1
|
Nasir A, Afridi M, Afridi OK, Khan MA, Khan A, Zhang J, Qian B. The persistent pain enigma: Molecular drivers behind acute-to-chronic transition. Neurosci Biobehav Rev 2025; 173:106162. [PMID: 40239909 DOI: 10.1016/j.neubiorev.2025.106162] [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: 02/26/2025] [Revised: 03/20/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025]
Abstract
The transition from acute to chronic pain is a complex and multifactorial process that presents significant challenges in both diagnosis and treatment. Key mechanisms of peripheral and central sensitization, neuroinflammation, and altered synaptic plasticity contribute to the amplification of pain signals and the persistence of pain. Glial cell activation, particularly microglia and astrocytes, is pivotal in developing chronic pain by releasing pro-inflammatory cytokines that enhance pain sensitivity. This review explores the molecular, cellular, and systemic mechanisms underlying the transition from acute to chronic pain, offering new insights into the molecular and neurobiological mechanisms involved, which are often underexplored in existing literature. It also addresses emerging therapeutic strategies beyond traditional pain management, offering valuable perspectives for future research and clinical applications.
Collapse
Affiliation(s)
- Abdul Nasir
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Maryam Afridi
- Department of Pharmacy, Qurtuba University, Peshawar, KP, Pakistan
| | | | | | - Amir Khan
- Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jun Zhang
- Department of Pain, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Bai Qian
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| |
Collapse
|
2
|
Hao S, Lin S, Tao W, Zhuo M. Cortical Potentiation in Chronic Neuropathic Pain and the Future Treatment. Pharmaceuticals (Basel) 2025; 18:363. [PMID: 40143140 PMCID: PMC11944705 DOI: 10.3390/ph18030363] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/09/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Pain, or the ability to feel pain and express the unpleasantness caused by peripheral injuries, are functions of the central nervous system. From peripheral sensory nerve terminals to certain cortical regions of the brain, activation of related neural networks underlies the sensory process. Recently, our knowledge of pain has been increasing dramatically, due to the advancement of scientific approaches. We no longer see the brain as a random matrix for pain but, rather, we are able to identify the step-by-step selective signaling proteins, neurons, and networks that preferentially contribute to the process of chronic pain and its related negative emotions, like anxiety and fear. However, there is still lacking the selective and effective drugs and methods for the treatment of chronic pain clinically. While first-line drugs for acute pain and mental diseases are also applied for the clinical management of chronic pain, their prolonged usage always causes serious side effects. In this short review, we will update and summarize the recent progress in this field and mainly focus on the roles of neural networks and synaptic mechanisms in chronic neuropathic pain. Furthermore, potential drug targets (such as plasticity-related signaling molecules, ionic channels, cytokines, and neuropeptides) and methods for the management of chronic neuropathic pain will be discussed as well. We hope this review can provide new, valuable insight into the treatment of chronic neuropathic pain.
Collapse
Affiliation(s)
- Shun Hao
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou 350122, China; (S.H.); (W.T.)
| | - Shen Lin
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China;
| | - Wucheng Tao
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou 350122, China; (S.H.); (W.T.)
| | - Min Zhuo
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou 350122, China; (S.H.); (W.T.)
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
3
|
Morgan M, Nazemian V, Thai J, Lin I, Northfield S, Ivanusic JJ. BDNF sensitizes bone and joint afferent neurons at different stages of MIA-induced osteoarthritis. Bone 2024; 189:117260. [PMID: 39299629 DOI: 10.1016/j.bone.2024.117260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
There is emerging evidence that Brain Derived Neurotrophic Factor (BDNF), and one of its receptors TrkB, play important roles in the pathogenesis of osteoarthritis (OA) pain. Whilst these studies clearly highlight the potential for targeting BDNF/TrkB signaling to treat OA pain, the mechanism for how BDNF/TrkB signaling contributes to OA pain remains unclear. In this study, we used an animal model of mono-iodoacetate (MIA)-induced OA, in combination with electrophysiology, behavioral testing, Western blot analysis, and retrograde tracing and immunohistochemistry, to identify roles for BDNF/TrkB signaling in the pathogenesis of OA pain. We found that: 1) TrkB is expressed in myelinated medium diameter neurons that innervate the knee joint and bone in naïve animals; 2) peripheral application of BDNF increases the sensitivity of Aδ, but not C knee joint and bone afferent neurons, in response to mechanical stimulation, in naïve animals; 3) BDNF expression increases in synovial tissue in early MIA-induced OA, when pathology is confined to the joint, and in the subchondral bone in late MIA-induced OA, when there is additional damage to the surrounding bone; and 4) TrkB inhibition reverses MIA-induced changes in the sensitivity of Aδ but not C knee joint afferent neurons early in MIA-induced OA, and Aδ but not C bone afferent neurons late in MIA-induced OA. Our findings suggest that BDNF/TrkB signaling may have a role to play in the pathogenesis of OA pain, through effects on knee joint afferent neurons early in disease when there is inflammation confined to the joint, and bone afferent neurons late in disease when there is involvement of damage to subchondral bone. Targeted manipulation of BDNF/TrkB signaling may provide therapeutic benefit for the management of OA pain.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Vida Nazemian
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Irene Lin
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria, Australia
| | - Susan Northfield
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Hayashi K, Lesnak JB, Plumb AN, Janowski AJ, Smith AF, Hill JK, Sluka KA. Brain-derived neurotrophic factor contributes to activity-induced muscle pain in male but not female mice. Brain Behav Immun 2024; 120:471-487. [PMID: 38925417 DOI: 10.1016/j.bbi.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
Activity-induced muscle pain increases interleukin-1β (IL-1β) release from muscle macrophages and the development of hyperalgesia is prevented by blockade of IL-1β in muscle. Brain derived neurotrophic factor (BDNF) is released from sensory neurons in response to IL-1β and mediates both inflammatory and neuropathic pain. Thus, we hypothesize that in activity-induced pain, fatigue metabolites combined with IL-1β activate sensory neurons to increase BDNF release, peripherally in muscle and centrally in the spinal dorsal horn, to produce hyperalgesia. We tested the effect of intrathecal or intramuscular injection of BDNF-Tropomyosin receptor kinase B (TrkB) inhibitors, ANA-12 or TrkB-Fc, on development of activity-induced pain. Both inhibitors prevented the hyperalgesia when given before or 24hr after induction of the model in male but not female mice. BDNF messenger ribonucleic acid (mRNA) and protein were significantly increased in dorsal root ganglion (DRG) 24hr after induction of the model in both male and female mice. Blockade of IL-1β in muscle had no effect on the increased BNDF mRNA observed in the activity-induced pain model, while IL-1β applied to cultured DRG significantly induced BDNF expression, suggesting IL-1β is sufficient but not necessary to induce BNDF. Thus, fatigue metabolites, combined with IL-1β, upregulate BDNF in primary DRG neurons in both male and female mice, but contribute to activity-induced pain only in males.
Collapse
Affiliation(s)
- Kazuhiro Hayashi
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA; Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Joseph B Lesnak
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Ashley N Plumb
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Adam J Janowski
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Angela F Smith
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Joslyn K Hill
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Kathleen A Sluka
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
5
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
6
|
Singh AA, Yadav D, Khan F, Song M. Indole-3-Carbinol and Its Derivatives as Neuroprotective Modulators. Brain Sci 2024; 14:674. [PMID: 39061415 PMCID: PMC11274471 DOI: 10.3390/brainsci14070674] [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: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its downstream tropomyosin receptor kinase B (TrkB) signaling pathway play pivotal roles in the resilience and action of antidepressant drugs, making them prominent targets in psychiatric research. Oxidative stress (OS) contributes to various neurological disorders, including neurodegenerative diseases, stroke, and mental illnesses, and exacerbates the aging process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) serves as the primary cellular defense mechanism against OS-induced brain damage. Thus, Nrf2 activation may confer endogenous neuroprotection against OS-related cellular damage; notably, the TrkB/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, stimulated by BDNF-dependent TrkB signaling, activates Nrf2 and promotes its nuclear translocation. However, insufficient neurotrophin support often leads to the downregulation of the TrkB signaling pathway in brain diseases. Thus, targeting TrkB activation and the Nrf2-ARE system is a promising therapeutic strategy for treating neurodegenerative diseases. Phytochemicals, including indole-3-carbinol (I3C) and its metabolite, diindolylmethane (DIM), exhibit neuroprotective effects through BDNF's mimetic activity; Akt phosphorylation is induced, and the antioxidant defense mechanism is activated by blocking the Nrf2-kelch-like ECH-associated protein 1 (Keap1) complex. This review emphasizes the therapeutic potential of I3C and its derivatives for concurrently activating neuronal defense mechanisms in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea;
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| |
Collapse
|
7
|
Merighi A. Brain-Derived Neurotrophic Factor, Nociception, and Pain. Biomolecules 2024; 14:539. [PMID: 38785946 PMCID: PMC11118093 DOI: 10.3390/biom14050539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
This article examines the involvement of the brain-derived neurotrophic factor (BDNF) in the control of nociception and pain. BDNF, a neurotrophin known for its essential role in neuronal survival and plasticity, has garnered significant attention for its potential implications as a modulator of synaptic transmission. This comprehensive review aims to provide insights into the multifaceted interactions between BDNF and pain pathways, encompassing both physiological and pathological pain conditions. I delve into the molecular mechanisms underlying BDNF's involvement in pain processing and discuss potential therapeutic applications of BDNF and its mimetics in managing pain. Furthermore, I highlight recent advancements and challenges in translating BDNF-related research into clinical practice.
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, 10095 Turin, Italy
| |
Collapse
|
8
|
Smith PA. BDNF in Neuropathic Pain; the Culprit that Cannot be Apprehended. Neuroscience 2024; 543:49-64. [PMID: 38417539 DOI: 10.1016/j.neuroscience.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/20/2024] [Indexed: 03/01/2024]
Abstract
In males but not in females, brain derived neurotrophic factor (BDNF) plays an obligatory role in the onset and maintenance of neuropathic pain. Afferent terminals of injured peripheral nerves release colony stimulating factor (CSF-1) and other mediators into the dorsal horn. These transform the phenotype of dorsal horn microglia such that they express P2X4 purinoceptors. Activation of these receptors by neuron-derived ATP promotes BDNF release. This microglial-derived BDNF increases synaptic activation of excitatory dorsal horn neurons and decreases that of inhibitory neurons. It also alters the neuronal chloride gradient such the normal inhibitory effect of GABA is converted to excitation. By as yet undefined processes, this attenuated inhibition increases NMDA receptor function. BDNF also promotes the release of pro-inflammatory cytokines from astrocytes. All of these actions culminate in the increase dorsal horn excitability that underlies many forms of neuropathic pain. Peripheral nerve injury also alters excitability of structures in the thalamus, cortex and mesolimbic system that are responsible for pain perception and for the generation of co-morbidities such as anxiety and depression. The weight of evidence from male rodents suggests that this preferential modulation of excitably of supra-spinal pain processing structures also involves the action of microglial-derived BDNF. Possible mechanisms promoting the preferential release of BDNF in pain signaling structures are discussed. In females, invading T-lymphocytes increase dorsal horn excitability but it remains to be determined whether similar processes operate in supra-spinal structures. Despite its ubiquitous role in pain aetiology neither BDNF nor TrkB receptors represent potential therapeutic targets.
