disruption. These data support a role for HA and macrophages in tissue integrity and muscle pain development in patients taking PEGPH20.
Collapse
|
7
|
Periodontal acidification contributes to tooth pain hypersensitivity during orthodontic tooth movement. Neurosci Res 2021; 177:103-110. [PMID: 34808249 DOI: 10.1016/j.neures.2021.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022]
Abstract
Tooth movements associated with orthodontic treatment often cause tooth pain. However, the detailed mechanism remains unclear. Here, we examined the involvement of periodontal acidification caused by tooth movement in mechanical tooth pain hypersensitivity. Elastics were inserted between the first and second molars to move the teeth in Sprague-Dawley rats. Mechanical head-withdrawal reflex threshold to first molar stimulation and the pH of the gingival sulcus around the tooth were measured. The expression of acid-sensing ion channel 3 (ASIC3) in trigeminal ganglion neurons and phosphorylation of ASIC3 in the periodontal tissue were analyzed. The mechanical head-withdrawal reflex threshold to first molar stimulation and pH in the gingival sulcus decreased on day 1 after the elastic insertion. These decreases recovered to the sham level by buffering periodontal acidification. Periodontal inhibition of ASIC3 channel activity reversed the decreased mechanical head-withdrawal reflex threshold to first molar stimulation. On day 1 after elastic insertion, the tooth movement did not change the number of ASIC3 immunoreactive trigeminal ganglion neurons innervating the periodontal tissue but increased phosphorylated-ASIC3 levels in the periodontal tissue. Periodontal acidification induced by tooth movement causes phosphorylation of ASIC3, resulting in mechanical pain hypersensitivity in mechanically forced tooth.
Collapse
|
8
|
de Azambuja G, Jorge CO, Gomes BB, Lourenço HR, Simabuco FM, Oliveira-Fusaro MCG. Regular swimming exercise prevented the acute and persistent mechanical muscle hyperalgesia by modulation of macrophages phenotypes and inflammatory cytokines via PPARγ receptors. Brain Behav Immun 2021; 95:462-476. [PMID: 33964434 DOI: 10.1016/j.bbi.2021.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/09/2021] [Accepted: 05/03/2021] [Indexed: 12/31/2022] Open
Abstract
Physically active individuals are less likely to develop chronic pain, and physical exercise is an established strategy to control inflammatory diseases. Here, we hypothesized that 1) peripheral pro-inflammatory macrophages phenotype contribute to predisposition of the musculoskeletal to chronic pain, and that 2) activation of PPARγ receptors, modulation of macrophage phenotypes and cytokines through physical exercise would prevent persistent muscle pain. We tested these hypotheses using swimming exercise, pharmacological and immunochemical techniques in a rodent model of persistent muscle hyperalgesia. Swimming prevented the persistent mechanical muscle hyperalgesia most likely through activation of PPARγ receptors, as well as activation of PPARγ receptors by 15d-PGJ2 and depletion of muscle macrophages in sedentary animals. Acute and persistent muscle hyperalgesia were characterized by an increase in pro-inflammatory macrophages phenotype, and swimming and the 15d-PGJ2 prevented this increase and increased anti-inflammatory macrophages phenotype. Finally, IL-1β concentration in muscle increased in the acute phase, which was also prevented by PPARγ receptors activation through swimming. Besides, swimming increased muscle concentration of IL-10 in both acute and chronic phases, but only in the persistent phase through PPARγ receptors. Our findings suggest physical exercise activates PPARγ receptors and increases anti-inflammatory responses in the muscle tissue by modulating macrophages phenotypes and cytokines, thereby preventing the establishment of persistent muscle hyperalgesia. These results further highlight the potential of physical exercise to prevent chronic muscle pain.
