1
|
Leite CBG, Fricke HP, Tavares LP, Nshimiyimana R, Mekhail J, Kilgallen E, Killick F, Whalen JD, Lehoczky JA, Serhan CN, Charles JF, Lattermann C. Maresin 1-LGR6 axis mitigates inflammation and posttraumatic osteoarthritis after transection of the anterior cruciate ligament in mice. Osteoarthritis Cartilage 2025:S1063-4584(25)00869-6. [PMID: 40139646 DOI: 10.1016/j.joca.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Anterior cruciate ligament (ACL) tears frequently cause chronic inflammation and posttraumatic osteoarthritis (PTOA), with therapies failing to resolve persistent post-injury inflammation. Specialized pro-resolving mediators (SPMs), including Maresin1 (MaR1), show promise in resolving inflammation and promoting tissue repair. However, their role in PTOA remains underexplored. This study investigated inflammatory markers and MaR1 dynamics post-ACL injury, the role of the MaR1 receptor Leucine-rich Repeat-containing G protein-coupled receptor 6 (LGR6) in PTOA, and MaR1's therapeutic potential in a mouse ACL transection (ACLT) model. DESIGN Eight-week-old C57BL6/J male mice underwent ACLT, and synovial fluid, periarticular tissue, and tibiofemoral joints were collected at various time points post-surgery for analysis. LGR6-deficient mice were utilized to investigate the role of MaR1 signaling in inflammation resolution. Additionally, the effect of intraarticular MaR1 administration on PTOA progression was assessed. RESULTS ACLT induced joint inflammation with leukocyte infiltration and elevated pro-inflammatory cytokines. MaR1 levels peaked early post-injury and were associated with a six-fold increase in LGR6 expression. LGR6 deficiency worsened inflammation and PTOA severity with higher histological Osteoarthritis Research Society International (OARSI) scores (mean difference 5.6[95%CI: 2.5-8.6], p<0.001) and microCT OA severity scores (mean difference 4.3[95%CI: 0.7-7.9], p=0.018). Intraarticular MaR1 treatment reduced leukocyte recruitment, suppressed pro-inflammatory gene expression, and ameliorated PTOA development, improving histological OARSI scores (mean difference -3.9[95%CI: -6.9 to -1.0], p=0.012) and microCT scores (mean difference -6.7[95%CI: -10.3 to -3.0], p=0.012). CONCLUSION This study suggests a critical role of MaR1 in resolving inflammation post-ACL injury and mitigating PTOA in mice. Targeting SPM pathways, particularly MaR1 and/or MaR1 mimetics, offers a promising strategy to prevent chronic joint inflammation and degeneration after ACL injury.
Collapse
Affiliation(s)
- Chilan B G Leite
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Hannah P Fricke
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Luciana P Tavares
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Robert Nshimiyimana
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Julie Mekhail
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Elliott Kilgallen
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Felix Killick
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Janey D Whalen
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Jessica A Lehoczky
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Julia F Charles
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Christian Lattermann
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Oggero S, Voisin MB, Picco F, Huerta MÁ, Cecconello C, Burgoyne T, Perretti M, Malcangio M. Activation of proresolving macrophages in dorsal root ganglia attenuates persistent arthritis pain. Proc Natl Acad Sci U S A 2025; 122:e2416343122. [PMID: 40063821 PMCID: PMC11929478 DOI: 10.1073/pnas.2416343122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025] Open
Abstract
Pain independent of disease activity is frequently reported by rheumatoid arthritis patients and remains undertreated. Preclinical evidence suggests that imbalance of neuroimmune proresolving interactions within dorsal root ganglia (DRG) rather than at the site of inflammation plays mechanistic roles in persistent arthritis pain. Here, we inhibited production of proresolving lipid mediators by silencing 12/15-lipoxygenase expression in CX3CR1+ monocyte/macrophages conditional knockout (cKO) mice. In an arthritis model, hind paw mechanical hypersensitivity is exacerbated in male and female cKO mice in association with DRG infiltration of neutrophils, which migrate in response to leukotriene B4 released by macrophages through 5-lipoxygenase conversion of arachidonic acid provided by neuron-derived vesicles. Neutrophils apoptosis promotes primary macrophage efferocytosis which is defective in cKO macrophages. In wild-type (WT) and cKO mice, intrathecal injection of MerTK activating antibody, attenuates persistent hypersensitivity and polarizes DRG macrophages toward a proresolving phenotype with production of antinociceptive lipoxin A4. Thus, we delineate a neuron-macrophage-neutrophil bidirectional circuit that can be exploited to reduce persistent arthritis pain.