Collapse
Affiliation(s)
- Peter A Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
9
|
Smith PA. The Known Biology of Neuropathic Pain and Its Relevance to Pain Management. Can J Neurol Sci 2024; 51:32-39. [PMID: 36799022 DOI: 10.1017/cjn.2023.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Patients with neuropathic pain are heterogeneous in pathophysiology, etiology, and clinical presentation. Signs and symptoms are determined by the nature of the injury and factors such as genetics, sex, prior injury, age, culture, and environment. Basic science has provided general information about pain etiology by studying the consequences of peripheral injury in rodent models. This is associated with the release of inflammatory cytokines, chemokines, and growth factors that sensitize sensory nerve endings, alter gene expression, promote post-translational modification of proteins, and alter ion channel function. This leads to spontaneous activity in primary afferent neurons that is crucial for the onset and persistence of pain and the release of secondary mediators such as colony-stimulating factor 1 from primary afferent terminals. These promote the release of tertiary mediators such as brain-derived neurotrophic factor and interleukin-1β from microglia and astrocytes. Tertiary mediators facilitate the transmission of nociceptive information at the spinal, thalamic, and cortical levels. For the most part, these findings have failed to identify new therapeutic approaches. More recent basic science has better mirrored the clinical situation by addressing the pathophysiology associated with specific types of injury, refinement of methodology, and attention to various contributory factors such as sex. Improved quantification of sensory profiles in each patient and their distribution into defined clusters may improve translation between basic science and clinical practice. If such quantification can be traced back to cellular and molecular aspects of pathophysiology, this may lead to personalized medicine approaches that dictate a rational therapeutic approach for each individual.
Collapse
Affiliation(s)
- Peter A Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| |
Collapse
|
10
|
Jang K, Garraway SM. A review of dorsal root ganglia and primary sensory neuron plasticity mediating inflammatory and chronic neuropathic pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100151. [PMID: 38314104 PMCID: PMC10837099 DOI: 10.1016/j.ynpai.2024.100151] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024]
Abstract
Pain is a sensory state resulting from complex integration of peripheral nociceptive inputs and central processing. Pain consists of adaptive pain that is acute and beneficial for healing and maladaptive pain that is often persistent and pathological. Pain is indeed heterogeneous, and can be expressed as nociceptive, inflammatory, or neuropathic in nature. Neuropathic pain is an example of maladaptive pain that occurs after spinal cord injury (SCI), which triggers a wide range of neural plasticity. The nociceptive processing that underlies pain hypersensitivity is well-studied in the spinal cord. However, recent investigations show maladaptive plasticity that leads to pain, including neuropathic pain after SCI, also exists at peripheral sites, such as the dorsal root ganglia (DRG), which contains the cell bodies of sensory neurons. This review discusses the important role DRGs play in nociceptive processing that underlies inflammatory and neuropathic pain. Specifically, it highlights nociceptor hyperexcitability as critical to increased pain states. Furthermore, it reviews prior literature on glutamate and glutamate receptors, voltage-gated sodium channels (VGSC), and brain-derived neurotrophic factor (BDNF) signaling in the DRG as important contributors to inflammatory and neuropathic pain. We previously reviewed BDNF's role as a bidirectional neuromodulator of spinal plasticity. Here, we shift focus to the periphery and discuss BDNF-TrkB expression on nociceptors, non-nociceptor sensory neurons, and non-neuronal cells in the periphery as a potential contributor to induction and persistence of pain after SCI. Overall, this review presents a comprehensive evaluation of large bodies of work that individually focus on pain, DRG, BDNF, and SCI, to understand their interaction in nociceptive processing.
Collapse
Affiliation(s)
- Kyeongran Jang
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Sandra M. Garraway
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
11
|
Hayashi K, Lesnak JB, Plumb AN, Janowski AJ, Smith AF, Hill JK, Sluka KA. Brain-derived neurotrophic factor contributes to activity-induced muscle pain in male but not female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.565022. [PMID: 37961342 PMCID: PMC10635076 DOI: 10.1101/2023.10.31.565022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Activity-induced muscle pain increases release of interleukin-1β (IL-1β) in muscle macrophages and the development of pain is prevented by blockade of IL-1β. Brain derived neurotrophic factor (BDNF) is released from sensory neurons in response to IL-1β and mediates both inflammatory and neuropathic pain. Thus, we hypothesized that metabolites released during fatiguing muscle contractions activate macrophages to release IL-1β, which subsequently activate sensory neurons to secrete BDNF. To test this hypothesis, we used an animal model of activity-induced pain induced by repeated intramuscular acidic saline injections combined with fatiguing muscle contractions. Intrathecal or intramuscular injection of inhibitors of BDNF-Tropomyosin receptor kinase B (TrkB) signaling, ANA-12 or TrkB-Fc, reduced the decrease in muscle withdrawal thresholds in male, but not in female, mice when given before or 24hr after, but not 1 week after induction of the model. BDNF messenger ribonucleic acid (mRNA) was significantly increased in L4-L6 dorsal root ganglion (DRG), but not the spinal dorsal horn or gastrocnemius muscle, 24hr after induction of the model in either male or female mice. No changes in TrkB mRNA or p75 neurotrophin receptor mRNA were observed. BDNF protein expression via immunohistochemistry was significantly increased in L4-L6 spinal dorsal horn and retrogradely labelled muscle afferent DRG neurons, at 24hr after induction of the model in both sexes. In cultured DRG, fatigue metabolites combined with IL-1β significantly increased BDNF expression in both sexes. In summary, fatigue metabolites release, combined with IL-1β, BDNF from primary DRG neurons and contribute to activity-induced muscle pain only in males, while there were no sex differences in the changes in expression observed in BDNF.
Collapse
Affiliation(s)
- Kazuhiro Hayashi
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Joseph B. Lesnak
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Ashley N. Plumb
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Adam J. Janowski
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Angela F. Smith
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Joslyn K. Hill
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| | - Kathleen A. Sluka
- Department of Physical Therapy and Rehabilitation Science, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
12
|
Pourbagher-Shahri AM, Forouzanfar F. Saffron (Crocus sativus) and its constituents for pain management: A review of current evidence. Phytother Res 2023; 37:5041-5057. [PMID: 37528638 DOI: 10.1002/ptr.7968] [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: 02/09/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 08/03/2023]
Abstract
Pain can become a chronic and deliberating experience with a significant burden. In preclinical and clinical studies, Saffron (Crocus sativus L.) has shown analgesic activities. Considering the unsatisfactory results of current therapeutic management for chronic pain conditions, we aimed to review saffron's analgesic activity and underlying mechanisms. Saffron showed antinociceptive activities in formalin-, carrageenan-, and capsaicin-induced experimental pain models. Saffron analgesic activities affected several targets, including ion channels of nociceptors; the adrenergic system and central histaminic system; inhibition of inflammatory pathways, apoptotic pathways, and oxidative stress; regulation of NO pathway, and the endocannabinoid system. Clinical studies showed analgesia of Saffron in rheumatoid arthritis, after-pain following childbirth, dysmenorrhea, and fibromyalgia. Our literature review showed that saffron can be beneficial as an adjunct therapy to commonly used analgesics in practice, particularly in chronic pain conditions.
Collapse
Affiliation(s)
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Xie RG, Xu GY, Wu SX, Luo C. Presynaptic glutamate receptors in nociception. Pharmacol Ther 2023; 251:108539. [PMID: 37783347 DOI: 10.1016/j.pharmthera.2023.108539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/19/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Chronic pain is a frequent, distressing and poorly understood health problem. Plasticity of synaptic transmission in the nociceptive pathways after inflammation or injury is assumed to be an important cellular basis for chronic, pathological pain. Glutamate serves as the main excitatory neurotransmitter at key synapses in the somatosensory nociceptive pathways, in which it acts on both ionotropic and metabotropic glutamate receptors. Although conventionally postsynaptic, compelling anatomical and physiological evidence demonstrates the presence of presynaptic glutamate receptors in the nociceptive pathways. Presynaptic glutamate receptors play crucial roles in nociceptive synaptic transmission and plasticity. They modulate presynaptic neurotransmitter release and synaptic plasticity, which in turn regulates pain sensitization. In this review, we summarize the latest understanding of the expression of presynaptic glutamate receptors in the nociceptive pathways, and how they contribute to nociceptive information processing and pain hypersensitivity associated with inflammation / injury. We uncover the cellular and molecular mechanisms of presynaptic glutamate receptors in shaping synaptic transmission and plasticity to mediate pain chronicity, which may provide therapeutic approaches for treatment of chronic pain.