Collapse
Affiliation(s)
- Graciana de Azambuja
- Laboratory of Pain and Inflammation Research, School of Applied Sciences, University of Campinas, Brazil
| | - Carolina O Jorge
- Laboratory of Pain and Inflammation Research, School of Applied Sciences, University of Campinas, Brazil
| | - Beatriz B Gomes
- Laboratory of Pain and Inflammation Research, School of Applied Sciences, University of Campinas, Brazil
| | - Hayla R Lourenço
- Laboratory of Pain and Inflammation Research, School of Applied Sciences, University of Campinas, Brazil
| | - Fernando M Simabuco
- Multidisciplinary Laboratory in Food and Health, School of Applied Sciences, University of Campinas, Brazil
| | | |
Collapse
|
9
|
Qin L, Li J. Nerve growth factor in muscle afferent neurons of peripheral artery disease and autonomic function. Neural Regen Res 2021; 16:694-699. [PMID: 33063730 PMCID: PMC8067946 DOI: 10.4103/1673-5374.293132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In peripheral artery disease patients, the blood supply directed to the lower limbs is reduced. This results in severe limb ischemia and thereby enhances pain sensitivity in lower limbs. The painful perception is induced and exaggerate during walking, and is relieved by rest. This symptom is termed by intermittent claudication. The limb ischemia also amplifies autonomic responses during exercise. In the process of pain and autonomic responses originating exercising muscle, a number of receptors in afferent nerves sense ischemic changes and send signals to the central nervous system leading to autonomic responses. This review integrates recent study results in terms of perspectives including how nerve growth factor affects muscle sensory nerve receptors in peripheral artery disease and thereby alters responses of sympathetic nerve activity and blood pressure to active muscle. For the sensory nerve receptors, we emphasize the role played by transient receptor potential vanilloid type 1, purinergic P2X purinoceptor 3 and acid sensing ion channel subtype 3 in amplified sympathetic nerve activity responses in peripheral artery disease.
Collapse
Affiliation(s)
- Lu Qin
- Heart & Vascular Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Jianhua Li
- Heart & Vascular Institute, Penn State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
10
|
Lesnak JB, Sluka KA. Mechanism of exercise-induced analgesia: what we can learn from physically active animals. Pain Rep 2020; 5:e850. [PMID: 33490844 PMCID: PMC7808683 DOI: 10.1097/pr9.0000000000000850] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/26/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022] Open
Abstract
Physical activity has become a first-line treatment in rehabilitation settings for individuals with chronic pain. However, research has only recently begun to elucidate the mechanisms of exercise-induced analgesia. Through the study of animal models, exercise has been shown to induce changes in the brain, spinal cord, immune system, and at the site of injury to prevent and reduce pain. Animal models have also explored beneficial effects of exercise through different modes of exercise including running, swimming, and resistance training. This review will discuss the central and peripheral mechanisms of exercise-induced analgesia through different modes, intensity, and duration of exercise as well as clinical applications of exercise with suggestions for future research directions.
Collapse
Affiliation(s)
- Joseph B. Lesnak
- Department of Physical Therapy and Rehabilitation Sciences, University of Iowa, Iowa City, IA, USA
| | - Kathleen A. Sluka
- Department of Physical Therapy and Rehabilitation Sciences, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
11
|
Khataei T, Harding AMS, Janahmadi M, El-Geneidy M, Agha-Alinejad H, Rajabi H, Snyder PM, Sluka KA, Benson CJ. ASICs are required for immediate exercise-induced muscle pain and are downregulated in sensory neurons by exercise training. J Appl Physiol (1985) 2020; 129:17-26. [PMID: 32463731 DOI: 10.1152/japplphysiol.00033.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exercise training is an effective therapy for many pain-related conditions, and trained athletes have lower pain perception compared with unconditioned people. Some painful conditions, including strenuous exercise, are associated with elevated levels of protons, metabolites, and inflammatory factors, which may activate receptors and/or ion channels, including acid-sensing ion channels (ASICs), on nociceptive sensory neurons. We hypothesized that ASICs are required for immediate exercise-induced muscle pain (IEIP) and that exercise training diminishes IEIP by modulating ASICs within muscle afferents. We found high-intensity interval training (HIIT) reduced IEIP in C57BL/6 mice and diminished ASIC mRNA levels in lumber dorsal root ganglia, and this downregulation of ASICs correlated with improved exercise capacity. Additionally, we found that ASIC3 -/- mice did not develop IEIP; however, the exercise capacity of ASIC3 -/- was similar to wild-type mice. These results suggest that ASICs are required for IEIP and that diminishment of IEIP after exercise training correlates with downregulation of ASICs in sensory neurons.NEW & NOTEWORTHY Exercise performance can be limited by the sensations of muscle fatigue and pain transmitted by muscle afferents. It has been proposed that exercise training abrogates these negative feedback signals. We found that acid-sensing ion channels (ASICs) are required for immediate exercise-induced muscle pain (IEIP). Moreover, exercise training prevented IEIP and was correlated with downregulation of ASICs in sensory neurons.