Collapse
Affiliation(s)
- Rou-Gang Xie
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Sheng-Xi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
14
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Nabhan AB. Pathophysiology, Clinical Implications and Management of Orofacial Neuropathic Pain- with special attention to Trigeminal neuralgia: A Narrative Review. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2023; 16:835-846. [DOI: 10.13005/bpj/2666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Background: It is a widely held belief that if the trigeminal nerve is damaged, the victim would experience agonising and unrelenting external pain. A lesion to the trigeminal nerve may have a wide-reaching effect, such as on one side of the face in particular, or it might have a more localised effect, such as on some or all of your gums. The risk of damage increases the likelihood that it will be difficult to speak and swallow. This nerve provides sensation to a part of your face that may be constantly aching or tingling for some people. However, the trigeminal nerve injury-related persistent orofacial pain might be brought on by a wide variety of unknown triggers. Aim: In this study investigate the clinical manifestations of chronic orofacial pain brought on by a damage to the trigeminal nerve, as well as the diagnostic and therapeutic approaches available to treat this condition. Methodology Through the use of search phrases such as "Trigeminal nerve injury," "Trigeminal ganglion," "Trigeminal spinal subnucleus caudalis," "Craniofacial pain," "Oral prognosis," and "treatment," the computerised databases for the last twenty years have been investigated. There are now two hundred objects in total that have been accumulated. There have been around fifty of them that are pertinent to the discussion that is going on in this work. Majority of the patients fair enough with the pharmacology treatment/drugs like the carbamazepine & oxcarbazepine which forms the first line treatment options followed by lamotrigine & baclofen encompassing the second line of drugs along with adjuvant drug support of topiramate, levetiracetam, gabapentin, pregabalin. As the field of science has explored &advanced for the latest treatment options include microvascular decompression, gamma knife radiosurgery, percutaneous rhizotomies variable based on the evidences & guidelines 54 Conclusion: New diagnostic criteria and treatment alternatives have become available for people who suffer from trigeminal neuropathy and orofacial neuropathic pain as a result of recent developments in fundamental animal research that have led to their development. Despite the results, more research needs to investigate a greater variety of distinct non-neuronal cell feature approaches.
Collapse
Affiliation(s)
- Abdullah Bin Nabhan
- Oral Medicine and Orofacial Pain, College of Dentistry, Prince Sattam Bin Abdulaziz University, Al Kharj, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Inyang KE, Sim J, Clark KB, Matan G, Monahan K, Evans C, Beng P, Ma JV, Heijnen CJ, Dantzer R, Scherrer G, Kavelaars A, Bernard M, Aldhamen Y, Folger JK, Laumet G. Tonic Meningeal Interleukin-10 Upregulates Delta Opioid Receptor to Prevent Relapse to Pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544200. [PMID: 37333074 PMCID: PMC10274865 DOI: 10.1101/2023.06.08.544200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chronic pain often alternates between transient remission and relapse of severe pain. While most research on chronic pain has focused on mechanisms maintaining pain, there is a critical unmet need to understand what prevents pain from re-emerging in those who recover from acute pain. We found that interleukin (IL)-10, a pain resolving cytokine, is persistently produced by resident macrophages in the spinal meninges during remission from pain. IL-10 upregulated expression and analgesic activity of δ-opioid receptor (δOR) in the dorsal root ganglion. Genetic or pharmacological inhibition of IL-10 signaling or δOR triggered relapse to pain in both sexes. These data challenge the widespread assumption that remission of pain is simply a return to the naïve state before pain was induced. Instead, our findings strongly suggest a novel concept that: remission is a state of lasting pain vulnerability that results from a long-lasting neuroimmune interactions in the nociceptive system.
Collapse
|
17
|
Tonyan S, Pospelova M, Krasnikova V, Fionik O, Alekseeva T, Samochernykh K, Ivanova N, Vavilova T, Vasilieva E, Makhanova A, Nikolaeva A, Bukkieva T, Combs S, Shevtsov M. Neurotrophin-3 (NT-3) as a Potential Biomarker of the Peripheral Nervous System Damage Following Breast Cancer Treatment. PATHOPHYSIOLOGY 2023; 30:110-122. [PMID: 37092524 PMCID: PMC10123681 DOI: 10.3390/pathophysiology30020010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023] Open
Abstract
Damage to the peripheral nervous system (PNS) is a common complication of breast cancer (BC) treatment, with 60 to 80% of breast cancer survivors experiencing symptoms of PNS damage. In the current study, the levels of brain-derived neurotrophic factor (BDNF), galectin-3 (Gal-3), and neurotrophin-3 (NT-3) were measured in the blood serum of BC patients by ELISA as potential biomarkers that might indicate the PNS damage. Sixty-seven patients were enrolled in this multi-center trial and compared to the aged-matched healthy female volunteers (control group) (n = 25). Intergroup comparison of biomarker levels (i.e., Gal-3 and BDNF) did not show significant differences in any of the studied subgroups. However, intriguingly, NT-3 levels were significantly higher in BC patients as compared to healthy volunteers, constituting 14.85 [10.3; 18.0] and 5.74 [4.56; 13.7] pg/mL, respectively (p < 0.001). In conclusion, NT-3 might be employed as a potential biomarker in BC patients with clinical manifestations of PNS damage. However, further studies to validate its correlation to the degree of peripheral nervous system lesions are of high value.
Collapse
Affiliation(s)
- Samvel Tonyan
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Maria Pospelova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Varvara Krasnikova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Olga Fionik
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Tatyana Alekseeva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Konstantin Samochernykh
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Nataliya Ivanova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Tatyana Vavilova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Elena Vasilieva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Albina Makhanova
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Aleksandra Nikolaeva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Tatyana Bukkieva
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Stephanie Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
| | - Maxim Shevtsov
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
18
|
Thakkar B, Acevedo EO. BDNF as a biomarker for neuropathic pain: Consideration of mechanisms of action and associated measurement challenges. Brain Behav 2023; 13:e2903. [PMID: 36722793 PMCID: PMC10013954 DOI: 10.1002/brb3.2903] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 01/08/2023] [Accepted: 01/16/2023] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION The primary objective of this paper is to (1) provide a summary of human studies that have used brain derived neurotrophic factor (BDNF) as a biomarker, (2) review animal studies that help to elucidate the mechanistic involvement of BDNF in the development and maintenance of neuropathic pain (NP), and (3) provide a critique of the existing measurement techniques to highlight the limitations of the methods utilized to quantify BDNF in different biofluids in the blood (i.e., serum and plasma) with the intention of presenting a case for the most reliable and valid technique. Lastly, this review also explores potential moderators that can influence the measurement of BDNF and provides recommendations to standardize its quantification to reduce the inconsistencies across studies. METHODS In this manuscript we examined the literature on BDNF, focusing on its role as a biomarker, its mechanism of action in NP, and critically analyzed its measurement in serum and plasma to identify factors that contribute to the discrepancy in results between plasma and serum BDNF values. RESULTS A large heterogenous literature was reviewed that detailed BDNF's utility as a potential biomarker in healthy volunteers, patients with chronic pain, and patients with neuropsychiatric disorders but demonstrated inconsistent findings. The literature provides insight into the mechanism of action of BDNF at different levels of the central nervous system using animal studies. We identified multiple factors that influence the measurement of BDNF in serum and plasma and based on current evidence, we recommend assessing serum BDNF levels to quantify peripheral BDNF as they are more stable and sensitive to changes than plasma BDNF. CONCLUSION Although mechanistic studies clearly explain the role of BDNF, results from human studies are inconsistent. More studies are needed to evaluate the methodological challenges in using serum BDNF as a biomarker in NP.
Collapse
Affiliation(s)
- Bhushan Thakkar
- Department of Physical Therapy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Edmund O Acevedo
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
19
|
Di-Bonaventura S, Fernández-Carnero J, Matesanz-García L, Arribas-Romano A, Polli A, Ferrer-Peña R. Effect of Different Physical Therapy Interventions on Brain-Derived Neurotrophic Factor Levels in Chronic Musculoskeletal Pain Patients: A Systematic Review. Life (Basel) 2023; 13:163. [PMID: 36676112 PMCID: PMC9867147 DOI: 10.3390/life13010163] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE The main objectives of this review were, firstly, to study the effect of different physiotherapy interventions on BDNF levels, and, secondly, to analyze the influence of physiotherapy on pain levels to subsequently draw conclusions about its possible relationship with BDNF. BACKGROUND Based on the theory that neurotrophic factors such as BDNF play a fundamental role in the initiation and/or maintenance of hyperexcitability of central neurons in pain, it was hypothesized that the levels of this neurotrophic factor may be modified by the application of therapeutic interventions, favoring a reduction in pain intensity. METHODS A literature search of multiple electronic databases (Pubmed, PsycINFO, Medline (Ebsco), Scopus, WOS, Embase) was conducted to identify randomized control trials (RCTs) published without language restrictions up to and including March 2022. The search strategy was based on the combination of medical terms (Mesh) and keywords relating to the following concepts: "pain", "chronic pain", "brain derived neurotrophic factor", "BDNF", "physiotherapy", and "physical therapy". A total of seven papers were included. RESULTS There were two studies that showed statistically significant differences in pain intensity reduction and an increase in the BDNF levels that used therapies such as rTMS and EIMS in patients with chronic myofascial pain. However, the same conclusions cannot be drawn for the other physical therapies applied. CONCLUSIONS rTMS and EIMS interventions achieved greater short-term reductions in pain intensity and increased BDNF over other types of interventions in chronic myofascial pain patients, as demonstrated by a moderate amount of evidence. In contrast, other types of physical therapy (PT) interventions did not appear to be more effective in decreasing pain intensity and increasing BDNF levels than placebo PT or minimal intervention, as a low amount of evidence was found.
Collapse
Affiliation(s)
- Silvia Di-Bonaventura
- Escuela Internacional de Doctorado, Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, 28933 Alcorcón, Spain
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, 28922 Alcorcón, Spain
| | - Josué Fernández-Carnero
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, 28922 Alcorcón, Spain
- Grupo Multidisciplinar de Investigación y Tratamiento del Dolor, Grupo de Excelencia Investigadora URJC-Banco de Santander, 28922 Madrid, Spain
- La Paz Hospital Institute for Health Research, IdiPAZ, 28029 Madrid, Spain
- Motion in Brains Research Group, Institute of Neuroscience and Movement Sciences (INCIMOV), Centro Superior de Estudios Universitarios La Salle, Universidad Autonóma de Madrid, 28023 Madrid, Spain
- Grupo de Investigación de Dolor Musculoesqueletico y Control Motor, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain
| | - Luis Matesanz-García
- Departamento de Fisioterapia, Facultad de Ciencias de la Salud, CSEU La Salle, Universidad Autonóma de Madrid, 28023 Madrid, Spain
| | - Alberto Arribas-Romano
- Escuela Internacional de Doctorado, Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, 28933 Alcorcón, Spain
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, 28922 Alcorcón, Spain
| | - Andrea Polli
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Vrije Universiteit Brussel, Pleinlaan 22, 1050 Brussels, Belgium
- Centre for Environment and Health, Department of Public Health and Primary Care, KU Leuven, Blok D, Bus 7001, 3000 Leuven, Belgium
| | - Raúl Ferrer-Peña
- La Paz Hospital Institute for Health Research, IdiPAZ, 28029 Madrid, Spain
- Motion in Brains Research Group, Institute of Neuroscience and Movement Sciences (INCIMOV), Centro Superior de Estudios Universitarios La Salle, Universidad Autonóma de Madrid, 28023 Madrid, Spain
- Departamento de Fisioterapia, Facultad de Ciencias de la Salud, CSEU La Salle, Universidad Autonóma de Madrid, 28023 Madrid, Spain
| |
Collapse
|
20
|
Pike CK, Kim M, Schnitzer K, Mercaldo N, Edwards R, Napadow V, Zhang Y, Morrissey EJ, Alshelh Z, Evins AE, Loggia ML, Gilman JM. Study protocol for a phase II, double-blind, randomised controlled trial of cannabidiol (CBD) compared with placebo for reduction of brain neuroinflammation in adults with chronic low back pain. BMJ Open 2022; 12:e063613. [PMID: 36123113 PMCID: PMC9486315 DOI: 10.1136/bmjopen-2022-063613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/02/2022] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Chronic pain is a debilitating medical problem that is difficult to treat. Neuroinflammatory pathways have emerged as a potential therapeutic target, as preclinical studies have demonstrated that glial cells and neuroglial interactions play a role in the establishment and maintenance of pain. Recently, we used positron emission tomography (PET) to demonstrate increased levels of 18 kDa translocator protein (TSPO) binding, a marker of glial activation, in patients with chronic low back pain (cLBP). Cannabidiol (CBD) is a glial inhibitor in animal models, but studies have not assessed whether CBD reduces neuroinflammation in humans. The principal aim of this trial is to evaluate whether CBD, compared with placebo, affects neuroinflammation, as measured by TSPO levels. METHODS AND ANALYSIS This is a double-blind, randomised, placebo-controlled, phase II clinical trial. Eighty adults (aged 18-75) with cLBP for >6 months will be randomised to either an FDA-approved CBD medication (Epidiolex) or matching placebo for 4 weeks using a dose-escalation design. All participants will undergo integrated PET/MRI at baseline and after 4 weeks of treatment to evaluate neuroinflammation using [11C]PBR28, a second-generation radioligand for TSPO. Our primary hypothesis is that participants randomised to CBD will demonstrate larger reductions in thalamic [11C]PBR28 signal compared with those receiving placebo. We will also assess the effect of CBD on (1) [11C]PBR28 signal from limbic regions, which our prior work has linked to depressive symptoms and (2) striatal activation in response to a reward task. Additionally, we will evaluate self-report measures of cLBP intensity and bothersomeness, depression and quality of life at baseline and 4 weeks. ETHICS AND DISSEMINATION This protocol is approved by the Massachusetts General Brigham Human Research Committee (protocol number: 2021P002617) and FDA (IND number: 143861) and registered with ClinicalTrials.gov. Results will be published in peer-reviewed journals and presented at conferences. TRIAL REGISTRATION NUMBER NCT05066308; ClinicalTrials.gov.
Collapse
Affiliation(s)
- Chelsea K Pike
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
- Massachusetts General Hospital Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
| | - Minhae Kim
- Massachusetts General Hospital Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
| | - Kristina Schnitzer
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathaniel Mercaldo
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert Edwards
- Department of Anesthesiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Vitaly Napadow
- Massachusetts General Hospital Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Charlestown, Massachusetts, USA
| | - Yi Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Erin Janas Morrissey
- Massachusetts General Hospital Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
| | - Zeynab Alshelh
- Massachusetts General Hospital Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts, USA
| | - A Eden Evins
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Marco L Loggia
- Massachusetts General Hospital Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
- Department of Radiology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jodi M Gilman
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
- Massachusetts General Hospital Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Shinoda M, Hitomi S, Iwata K, Hayashi Y. Plastic changes in nociceptive pathways contributing to persistent orofacial pain. J Oral Biosci 2022; 64:263-270. [PMID: 35840073 DOI: 10.1016/j.job.2022.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Pain is a warning signal for the body defense mechanisms and is a critical sensation for supporting life. However, orofacial pain is not a vital sensation, but a disease. However, there are still many unclear points about the pathophysiological mechanism of orofacial pain. This situation makes it difficult for many clinicians to treat orofacial pain hypersensitivity. HIGHLIGHT Noxious information on the orofacial region received by trigeminal ganglion neurons is recognized as "orofacial pain" by being transmitted to the somatosensory cortex and limbic system via the spinal trigeminal nucleus and the thalamic sensory nuclei. Orofacial inflammation or trigeminal nerve injury causes neuropathic changes in various nociceptive signaling pathways, resulting in persistent orofacial pain. It is considered that persistent oral facial pain is triggered by plastic changes in nociceptive signaling pathways involving various cells such as satellite glial cells, astrocytes, microglia, and macrophages, as well as nociceptive neurons. CONCLUSION Recent studies have shown that hyperexcitability of nociceptive neurons in the nociceptive signaling pathways of the orofacial region caused by a variety of factors causes persistent orofacial pain. This review outlines the pathophysiology of orofacial pain along with the results of our study.
Collapse
Affiliation(s)
- Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| |
Collapse
|
22
|
The BDNF-TrkB signaling pathway in the rostral anterior cingulate cortex is involved in the development of pain aversion in rats with bone cancer via NR2B and ERK-CREB signaling. Brain Res Bull 2022; 185:18-27. [DOI: 10.1016/j.brainresbull.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/15/2022] [Accepted: 04/05/2022] [Indexed: 11/19/2022]
|
23
|
Dedek A, Hildebrand ME. Advances and Barriers in Understanding Presynaptic N-Methyl-D-Aspartate Receptors in Spinal Pain Processing. Front Mol Neurosci 2022; 15:864502. [PMID: 35431805 PMCID: PMC9008455 DOI: 10.3389/fnmol.2022.864502] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022] Open
Abstract
For decades, N-methyl-D-aspartate (NMDA) receptors have been known to play a critical role in the modulation of both acute and chronic pain. Of particular interest are NMDA receptors expressed in the superficial dorsal horn (SDH) of the spinal cord, which houses the nociceptive processing circuits of the spinal cord. In the SDH, NMDA receptors undergo potentiation and increases in the trafficking of receptors to the synapse, both of which contribute to increases in excitability and plastic increases in nociceptive output from the SDH to the brain. Research efforts have primarily focused on postsynaptic NMDA receptors, despite findings that presynaptic NMDA receptors can undergo similar plastic changes to their postsynaptic counterparts. Recent technological advances have been pivotal in the discovery of mechanisms of plastic changes in presynaptic NMDA receptors within the SDH. Here, we highlight these recent advances in the understanding of presynaptic NMDA receptor physiology and their modulation in models of chronic pain. We discuss the role of specific NMDA receptor subunits in presynaptic membranes of nociceptive afferents and local SDH interneurons, including their modulation across pain modalities. Furthermore, we discuss how barriers such as lack of sex-inclusive research and differences in neurodevelopmental timepoints have complicated investigations into the roles of NMDA receptors in pathological pain states. A more complete understanding of presynaptic NMDA receptor function and modulation across pain states is needed to shed light on potential new therapeutic treatments for chronic pain.
Collapse
Affiliation(s)
- Annemarie Dedek
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- Neuroscience Department, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Michael E. Hildebrand
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- Neuroscience Department, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- *Correspondence: Michael E. Hildebrand,
| |
Collapse
|
24
|
Xie RG, Chu WG, Liu DL, Wang X, Ma SB, Wang F, Wang FD, Lin Z, Wu WB, Lu N, Liu YY, Han WJ, Zhang H, Bai ZT, Hu SJ, Tao HR, Kuner T, Zhang X, Kuner R, Wu SX, Luo C. Presynaptic NMDARs on spinal nociceptor terminals state-dependently modulate synaptic transmission and pain. Nat Commun 2022; 13:728. [PMID: 35132099 PMCID: PMC8821657 DOI: 10.1038/s41467-022-28429-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 01/21/2022] [Indexed: 12/24/2022] Open
Abstract
Postsynaptic NMDARs at spinal synapses are required for postsynaptic long-term potentiation and chronic pain. However, how presynaptic NMDARs (PreNMDARs) in spinal nociceptor terminals control presynaptic plasticity and pain hypersensitivity has remained unclear. Here we report that PreNMDARs in spinal nociceptor terminals modulate synaptic transmission in a nociceptive tone-dependent manner. PreNMDARs depresses presynaptic transmission in basal state, while paradoxically causing presynaptic potentiation upon injury. This state-dependent modulation is dependent on Ca2+ influx via PreNMDARs. Small conductance Ca2+-activated K+ (SK) channels are responsible for PreNMDARs-mediated synaptic depression. Rather, tissue inflammation induces PreNMDARs-PKG-I-dependent BDNF secretion from spinal nociceptor terminals, leading to SK channels downregulation, which in turn converts presynaptic depression to potentiation. Our findings shed light on the state-dependent characteristics of PreNMDARs in spinal nociceptor terminals on modulating nociceptive transmission and revealed a mechanism underlying state-dependent transition. Moreover, we identify PreNMDARs in spinal nociceptor terminals as key constituents of activity-dependent pain sensitization. Postsynaptic NMDARs at spinal synapses are required for postsynaptic long-term potentiation and chronic pain. Here, the authors show that also presynaptic NMDARs in spinal nociceptor terminals modulate synaptic transmission in a nociceptive tone-dependent manner.
Collapse
|
25
|
Király K, Karádi DÁ, Zádor F, Mohammadzadeh A, Galambos AR, Balogh M, Riba P, Tábi T, Zádori ZS, Szökő É, Fürst S, Al-Khrasani M. Shedding Light on the Pharmacological Interactions between μ-Opioid Analgesics and Angiotensin Receptor Modulators: A New Option for Treating Chronic Pain. Molecules 2021; 26:6168. [PMID: 34684749 PMCID: PMC8537077 DOI: 10.3390/molecules26206168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/20/2022] Open
Abstract
The current protocols for neuropathic pain management include µ-opioid receptor (MOR) analgesics alongside other drugs; however, there is debate on the effectiveness of opioids. Nevertheless, dose escalation is required to maintain their analgesia, which, in turn, contributes to a further increase in opioid side effects. Finding novel approaches to effectively control chronic pain, particularly neuropathic pain, is a great challenge clinically. Literature data related to pain transmission reveal that angiotensin and its receptors (the AT1R, AT2R, and MAS receptors) could affect the nociception both in the periphery and CNS. The MOR and angiotensin receptors or drugs interacting with these receptors have been independently investigated in relation to analgesia. However, the interaction between the MOR and angiotensin receptors has not been excessively studied in chronic pain, particularly neuropathy. This review aims to shed light on existing literature information in relation to the analgesic action of AT1R and AT2R or MASR ligands in neuropathic pain conditions. Finally, based on literature data, we can hypothesize that combining MOR agonists with AT1R or AT2R antagonists might improve analgesia.