Collapse
Affiliation(s)
- Tahsin Khataei
- Department of Exercise Physiology, Tarbiat Modares University, Tehran, Iran.,Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | - Anne Marie S Harding
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | - Mahyar Janahmadi
- Department of Physiology and Neuroscience Research Center, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maram El-Geneidy
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | | | - Hamid Rajabi
- Department of Exercise Physiology, Kharazmi University, Tehran, Iran
| | - Peter M Snyder
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa
| | - Kathleen A Sluka
- Department of Physical Therapy and Rehabilitation Science, The University of Iowa, Iowa City, Iowa.,Neuroscience Institute, The University of Iowa, Iowa City, Iowa
| | - Christopher J Benson
- Department of Internal Medicine, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa.,Department of Pharmacology, University of Iowa, Roy J. and Lucile A. Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
12
|
A dual role for peripheral GDNF signaling in nociception and cardiovascular reflexes in the mouse. Proc Natl Acad Sci U S A 2019; 117:698-707. [PMID: 31848242 DOI: 10.1073/pnas.1910905116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Group III/IV muscle afferents transduce nociceptive signals and modulate exercise pressor reflexes (EPRs). However, the mechanisms governing afferent responsiveness to dually modulate these processes are not well characterized. We and others have shown that ischemic injury can induce both nociception-related behaviors and exacerbated EPRs in the same mice. This correlated with primary muscle afferent sensitization and increased expression of glial cell line-derived neurotrophic factor (GDNF) in injured muscle and increased expression of GDNF family receptor α1 (GFRα1) in dorsal root ganglia (DRG). Here, we report that increased GDNF/GFRα1 signaling to sensory neurons from ischemia/reperfusion-affected muscle directly modulated nociceptive-like behaviors and increased exercise-mediated reflexes and group III/IV muscle afferent sensitization. This appeared to have taken effect through increased cyclic adenosine monophosphate (cAMP) response element binding (CREB)/CREB binding protein-mediated expression of the purinergic receptor P2X5 in the DRGs. Muscle GDNF signaling to neurons may, therefore, play an important dual role in nociception and sympathetic reflexes and could provide a therapeutic target for treating complications from ischemic injuries.
Collapse
|
13
|
MiR-187-3p mimic alleviates ischemia-reperfusion-induced pain hypersensitivity through inhibiting spinal P2X7R and subsequent mature IL-1β release in mice. Brain Behav Immun 2019; 79:91-101. [PMID: 31100367 DOI: 10.1016/j.bbi.2019.05.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Ischemia-reperfusion (IR)-induced pain hypersensitivity shares features of neuroinflammation and neuropathic pain, accompanied by overproduction of interleukin (IL)-1β. Multiple microRNAs (miRs) are dysregulated during IR; among these miRs, miR-187-3p was recently reported to drive IL-1β release in retinal disease by activating members of the purinergic receptor family. However, the roles of miR-187-3p in the spinal cord are unclear. Thus, we investigated whether miR-187-3p is involved in the pathogenesis of IR-induced pain hypersensitivity by regulating the P2X7R signal and subsequent IL-1β release. METHODS A mouse model was established by 5-min occlusion of the aortic arch. Pain hypersensitivity was assessed by the paw withdrawal threshold (PWT) and paw withdrawal latency (PWL). MiR-187-3p, P2X7R, cleaved caspase-1 and mature IL-1β expression levels were measured by RT-PCR and Western blotting. The in vivo roles of miR-187-3p, P2X7R and IL-1β were explored by intrathecal treatment with synthetic miRs, selective agonists and antagonists in separate experiments. Double immunofluorescence staining was performed to delineate the cellular distribution of P2X7R and IL-1β. RESULTS IR-induced progressively decreased PWT and PWL values were closely related to decreases in miR-187-3p and increases in P2X7R expression levels over time. The functional miR-187-3p/P2X7R pair was preliminarily predicted by a bioinformatic database and confirmed in vivo by quantitative analysis, as mimic-187 greatly increased miR-187-3p but decreased P2X7R expression levels, whereas inhibitor-187 reversed these changes. In contrast, downregulating P2X7R by mimic-187 or A-438079 treatment comparably increased PWT and PWL values in IR-injured mice, while upregulating P2X7R by inhibitor-187 or BzATP treatment decreased PWT and PWL values in sham-operated mice. Moreover, P2X7R and IL-1β immunoreactivities in each group were changed in the same patterns. This finding was further supported by results showing that downregulating IL-1β by A-438079 and IL-1β-neutralizing antibody similarly decreased P2X7R, cleaved caspase-1 and mature IL-1β expression levels, whereas BzATP treatment increased these levels. Expectedly, mimic-187 treatment preserved PWT and PWL values, with decreased cleaved caspase-1 and mature IL-1β expression levels, whereas inhibitor-187 reversed these effects. CONCLUSIONS The spinal miR-187-3p/P2X7R pair functioned in a mouse IR model. Increasing miR-187-3p protected against pain hypersensitivity and mature IL-1β overproduction, partially through inhibiting P2X7R activation.