Collapse
MESH Headings
- Analgesics/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Chronic Pain/drug therapy
- Humans
- Neuralgia/drug therapy
- Nociception/drug effects
- Pain Management/methods
- Proto-Oncogene Mas
- Receptors, Angiotensin/drug effects
- Receptors, Angiotensin/metabolism
- Receptors, Opioid/agonists
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Kornél Király
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Dávid Á. Karádi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Ferenc Zádor
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Amir Mohammadzadeh
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Anna Rita Galambos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Mihály Balogh
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Pál Riba
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Tamás Tábi
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Zoltán S. Zádori
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Éva Szökő
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Susanna Fürst
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| |
Collapse
|
26
|
Boakye PA, Tang SJ, Smith PA. Mediators of Neuropathic Pain; Focus on Spinal Microglia, CSF-1, BDNF, CCL21, TNF-α, Wnt Ligands, and Interleukin 1β. FRONTIERS IN PAIN RESEARCH 2021; 2:698157. [PMID: 35295524 PMCID: PMC8915739 DOI: 10.3389/fpain.2021.698157] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/14/2021] [Indexed: 01/04/2023] Open
Abstract
Intractable neuropathic pain is a frequent consequence of nerve injury or disease. When peripheral nerves are injured, damaged axons undergo Wallerian degeneration. Schwann cells, mast cells, fibroblasts, keratinocytes and epithelial cells are activated leading to the generation of an "inflammatory soup" containing cytokines, chemokines and growth factors. These primary mediators sensitize sensory nerve endings, attract macrophages, neutrophils and lymphocytes, alter gene expression, promote post-translational modification of proteins, and alter ion channel function in primary afferent neurons. This leads to increased excitability and spontaneous activity and the generation of secondary mediators including colony stimulating factor 1 (CSF-1), chemokine C-C motif ligand 21 (CCL-21), Wnt3a, and Wnt5a. Release of these mediators from primary afferent neurons alters the properties of spinal microglial cells causing them to release tertiary mediators, in many situations via ATP-dependent mechanisms. Tertiary mediators such as BDNF, tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), and other Wnt ligands facilitate the generation and transmission of nociceptive information by increasing excitatory glutamatergic transmission and attenuating inhibitory GABA and glycinergic transmission in the spinal dorsal horn. This review focusses on activation of microglia by secondary mediators, release of tertiary mediators from microglia and a description of their actions in the spinal dorsal horn. Attention is drawn to the substantial differences in the precise roles of various mediators in males compared to females. At least 25 different mediators have been identified but the similarity of their actions at sensory nerve endings, in the dorsal root ganglia and in the spinal cord means there is considerable redundancy in the available mechanisms. Despite this, behavioral studies show that interruption of the actions of any single mediator can relieve signs of pain in experimental animals. We draw attention this paradox. It is difficult to explain how inactivation of one mediator can relieve pain when so many parallel pathways are available.
Collapse
Affiliation(s)
- Paul A. Boakye
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Shao-Jun Tang
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Shinoda M, Imamura Y, Hayashi Y, Noma N, Okada-Ogawa A, Hitomi S, Iwata K. Orofacial Neuropathic Pain-Basic Research and Their Clinical Relevancies. Front Mol Neurosci 2021; 14:691396. [PMID: 34295221 PMCID: PMC8291146 DOI: 10.3389/fnmol.2021.691396] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/21/2021] [Indexed: 01/01/2023] Open
Abstract
Trigeminal nerve injury is known to cause severe persistent pain in the orofacial region. This pain is difficult to diagnose and treat. Recently, many animal studies have reported that rewiring of the peripheral and central nervous systems, non-neuronal cell activation, and up- and down-regulation of various molecules in non-neuronal cells are involved in the development of this pain following trigeminal nerve injury. However, there are many unknown mechanisms underlying the persistent orofacial pain associated with trigeminal nerve injury. In this review, we address recent animal data regarding the involvement of various molecules in the communication of neuronal and non-neuronal cells and examine the possible involvement of ascending pathways in processing pathological orofacial pain. We also address the clinical observations of persistent orofacial pain associated with trigeminal nerve injury and clinical approaches to their diagnosis and treatment.
Collapse
Affiliation(s)
- Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiki Imamura
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Noboru Noma
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, Tokyo, Japan
| | - Akiko Okada-Ogawa
- Department of Oral Diagnostic Sciences, Nihon University School of Dentistry, Tokyo, Japan
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
28
|
Li Y, Bao Y, Zheng H, Qin Y, Hua B. The nonreceptor protein tyrosine kinase Src participates in every step of cancer-induced bone pain. Biomed Pharmacother 2021; 141:111822. [PMID: 34147901 DOI: 10.1016/j.biopha.2021.111822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/30/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer-induced bone pain (CIBP) is a refractory form of pain that has a high incidence in advanced tumors. Src protein tyrosine kinase is mainly composed of six domains, with two states of automatic inhibition and activation. The modular domain allows Src to conveniently regulate by and communicate with a variety of proteins, directly or indirectly participate in each step of the CIBP process. Src is beneficial to the growth and proliferation of tumor cells, and it can promote the metastases of primary tumors to bone. In the microenvironment of bone metastasis, it mainly mediates bone resorption, activates related peripheral receptors to participate in the formation of pain signals, and may promote the generation of pathological sensory nerve fibers. In the process of pain signal transmission, it mainly mediates NMDAR and central glial cells to regulate pain signal intensity and central sensitization, but it is not limited to these two aspects. Both basic experimentation and clinical research have shown encouraging potential, providing new ideas and inspiration for the prevention and treatment of CIBP.
Collapse
Affiliation(s)
- Yaoyuan Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yinggang Qin
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
29
|
Lituma PJ, Kwon HB, Alviña K, Luján R, Castillo PE. Presynaptic NMDA receptors facilitate short-term plasticity and BDNF release at hippocampal mossy fiber synapses. eLife 2021; 10:e66612. [PMID: 34061025 PMCID: PMC8186907 DOI: 10.7554/elife.66612] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/28/2021] [Indexed: 01/12/2023] Open
Abstract
Neurotransmitter release is a highly controlled process by which synapses can critically regulate information transfer within neural circuits. While presynaptic receptors - typically activated by neurotransmitters and modulated by neuromodulators - provide a powerful way of fine-tuning synaptic function, their contribution to activity-dependent changes in transmitter release remains poorly understood. Here, we report that presynaptic NMDA receptors (preNMDARs) at mossy fiber boutons in the rodent hippocampus can be activated by physiologically relevant patterns of activity and selectively enhance short-term synaptic plasticity at mossy fiber inputs onto CA3 pyramidal cells and mossy cells, but not onto inhibitory interneurons. Moreover, preNMDARs facilitate brain-derived neurotrophic factor release and contribute to presynaptic calcium rise. Taken together, our results indicate that by increasing presynaptic calcium, preNMDARs fine-tune mossy fiber neurotransmission and can control information transfer during dentate granule cell burst activity that normally occur in vivo.
Collapse
Affiliation(s)
- Pablo J Lituma
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Hyung-Bae Kwon
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Karina Alviña
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Rafael Luján
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad Castilla-La ManchaAlbaceteSpain
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of MedicineBronxUnited States
| |
Collapse
|
30
|
Driving effect of BDNF in the spinal dorsal horn on neuropathic pain. Neurosci Lett 2021; 756:135965. [PMID: 34022262 DOI: 10.1016/j.neulet.2021.135965] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/30/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022]
Abstract
Neuropathic pain (NP) is caused by direct or indirect damage to the nervous system and is a common symptom of many diseases. The mechanisms underlying the onset and persistence of NP are unclear. Therefore, research concerning these mechanisms has become an important focus in the medical field. Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophic factor family of signaling molecules. BDNF is an important regulator of neuronal development, synaptic transmission, and cellular and synaptic plasticity, which are essential for nerve maintenance and repair. However, BDNF is upregulated in the spinal dorsal horn and can promote NP by activating glial cells, reducing inhibitory functions and enhancing excitement after nociceptive stimulation. This review considers the relationship between NP and BDNF signaling in the spinal dorsal horn and discusses potentially related pathological mechanisms.
Collapse
|
31
|
Lee SW, Han HC. Methylene Blue Application to Lessen Pain: Its Analgesic Effect and Mechanism. Front Neurosci 2021; 15:663650. [PMID: 34079436 PMCID: PMC8165385 DOI: 10.3389/fnins.2021.663650] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Methylene blue (MB) is a cationic thiazine dye, widely used as a biological stain and chemical indicator. Growing evidence have revealed that MB functions to restore abnormal vasodilation and notably it is implicated even in pain relief. Physicians began to inject MB into degenerated disks to relieve pain in patients with chronic discogenic low back pain (CDLBP), and some of them achieved remarkable outcomes. For osteoarthritis and colitis, MB abates inflammation by suppressing nitric oxide production, and ultimately relieves pain. However, despite this clinical efficacy, MB has not attracted much public attention in terms of pain relief. Accordingly, this review focuses on how MB lessens pain, noting three major actions of this dye: anti-inflammation, sodium current reduction, and denervation. Moreover, we showed controversies over the efficacy of MB on CDLBP and raised also toxicity issues to look into the limitation of MB application. This analysis is the first attempt to illustrate its analgesic effects, which may offer a novel insight into MB as a pain-relief dye.
Collapse
Affiliation(s)
- Seung Won Lee
- Good Doctor Research Institute, College of Medicine, Korea University, Seoul, South Korea
| | - Hee Chul Han
- Department of Physiology, College of Medicine and Neuroscience Research Institute, Korea University, Seoul, South Korea
| |
Collapse
|
32
|
Borges JP, Mekhail K, Fairn GD, Antonescu CN, Steinberg BE. Modulation of Pathological Pain by Epidermal Growth Factor Receptor. Front Pharmacol 2021; 12:642820. [PMID: 34054523 PMCID: PMC8149758 DOI: 10.3389/fphar.2021.642820] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic pain has been widely recognized as a major public health problem that impacts multiple aspects of patient quality of life. Unfortunately, chronic pain is often resistant to conventional analgesics, which are further limited by their various side effects. New therapeutic strategies and targets are needed to better serve the millions of people suffering from this devastating disease. To this end, recent clinical and preclinical studies have implicated the epidermal growth factor receptor signaling pathway in chronic pain states. EGFR is one of four members of the ErbB family of receptor tyrosine kinases that have key roles in development and the progression of many cancers. EGFR functions by activating many intracellular signaling pathways following binding of various ligands to the receptor. Several of these signaling pathways, such as phosphatidylinositol 3-kinase, are known mediators of pain. EGFR inhibitors are known for their use as cancer therapeutics but given recent evidence in pilot clinical and preclinical investigations, may have clinical use for treating chronic pain. Here, we review the clinical and preclinical evidence implicating EGFR in pathological pain states and provide an overview of EGFR signaling highlighting how EGFR and its ligands drive pain hypersensitivity and interact with important pain pathways such as the opioid system.
Collapse
Affiliation(s)
- Jazlyn P Borges
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Katrina Mekhail
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Gregory D Fairn
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Costin N Antonescu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Benjamin E Steinberg
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
33
|
García-Magro N, Martin YB, Negredo P, Zafra F, Avendaño C. Microglia and Inhibitory Circuitry in the Medullary Dorsal Horn: Laminar and Time-Dependent Changes in a Trigeminal Model of Neuropathic Pain. Int J Mol Sci 2021; 22:4564. [PMID: 33925417 PMCID: PMC8123867 DOI: 10.3390/ijms22094564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Craniofacial neuropathic pain affects millions of people worldwide and is often difficult to treat. Two key mechanisms underlying this condition are a loss of the negative control exerted by inhibitory interneurons and an early microglial reaction. Basic features of these mechanisms, however, are still poorly understood. Using the chronic constriction injury of the infraorbital nerve (CCI-IoN) model of neuropathic pain in mice, we have examined the changes in the expression of GAD, the synthetic enzyme of GABA, and GlyT2, the membrane transporter of glycine, as well as the microgliosis that occur at early (5 days) and late (21 days) stages post-CCI in the medullary and upper spinal dorsal horn. Our results show that CCI-IoN induces a down-regulation of GAD at both postinjury survival times, uniformly across the superficial laminae. The expression of GlyT2 showed a more discrete and heterogeneous reduction due to the basal presence in lamina III of 'patches' of higher expression, interspersed within a less immunoreactive 'matrix', which showed a more substantial reduction in the expression of GlyT2. These patches coincided with foci lacking any perceptible microglial reaction, which stood out against a more diffuse area of strong microgliosis. These findings may provide clues to better understand the neural mechanisms underlying allodynia in neuropathic pain syndromes.
Collapse
Affiliation(s)
- Nuria García-Magro
- Department of Anatomy, Histology and Neuroscience, Medical School, Autónoma University of Madrid, 28029 Madrid, Spain; (N.G.-M.); (P.N.)
- Ph.D. Programme in Neuroscience, Doctoral School, Autónoma University of Madrid, 28049 Madrid, Spain
| | - Yasmina B. Martin
- Departamento de Anatomía, Facultad de Medicina, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain;
| | - Pilar Negredo
- Department of Anatomy, Histology and Neuroscience, Medical School, Autónoma University of Madrid, 28029 Madrid, Spain; (N.G.-M.); (P.N.)
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| | - Carlos Avendaño
- Department of Anatomy, Histology and Neuroscience, Medical School, Autónoma University of Madrid, 28029 Madrid, Spain; (N.G.-M.); (P.N.)
| |
Collapse
|
34
|
Miller KM, Mercado NM, Sortwell CE. Synucleinopathy-associated pathogenesis in Parkinson's disease and the potential for brain-derived neurotrophic factor. NPJ PARKINSONS DISEASE 2021; 7:35. [PMID: 33846345 PMCID: PMC8041900 DOI: 10.1038/s41531-021-00179-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The lack of disease-modifying treatments for Parkinson’s disease (PD) is in part due to an incomplete understanding of the disease’s etiology. Alpha-synuclein (α-syn) has become a point of focus in PD due to its connection to both familial and idiopathic cases—specifically its localization to Lewy bodies (LBs), a pathological hallmark of PD. Within this review, we will present a comprehensive overview of the data linking synuclein-associated Lewy pathology with intracellular dysfunction. We first present the alterations in neuronal proteins and transcriptome associated with LBs in postmortem human PD tissue. We next compare these findings to those associated with LB-like inclusions initiated by in vitro exposure to α-syn preformed fibrils (PFFs) and highlight the profound and relatively unique reduction of brain-derived neurotrophic factor (BDNF) in this model. Finally, we discuss the multitude of ways in which BDNF offers the potential to exert disease-modifying effects on the basal ganglia. What remains unknown is the potential for BDNF to mitigate inclusion-associated dysfunction within the context of synucleinopathy. Collectively, this review reiterates the merit of using the PFF model as a tool to understand the physiological changes associated with LBs, while highlighting the neuroprotective potential of harnessing endogenous BDNF.
Collapse
Affiliation(s)
- Kathryn M Miller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA. .,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
35
|
Durante M, Squillace S, Lauro F, Giancotti LA, Coppi E, Cherchi F, Di Cesare Mannelli L, Ghelardini C, Kolar G, Wahlman C, Opejin A, Xiao C, Reitman ML, Tosh DK, Hawiger D, Jacobson KA, Salvemini D. Adenosine A3 agonists reverse neuropathic pain via T cell-mediated production of IL-10. J Clin Invest 2021; 131:139299. [PMID: 33621215 DOI: 10.1172/jci139299] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 02/19/2021] [Indexed: 12/16/2022] Open
Abstract
The A3 adenosine receptor (A3AR) has emerged as a therapeutic target with A3AR agonists to tackle the global challenge of neuropathic pain, and investigation into its mode of action is essential for ongoing clinical development. Immune cell A3ARs, and their activation during pathology, modulate cytokine release. Thus, the use of immune cells as a cellular substrate for the pharmacological action of A3AR agonists is enticing, but unknown. The present study discovered that Rag-KO mice lacking T and B cells, as compared with WT mice, are insensitive to the anti-allodynic effects of A3AR agonists. Similar findings were observed in interleukin-10 and interleukin-10 receptor knockout mice. Adoptive transfer of CD4+ T cells from WT mice infiltrated the dorsal root ganglion (DRG) and restored A3AR agonist-mediated anti-allodynia in Rag-KO mice. CD4+ T cells from Adora3-KO or Il10-KO mice did not. Transfer of CD4+ T cells from WT mice, but not Il10-KO mice, into Il10-KO mice or Adora3-KO mice fully reinstated the anti-allodynic effects of A3AR activation. Notably, A3AR agonism reduced DRG neuron excitability when cocultured with CD4+ T cells in an IL-10-dependent manner. A3AR action on CD4+ T cells infiltrated in the DRG decreased phosphorylation of GluN2B-containing N-methyl-D-aspartate receptors at Tyr1472, a modification associated with regulating neuronal hypersensitivity. Our findings establish that activation of A3AR on CD4+ T cells to release IL-10 is required and sufficient evidence for the use of A3AR agonists as therapeutics.
Collapse
Affiliation(s)
- Mariaconcetta Durante
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology, University of Florence, Florence, Italy
| | - Silvia Squillace
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome, Rome, Italy
| | - Filomena Lauro
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Institute of Research for Food Safety & Health, Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Luigino Antonio Giancotti
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology, University of Florence, Florence, Italy
| | - Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology, University of Florence, Florence, Italy
| | - Grant Kolar
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology, and
| | - Carrie Wahlman
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Adeleye Opejin
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Cuiying Xiao
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Marc L Reitman
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Dilip K Tosh
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Kenneth A Jacobson
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
36
|
Mizoguchi Y, Ohgidani M, Haraguchi Y, Murakawa-Hirachi T, Kato TA, Monji A. ProBDNF induces sustained elevation of intracellular Ca 2+ possibly mediated by TRPM7 channels in rodent microglial cells. Glia 2021; 69:1694-1708. [PMID: 33740269 DOI: 10.1002/glia.23996] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 01/07/2023]
Abstract
Microglia are intrinsic immune cells that release factors including pro- and anti-inflammatory cytokines, nitric oxide (NO) and neurotrophins following activation in the brain. Elevation of intracellular Ca2+ concentration ([Ca2+ ]i) is important for microglial functions, such as the release of cytokines or NO from activated microglia. Brain-derived neurotrophic factor (BDNF) is a neurotrophin well known for its roles in the activation of microglia. Interestingly, proBDNF, the precursor form of mature BDNF, and mature BDNF elicit opposing neuronal responses in the brain. Mature BDNF induces sustained intracellular Ca2+ elevation through the upregulation of the surface expression of TRPC3 channels in rodent microglial cells. In addition, TRPC3 channels are important for the BDNF-induced suppression of NO production in activated microglia. In this study, we observed that proBDNF and mature BDNF have opposite effects on the relative expression of surface p75 neurotrophin receptor (p75NTR ) in rodent microglial cells. ProBDNF induces a sustained elevation of [Ca2+ ]i through binding to the p75NTR , which is possibly mediated by Rac 1 activation and TRPM7 channels in rodent microglial cells. Flow cytometry showed that proBDNF increased the relative surface expression of TRPM7. Although proBDNF did not affect either mRNA expression of pro- and anti-inflammatory cytokines or the phagocytic activity, proBDNF potentiates the generation of NO induced by IFN-γ and TRPM7 channels could be involved in the proBDNF-induced potentiation of IFN-γ-mediated production of NO. We show direct evidence that rodent microglial cells are able to respond to proBDNF, which might be important for the regulation of inflammatory responses in the brain.