Collapse
|
14
|
|
15
|
Abstract
Clinical conditions resulting in musculoskeletal pain show important sex differences in both prevalence and degree of functional disability. The underlying mechanisms for these distinctions in pain manifestation are not fully known. However, recent preclinical studies have shown at the primary afferent level that males and females present fundamental differences in their peripheral response properties and injury-related gene expression patterns that may underlie observed afferent sensitization. At the spinal cord level, studies in various models of pain suggest important roles for the immune system, glutamate signaling and hormones in modulating sex differences. While preclinical studies have been able to characterize some of the basic underlying molecular mechanisms of sex differences in muscle pain, human studies have relied mainly on functional brain imaging studies to explain differences. Further complicating our understanding of how sex influences muscle pain is the notion that the type of injury sustained, or clinical condition may differentially activate distinct mechanisms of muscle pain development in males versus females. More research is necessary to better understand how the sexes differ in their perception of muscle pain. This review highlights recent advances in both human and animal studies of sex differences in muscle pain.
Collapse
|
16
|
Gong X, Chen Y, Chang J, Huang Y, Cai M, Zhang M. Environmental enrichment reduces adolescent anxiety- and depression-like behaviors of rats subjected to infant nerve injury. J Neuroinflammation 2018; 15:262. [PMID: 30208926 PMCID: PMC6134705 DOI: 10.1186/s12974-018-1301-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/31/2018] [Indexed: 01/12/2023] Open
Abstract
Background Infant nerve injury causes delayed adolescent neuropathic pain, but whether it also leads to psychiatric illness is unknown. Environmental enrichment (EE) increases social communication and activity. Thus, our goal was to test anxiety- and depression-like behaviors after infant peripheral nerve injury and evaluate the effect of environmental enrichment on these models of affective disorders. Methods Open field, elevated plus maze, sucrose preference, and pain behaviors (paw withdrawal threshold, spontaneous guarding score, and cold response to acetone) were measured in rats that received infant spared nerve injury (SNI). Enzyme-linked immune absorbent assay of cytokines was performed to evaluate the inflammatory response in the brain. Then, the ability of intracerebroventricular (ICV) injection of a microglia inhibitor, minocycline (MIN), and EE (a free-running wheel, a staircase, a plastic tunnel, a raised platform, and various colored balls) to reverse the infant SNI effects on behaviors and cytokines was examined. Results Infant nerve injury resulted in adolescent anxiety- and depression-like behaviors. The medial prefrontal cortex, basolateral amygdala, and ventral hippocampus were skewed to a pro-inflammatory profile. ICV injection of MIN reduced anxiety- and depression-like behaviors without affecting pain behaviors. In addition, ICV MIN skewed the brain towards an anti-inflammatory profile. Finally, environmental enrichment improved anxiety- and depression-like behaviors, as well as pain behaviors. EE increased brain IL-10 and decreased IL-1β and TNF-α. Conclusions Infant nerve injury induces adolescent anxiety- and depression-like behaviors and central nervous inflammation. Environmental enrichment reduces these behaviors by normalizing the inflammation balance in the brain.
Collapse
Affiliation(s)
- Xingrui Gong
- Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, 200127, Shanghai, China.,Department of Anesthesiology, Shiyan Taihe Hospital (Affiliated Hospital of Hubei University of Medicine), Shiyan, Hubei, China.,Institute of Anesthesiology, Department of Anesthesiology, Shiyan Taihe Hospital (Affiliated Hospital of Hubei University of Medicine), Shiyan, Hubei, China
| | - Yongmei Chen
- Department of Laboratory, Shiyan Taihe Hospital (Affiliated Hospital of Hubei University of Medicine), Shiyan, Hubei, China
| | - Jing Chang
- Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, 200127, Shanghai, China
| | - Yue Huang
- Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, 200127, Shanghai, China
| | - Meihau Cai
- Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, 200127, Shanghai, China
| | - Mazhong Zhang
- Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, 200127, Shanghai, China.