Collapse
Affiliation(s)
- Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, Saga, Japan
| | - Masahiro Ohgidani
- Department of Psychiatry, Faculty of Medicine, Saga University, Saga, Japan.,Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
37
|
Chen W, McRoberts JA, Ennes HS, Marvizon JC. cAMP signaling through protein kinase A and Epac2 induces substance P release in the rat spinal cord. Neuropharmacology 2021; 189:108533. [PMID: 33744339 DOI: 10.1016/j.neuropharm.2021.108533] [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: 08/12/2020] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 11/18/2022]
Abstract
Using neurokinin 1 receptor (NK1R) internalization to measure of substance P release in rat spinal cord slices, we found that it was induced by the adenylyl cyclase (AC) activator forskolin, by the protein kinase A (PKA) activators 6-Bnz-cAMP and 8-Br-cAMP, and by the activator of exchange protein activated by cAMP (Epac) 8-pCPT-2-O-Me-cAMP (CPTOMe-cAMP). Conversely, AC and PKA inhibitors decreased substance P release induced by electrical stimulation of the dorsal root. Therefore, the cAMP signaling pathway mediates substance P release in the dorsal horn. The effects of forskolin and 6-Bnz-cAMP were not additive with NMDA-induced substance P release and were decreased by the NMDA receptor blocker MK-801. In cultured dorsal horn neurons, forskolin increased NMDA-induced Ca2+ entry and the phosphorylation of the NR1 and NR2B subunits of the NMDA receptor. Therefore, cAMP-induced substance P release is mediated by the activating phosphorylation by PKA of NMDA receptors. Voltage-gated Ca2+ channels, but not by TRPV1 or TRPA1, also contributed to cAMP-induced substance P release. Activation of PKA was required for the effects of forskolin and the three cAMP analogs. Epac2 contributed to the effects of forskolin and CPTOMe-cAMP, signaling through a Raf - mitogen-activated protein kinase pathway to activate Ca2+ channels. Epac1 inhibitors induced NK1R internalization independently of substance P release. In rats with latent sensitization to pain, the effect of 6-Bnz-cAMP was unchanged, whereas the effect of forskolin was decreased due to the loss of the stimulatory effect of Epac2. Hence, substance P release induced by cAMP decreases during pain hypersensitivity.
Collapse
Affiliation(s)
- Wenling Chen
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - James A McRoberts
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Helena S Ennes
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Juan Carlos Marvizon
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA.
| |
Collapse
|
38
|
Abstract
Brain-derived neurotrophic factor (BDNF) and the high-affinity receptor tropomyosin receptor kinase B (TrkB) have important roles in neuronal survival and in spinal sensitization mechanisms associated with chronic pain. Recent clinical evidence also supports a peripheral role of BDNF in osteoarthritis (OA), with synovial expression of TrkB associated with higher OA pain. The aim of this study was to use clinical samples and animal models to explore the potential contribution of knee joint BDNF/TrkB signalling to chronic OA pain. Brain-derived neurotrophic factor and TrkB mRNA and protein were present in knee synovia from OA patients (16 women, 14 men, median age 67 years [interquartile range: 61-73]). There was a significant positive correlation between mRNA expression of NTRK2 (TrkB) and the proinflammatory chemokine fractalkine in the OA synovia. Using the surgical medial meniscal transection (MNX) model and the chemical monosodium iodoacetate (MIA) model of OA pain in male rats, the effects of peripheral BDNF injection, vs sequestering endogenous BDNF with TrkB-Fc chimera, on established pain behaviour were determined. Intra-articular injection of BDNF augmented established OA pain behaviour in MIA rats, but had no effect in controls. Intra-articular injection of the TrkB-Fc chimera acutely reversed pain behaviour to a similar extent in both models of OA pain (weight-bearing asymmetry MIA: -11 ± 4%, MNX: -12 ± 4%), compared to vehicle treatment. Our data suggesting a contribution of peripheral knee joint BDNF/TrkB signalling in the maintenance of chronic OA joint pain support further investigation of the therapeutic potential of this target.
Collapse
|
39
|
Ferrini F, Salio C, Boggio EM, Merighi A. Interplay of BDNF and GDNF in the Mature Spinal Somatosensory System and Its Potential Therapeutic Relevance. Curr Neuropharmacol 2021; 19:1225-1245. [PMID: 33200712 PMCID: PMC8719296 DOI: 10.2174/1570159x18666201116143422] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 11/22/2022] Open
Abstract
The growth factors BDNF and GDNF are gaining more and more attention as modulators of synaptic transmission in the mature central nervous system (CNS). The two molecules undergo a regulated secretion in neurons and may be anterogradely transported to terminals where they can positively or negatively modulate fast synaptic transmission. There is today a wide consensus on the role of BDNF as a pro-nociceptive modulator, as the neurotrophin has an important part in the initiation and maintenance of inflammatory, chronic, and/or neuropathic pain at the peripheral and central level. At the spinal level, BDNF intervenes in the regulation of chloride equilibrium potential, decreases the excitatory synaptic drive to inhibitory neurons, with complex changes in GABAergic/glycinergic synaptic transmission, and increases excitatory transmission in the superficial dorsal horn. Differently from BDNF, the role of GDNF still remains to be unraveled in full. This review resumes the current literature on the interplay between BDNF and GDNF in the regulation of nociceptive neurotransmission in the superficial dorsal horn of the spinal cord. We will first discuss the circuitries involved in such a regulation, as well as the reciprocal interactions between the two factors in nociceptive pathways. The development of small molecules specifically targeting BDNF, GDNF and/or downstream effectors is opening new perspectives for investigating these neurotrophic factors as modulators of nociceptive transmission and chronic pain. Therefore, we will finally consider the molecules of (potential) pharmacological relevance for tackling normal and pathological pain.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- Department of Psychiatry & Neuroscience, Université Laval, Québec, Canada
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Elena M. Boggio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- National Institute of Neuroscience, Grugliasco, Italy
| |
Collapse
|
40
|
Tran EL, Crawford LK. Revisiting PNS Plasticity: How Uninjured Sensory Afferents Promote Neuropathic Pain. Front Cell Neurosci 2020; 14:612982. [PMID: 33362476 PMCID: PMC7759741 DOI: 10.3389/fncel.2020.612982] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/12/2020] [Indexed: 11/13/2022] Open
Abstract
Despite the widespread study of how injured nerves contribute to chronic pain, there are still major gaps in our understanding of pain mechanisms. This is particularly true of pain resulting from nerve injury, or neuropathic pain, wherein tactile or thermal stimuli cause painful responses that are particularly difficult to treat with existing therapies. Curiously, this stimulus-driven pain relies upon intact, uninjured sensory neurons that transmit the signals that are ultimately sensed as painful. Studies that interrogate uninjured neurons in search of cell-specific mechanisms have shown that nerve injury alters intact, uninjured neurons resulting in an activity that drives stimulus-evoked pain. This review of neuropathic pain mechanisms summarizes cell-type-specific pathology of uninjured sensory neurons and the sensory ganglia that house their cell bodies. Uninjured neurons have demonstrated a wide range of molecular and neurophysiologic changes, many of which are distinct from those detected in injured neurons. These intriguing findings include expression of pain-associated molecules, neurophysiological changes that underlie increased excitability, and evidence that intercellular signaling within sensory ganglia alters uninjured neurons. In addition to well-supported findings, this review also discusses potential mechanisms that remain poorly understood in the context of nerve injury. This review highlights key questions that will advance our understanding of the plasticity of sensory neuron subpopulations and clarify the role of uninjured neurons in developing anti-pain therapies.
Collapse
Affiliation(s)
- Emily L Tran
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| | - LaTasha K Crawford
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| |
Collapse
|
41
|
Chen W, Marvizón JC. A Src family kinase maintains latent sensitization in rats, a model of inflammatory and neuropathic pain. Brain Res 2020; 1746:146999. [PMID: 32579948 PMCID: PMC10866137 DOI: 10.1016/j.brainres.2020.146999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Latent sensitization is a long-term model of chronic pain in which hyperalgesia is continuously suppressed by opioid receptors, as demonstrated by the induction of mechanical allodynia by opioid antagonists. Different intracellular signals may mediate the initiation, maintenance and expression of latent sensitization. Our criterion for the involvement of a signal in the maintenance of latent sensitization is that inhibitors should permanently eliminate the allodynia produced by an opioid antagonist. We hypothesized that Src family kinases (SFKs) maintain latent sensitization and tested this hypothesis by inducing latent sensitization in rats with complete Freund's adjuvant (CFA) or spared nerve injury. After measures of mechanical allodynia returned to baseline, vehicle or the SFK inhibitor PP2 were injected intrathecally. The opioid antagonist naltrexone injected intrathecally 15 min later produced allodynia in control rats but not in rats injected with PP2. Vehicle or PP2 were injected daily for two more days and naltrexone was injected five days later. Again, naltrexone induced allodynia in the control rats but not in the rats injected with PP2. Results were similar when latent sensitization was induced with CFA or spared nerve injury. We concluded that an SFK, likely Fyn, maintains latent sensitization induced by inflammation or nerve injury.
Collapse
Affiliation(s)
- Wenling Chen
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Juan Carlos Marvizón
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States; Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
42
|
Chen W, Marvizon JC. Neurokinin 1 receptor activation in the rat spinal cord maintains latent sensitization, a model of inflammatory and neuropathic chronic pain. Neuropharmacology 2020; 177:108253. [PMID: 32736088 PMCID: PMC10863619 DOI: 10.1016/j.neuropharm.2020.108253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022]
Abstract
Latent sensitization is a model of chronic pain in which a persistent state of pain hypersensitivity is suppressed by opioid receptors, as evidenced by the ability of opioid antagonists to induce a period of mechanical allodynia. Our objective was to determine if substance P and its neurokinin 1 receptor (NK1R) mediate the maintenance of latent sensitization. Latent sensitization was induced by injecting rats in the hindpaw with complete Freund's adjuvant (CFA), or by tibial spared nerve injury (SNI). When responses to von Frey filaments returned to baseline (day 28), the rats were injected intrathecally with saline or the NK1R antagonist RP67580, followed 15 min later by intrathecal naltrexone. In both pain models, the saline-injected rats developed allodynia for 2 h after naltrexone, but not the RP67580-injected rats. Saline or RP67580 were injected daily for two more days. Five days later (day 35), naltrexone was injected intrathecally. Again, the saline-injected rats, but not the RP67580-injected rats, developed allodynia in response to naltrexone. To determine if there is sustained activation of NK1Rs during latent sensitization, NK1R internalization was measured in lamina I neurons in rats injected in the paw with saline or CFA, and then injected intrathecally with saline or naltrexone on day 28. The rats injected with CFA had a small amount of NK1R internalization that was significantly higher than in the saline-injected rats. Naltrexone increased NK1R internalization in the CFA-injected rats but nor in the saline-injected rats. Therefore, sustained activation of NK1Rs maintains pain hypersensitivity during latent sensitization.
Collapse
Affiliation(s)
- Wenling Chen
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Juan Carlos Marvizon
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA.
| |
Collapse
|
43
|
Cappoli N, Tabolacci E, Aceto P, Dello Russo C. The emerging role of the BDNF-TrkB signaling pathway in the modulation of pain perception. J Neuroimmunol 2020; 349:577406. [PMID: 33002723 DOI: 10.1016/j.jneuroim.2020.577406] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
The brain derived neurotrophic factor (BDNF) is a crucial neuromodulator in pain transmission both in peripheral and central nervous system (CNS). Despite evidence of a pro-nociceptive role of BDNF, recent studies have reported contrasting results, including anti-nociceptive and anti-inflammatory activities. Moreover, BDNF polymorphisms can interfere with BDNF role in pain perception. In Val66Met carriers, the Met allele may have a dual role, with anti-nociceptive actions in normal condition and pro-nociceptive effects during chronic pain. In order to elucidate the main effects of BDNF in nociception, we reviewed the main characteristics of this neurotrophin, focusing on its involvement in pain.
Collapse
Affiliation(s)
- Natalia Cappoli
- Università Cattolica del Sacro Cuore, Dipartimento di Sicurezza e Bioetica, Sezione di Farmacologia, Rome, Italy
| | - Elisabetta Tabolacci
- Università Cattolica del Sacro Cuore, Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Aceto
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Dipartimento di Scienze dell'Emergenza, Anestesiologiche e della Rianimazione, Rome, Italy; Università Cattolica del Sacro Cuore, Dipartimento di Scienze biotecnologiche di base, cliniche intensivologiche e perioperatorie, Rome, Italy.
| | - Cinzia Dello Russo
- Università Cattolica del Sacro Cuore, Dipartimento di Sicurezza e Bioetica, Sezione di Farmacologia, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
44
|
Mitchell R, Mikolajczak M, Kersten C, Fleetwood-Walker S. ErbB1-dependent signalling and vesicular trafficking in primary afferent nociceptors associated with hypersensitivity in neuropathic pain. Neurobiol Dis 2020; 142:104961. [DOI: 10.1016/j.nbd.2020.104961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/26/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
|
45
|
Ge MM, Zhou YQ, Tian XB, Manyande A, Tian YK, Ye DW, Yang H. Src-family protein tyrosine kinases: A promising target for treating chronic pain. Biomed Pharmacother 2020; 125:110017. [DOI: 10.1016/j.biopha.2020.110017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022] Open
|
46
|
Cellular mechanisms and molecular signaling pathways in stress-induced anxiety, depression, and blood-brain barrier inflammation and leakage. Inflammopharmacology 2020; 28:643-665. [PMID: 32333258 DOI: 10.1007/s10787-020-00712-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022]
Abstract
Depression and anxiety are comorbid conditions in many neurological or psychopathological disorders. Stress is an underlying event that triggers development of anxiety and depressive-like behaviors. Recent experimental data indicate that anxiety and depressive-like behaviors occurring as a result of stressful situations can cause blood-brain barrier (BBB) dysfunction, which is characterized by inflammation and leakage. However, the underlying mechanisms are not completely understood. This paper sought to review recent experimental preclinical and clinical data that suggest possible molecular mechanisms involved in development of stress-induced anxiety and depression with associated BBB inflammation and leakage. Critical therapeutic targets and potential pharmacological candidates for treatment of stress-induced anxiety and depression with associated BBB dysfunctions are also discussed.
Collapse
|
47
|
Shinoda M, Hayashi Y, Kubo A, Iwata K. Pathophysiological mechanisms of persistent orofacial pain. J Oral Sci 2020; 62:131-135. [PMID: 32132329 DOI: 10.2334/josnusd.19-0373] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Nociceptive stimuli to the orofacial region are typically received by the peripheral terminal of trigeminal ganglion (TG) neurons, and noxious orofacial information is subsequently conveyed to the trigeminal spinal subnucleus caudalis and the upper cervical spinal cord (C1-C2). This information is further transmitted to the cortical somatosensory regions and limbic system via the thalamus, which then leads to the perception of pain. It is a well-established fact that the presence of abnormal pain in the orofacial region is etiologically associated with neuroplastic changes that may occur at any point in the pain transmission pathway from the peripheral to the central nervous system (CNS). Recently, several studies have reported that functional plastic changes in a large number of cells, including TG neurons, glial cells (satellite cells, microglia, and astrocytes), and immune cells (macrophages and neutrophils), contribute to the sensitization and disinhibition of neurons in the peripheral and CNS, which results in orofacial pain hypersensitivity.
Collapse
Affiliation(s)
| | | | - Asako Kubo
- Department of Physiology, Nihon University School of Dentistry
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry
| |
Collapse
|
48
|
Meng X, Jin X, Wei X, Wang L, Yang J, Ji F. Low‑affinity neurotrophin receptor p75 of brain‑derived neurotrophic factor contributes to cancer‑induced bone pain by upregulating mTOR signaling. Exp Ther Med 2019; 18:4379-4387. [PMID: 31777542 PMCID: PMC6862244 DOI: 10.3892/etm.2019.8097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/04/2019] [Indexed: 11/06/2022] Open
Abstract
Crucial to the development and maintenance of pain sensations is neurotrophin receptor p75 (p75NTR), the low affinity receptor of brain-derived neurotrophic factor (BDNF). This receptor is widespread among dorsal root ganglion (DRG) neurons and the spinal cord. Few reports have demonstrated the specific role of p75NTR in the development of cancer-induced bone pain (CIBP). Therefore the present study examined whether p75NTR contributed to CIBP by upregulating mammalian target of rapamycin (mTOR) signaling. A CIBP rat model was induced and reverse transcription-quantitative polymerase chain reaction was employed to determine p75NTR and mTOR mRNA expression. Immunofluorescence analysis was performed to determine the coexpression of p75NTR and mTOR in DRG neurons, as well as the spinal cord. Von Frey filaments were used to measure the 50% likelihood of paw withdrawal thresholds (PWTs). Spontaneous pain was assessed by ambulatory score. The results demonstrated that compared with the control group, mTOR activation in primary cultured DRG neurons was significantly increased. In addition, mTOR and p75NTR expression was significantly enhanced in the BDNF-treated primary DRG in the BDNF group. In vivo experiments determined that mTOR and p75NTR levels were increased in the CIBP rats compared with the sham group. PWT, in response to mechanical stimulation, was significantly lower compared with that in sham rats and the ambulatory score was significantly higher than that in sham rats. Finally, intrathecal injection of a p75NTR-targeting small interfering RNA significantly decreased mTOR and p75NTR expression levels in DRG neurons and the spinal cord of CIBP rats, as well as partially reversing the decline in PWTs and the increase in ambulatory score. In conclusion, the present study determined that the activation of BDNF/p75NTR/mTOR signaling may participate in nociceptive transmission in CIBP, suggesting a novel mechanism and potential therapeutic target for CIBP treatment and management.
Collapse
Affiliation(s)
- Xiao‑Wen Meng
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xiao‑Hong Jin
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xiang Wei
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Li‑Na Wang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jian‑Ping Yang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Fu‑Hai Ji
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
49
|
Price TJ, Gold MS. From Mechanism to Cure: Renewing the Goal to Eliminate the Disease of Pain. PAIN MEDICINE 2019; 19:1525-1549. [PMID: 29077871 DOI: 10.1093/pm/pnx108] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective Persistent pain causes untold misery worldwide and is a leading cause of disability. Despite its astonishing prevalence, pain is undertreated, at least in part because existing therapeutics are ineffective or cause intolerable side effects. In this review, we cover new findings about the neurobiology of pain and argue that all but the most transient forms of pain needed to avoid tissue damage should be approached as a disease where a cure can be the goal of all treatment plans, even if attaining this goal is not yet always possible. Design We reviewed the literature to highlight recent advances in the area of the neurobiology of pain. Results We discuss barriers that are currently hindering the achievement of this goal, as well as the development of new therapeutic strategies. We also discuss innovations in the field that are creating new opportunities to treat and even reverse persistent pain, some of which are in late-phase clinical trials. Conclusion We conclude that the confluence of new basic science discoveries and development of new technologies are creating a path toward pain therapeutics that should offer significant hope of a cure for patients and practitioners alike. Classification of Evidence. Our review points to new areas of inquiry for the pain field to advance the goal of developing new therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, Texas
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
50
|
Marvizon JC, Chen W, Fu W, Taylor BK. Neuropeptide Y release in the rat spinal cord measured with Y1 receptor internalization is increased after nerve injury. Neuropharmacology 2019; 158:107732. [PMID: 31377198 DOI: 10.1016/j.neuropharm.2019.107732] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
Abstract
Neuropeptide Y (NPY) modulates nociception in the spinal cord, but little is known about its mechanisms of release. We measured NPY release in situ using the internalization of its Y1 receptor in dorsal horn neurons. Y1 receptor immunoreactivity was normally localized to the cell surface, but addition of NPY to spinal cord slices increased the number of neurons with Y1 internalization in a biphasic fashion (EC50s of 1 nM and 1 μM). Depolarization with KCl, capsaicin, or the protein kinase A activator 6-benzoyl-cAMP also induced Y1 receptor internalization, presumably by releasing NPY. NMDA receptor activation in the presence of BVT948, an inhibitor of protein tyrosine phosphatases, also released NPY. Electrical stimulation of the dorsal horn frequency-dependently induced NPY release; and this was decreased by the Y1 antagonist BIBO3304, the Nav channel blocker lidocaine, or the Cav2 channel blocker ω-conotoxin MVIIC. Dorsal root immersion in capsaicin, but not its electrical stimulation, also induced NPY release. This was blocked by CNQX, suggesting that part of the NPY released by capsaicin was from dorsal horn neurons receiving synapses from primary afferents and not from the afferent themselves. Mechanical stimulation in vivo, with rub or clamp of the hindpaw, elicited robust Y1 receptor internalization in rats with spared nerve injury but not sham surgery. In summary, NPY is released from dorsal horn interneurons or primary afferent terminals by electrical stimulation and by activation of TRPV1, PKA or NMDA receptors in. Furthermore, NPY release evoked by noxious and tactile stimuli increases after peripheral nerve injury.
Collapse
Affiliation(s)
- Juan Carlos Marvizon
- Vatche and Tamar Manoukian Division of Digestive Diseases, 900 Veterans Ave., Warren Hall Building, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, 11310 Wilshire Blvd., Building 115, Los Angeles, CA, 90073, USA.
| | - Wenling Chen
- Vatche and Tamar Manoukian Division of Digestive Diseases, 900 Veterans Ave., Warren Hall Building, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, 90095, USA; Veteran Affairs Greater Los Angeles Healthcare System, 11310 Wilshire Blvd., Building 115, Los Angeles, CA, 90073, USA.
| | - Weisi Fu
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY, USA.
| | - Bradley K Taylor
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY, USA; Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research and the Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|