| |
Collapse
|
17
|
Yu J, Francisco AMC, Patel BG, Cline JM, Zou E, Berga SL, Taylor RN. IL-1β Stimulates Brain-Derived Neurotrophic Factor Production in Eutopic Endometriosis Stromal Cell Cultures: A Model for Cytokine Regulation of Neuroangiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2281-2292. [PMID: 30031725 DOI: 10.1016/j.ajpath.2018.06.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/15/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022]
Abstract
Endometriosis implants are comprised of glandular and stromal elements, macrophages, nerves, and blood vessels and are commonly accompanied by pelvic pain. We propose that activated macrophages are recruited to and infiltrate nascent lesions, where they secrete proinflammatory cytokines, promoting the production of chemokines, neurotrophins, and angiogenic growth factors that sustain an inflammatory microenvironment. Immunohistochemical evaluation of endometriosis lesions reveals in situ colocalization of concentrated macrophages, brain-derived neurotrophic factor (BDNF), and nerve fibers. These observations were coupled with biochemical analyses of primary eutopic endometriosis stromal cell (EESC) cultures, which allowed defining potential pathways leading to the neuroangiogenic phenotype of these lesions. Our findings indicate that IL-1β potently (EC50 = 7 ± 2 ng/mL) stimulates production of EESC BDNF at the mRNA and protein levels in an IL-1 receptor-dependent fashion. Selective kinase inhibitors demonstrate that this IL-1β effect is mediated by c-Jun N-terminal kinase (JNK), NF-κB, and mechanistic target of rapamycin signal transduction pathways. IL-1β regulation of regulated on activation normal T cell expressed and secreted (RANTES), a prominent EESC chemokine, also relies on JNK and NF-κB. An important clinical implication of the study is that interference with BDNF and RANTES production, by selectively targeting the JNK and NF-κB cascades, may offer a tractable therapeutic strategy to mitigate the pain and inflammation associated with endometriosis.
Collapse
Affiliation(s)
- Jie Yu
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Antônio M C Francisco
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Health Sciences School, University of Vale do Sapucaí, Pouso Alegre, Brazil
| | - Bansari G Patel
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - J Mark Cline
- Department of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; Molecular Medicine and Translational Sciences Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Eric Zou
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sarah L Berga
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Molecular Medicine and Translational Sciences Program, Wake Forest School of Medicine, Winston-Salem, North Carolina; Clinical and Translational Science Institute, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
18
|
Tai LW, Yeung SC, Cheung CW. Enriched Environment and Effects on Neuropathic Pain: Experimental Findings and Mechanisms. Pain Pract 2018; 18:1068-1082. [PMID: 29722923 DOI: 10.1111/papr.12706] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/04/2018] [Accepted: 04/22/2018] [Indexed: 12/16/2022]
Abstract
Neuropathic pain inflicts tremendous biopsychosocial suffering for patients worldwide. However, safe and effective treatment of neuropathic pain is a prominent unmet clinical need. Environmental enrichment (EE) is an emerging cost-effective nonpharmacological approach to alleviate neuropathic pain and complement rehabilitation care. We present here a review of preclinical studies in ascertaining the efficacy of EE for neuropathic pain. Their proposed mechanisms, including the suppression of ascending nociceptive signaling to the brain, enhancement of the descending inhibitory system, and neuroprotection of the peripheral and central nervous systems, may collectively reduce pain perception and improve somatic and emotional functioning in neuropathic pain. The current evidence offers critical insights for future preclinical research and the translational application of EE in clinical pain management.
Collapse
Affiliation(s)
- Lydia Wai Tai
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, The University of Hong Kong, Hong Kong, Special Administrative Region, China.,Department of Anaesthesiology, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Sung Ching Yeung
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, The University of Hong Kong, Hong Kong, Special Administrative Region, China.,Department of Anaesthesiology, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Chi Wai Cheung
- Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, The University of Hong Kong, Hong Kong, Special Administrative Region, China.,Department of Anaesthesiology, The University of Hong Kong, Hong Kong, Special Administrative Region, China.,Research Centre of Heart, Brain, Hormone & Healthy Aging, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| |
Collapse
|
19
|
Beazley-Long N, Durrant AM, Swift MN, Donaldson LF. The physiological functions of central nervous system pericytes and a potential role in pain. F1000Res 2018; 7:341. [PMID: 29623199 PMCID: PMC5861511 DOI: 10.12688/f1000research.13548.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2018] [Indexed: 12/29/2022] Open
Abstract
Central nervous system (CNS) pericytes regulate critical functions of the neurovascular unit in health and disease. CNS pericytes are an attractive pharmacological target for their position within the neurovasculature and for their role in neuroinflammation. Whether the function of CNS pericytes also affects pain states and nociceptive mechanisms is currently not understood. Could it be that pericytes hold the key to pain associated with CNS blood vessel dysfunction? This article reviews recent findings on the important physiological functions of CNS pericytes and highlights how these neurovascular functions could be linked to pain states.
Collapse
Affiliation(s)
- Nicholas Beazley-Long
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Alexandra M Durrant
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Matthew N Swift
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| | - Lucy F Donaldson
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham, UK
| |
Collapse
|