Collapse
Affiliation(s)
- Silvia Oggero
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
| | - Mathieu-Benoit Voisin
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, LondonEC1M 6BQ, United Kingdom
| | - Francesca Picco
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
| | - Miguel Á. Huerta
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
- Department of Pharmacology, University of Granada, Granada18016, Spain
| | - Chiara Cecconello
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, LondonEC1M 6BQ, United Kingdom
| | - Thomas Burgoyne
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, LondonEC1V 9EL, United Kingdom
- Pediatric Respiratory Medicine, Royal Brompton Hospital, Guy’s and St Thomas’ National Health System Foundation Trust, LondonSW3 6NP, United Kingdom
| | - Mauro Perretti
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, LondonEC1M 6BQ, United Kingdom
| | - Marzia Malcangio
- Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, Guys’ Campus, LondonSE1 1UL, United Kingdom
| |
Collapse
|
3
|
Zhang Q, Wang Y, Zhu J, Zou M, Zhang Y, Wu H, Jin T. Specialized pro-resolving lipid mediators: a key player in resolving inflammation in autoimmune diseases. Sci Bull (Beijing) 2025; 70:778-794. [PMID: 39837719 DOI: 10.1016/j.scib.2024.07.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/21/2024] [Accepted: 07/16/2024] [Indexed: 01/23/2025]
Abstract
Uncontrolled hyperactivation of the immune system is the central mechanism underlying the pathogenesis of autoimmune diseases. Timely control of the inflammatory response is essential to prevent inflammation progression and organ damage. Specialized pro-resolving lipid mediators (SPMs) are autacoid molecules derived from essential polyunsaturated fatty acids during acute inflammatory responses. They promote the resolution of inflammation and orchestrate endogenous reparative responses. The SPM superfamily includes lipoxins, resolvins, protectins, and maresins, as well as novel conjugates involved in tissue regeneration. Much work has been done focusing on the actions of SPMs in autoimmunity and has identified their deficiencies and therapeutic effects in autoimmune diseases. In this review, we provide a brief introduction of SPMs, summarize their effects on key cells involved in innate and adaptive immunity, and highlight their role and therapeutic potential in autoimmune diseases.
Collapse
Affiliation(s)
- Qingxiang Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Ying Wang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm 17176, Sweden
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
4
|
Shih YRV, Tao H, Gilpin A, Lee YW, Perikamana SM, Varghese S. Specialized pro-resolving mediator Maresin 1 attenuates pain in a mouse model of osteoarthritis. Osteoarthritis Cartilage 2025; 33:341-350. [PMID: 39617202 PMCID: PMC11842212 DOI: 10.1016/j.joca.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVE We test whether the specialized pro-resolving molecule Maresin 1 (MaR1) attenuates nociceptive behaviors in mice with osteoarthritis-like pain. DESIGN Osteoarthritis (OA)-like pain behavior was induced by intra-articular injection of monosodium iodoacetate (MIA) and treated with MaR1 (N=6) or vehicle (N=5) by intraperitoneal injection 8 weeks after injury. Mice without MIA injection were used as control (N=6). Nociceptive behaviors were examined by von Frey and dynamic weight bearing measurements. Calcitonin gene-related peptide (CGRP) expression and activated macrophages in the dorsal root ganglion (DRG) were examined by immunofluorescence staining. The inflammatory profile in circulation was assessed by cytokine array. Calcium imaging was performed to assess the in vitro functional response of DRG neurons from animals with OA-like pain behavior to MaR1 with or without RAR Related Orphan Receptor A (RORA) inverse agonist SR3335. RESULTS MaR1 attenuated knee pain behavior in treated mice (N=6) compared to non-treated mice (N=5) as shown by increased paw withdrawal threshold with a mean difference of 112.2% (95% CI [49.79, 174.6], p=0.0784) at 4 h and 150.9% (95% CI [104.2, 197.5], p=0.0001) at 4 days post-MaR1 treatment, and increased weight bearing with a mean difference of 20.08% (95% CI [2.798, 37.37], p=0.0277) at 1 day post-MaR1 treatment. CGRP expression and activated macrophages were decreased in the DRG, and inflammatory cytokine levels in the circulation were attenuated. Calcium imaging showed MaR1 reduced the functional response of DRG neurons through RORA. CONCLUSIONS Our results show that MaR1 reduces OA-like pain behavior in mice and could be a potential treatment for OA pain.
Collapse
MESH Headings
- Animals
- Docosahexaenoic Acids/pharmacology
- Docosahexaenoic Acids/therapeutic use
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/drug effects
- Mice
- Disease Models, Animal
- Calcitonin Gene-Related Peptide/metabolism
- Calcitonin Gene-Related Peptide/drug effects
- Male
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/metabolism
- Osteoarthritis, Knee/drug therapy
- Osteoarthritis, Knee/metabolism
- Macrophages/metabolism
- Macrophages/drug effects
- Pain Measurement
- Osteoarthritis/drug therapy
- Behavior, Animal/drug effects
- Injections, Intra-Articular
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Yu-Ru V Shih
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Huchen Tao
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA.
| | - Anna Gilpin
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| | - Yuan-Wen Lee
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei 11031, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | | | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
5
|
Ferreira MV, Jesus CHA, Bonfim da Costa JP, Oliveira G, Liebl B, Verri Junior W, Zanoveli JM, Cunha JMD. Aspirin-triggered lipoxin A4 reduces neuropathic pain and anxiety-like behaviours in male diabetic rats: antinociceptive enhancement by cannabinoid receptor agonists. Eur J Pharmacol 2025; 989:177254. [PMID: 39788405 DOI: 10.1016/j.ejphar.2025.177254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
Neuropathy is the most common complication of diabetes, leading to painful symptoms like hyperalgesia. Current treatments for diabetic painful neuropathy often prove inadequate, necessitating the exploration of new pharmacological approaches. Therefore, this study aimed to investigate the potential antinociceptive effect of aspirin-triggered lipoxin A4 (ATL), a specialized pro-resolving lipid mediator, when administered alone or in combination with cannabinoid agonists, to alleviate diabetic neuropathic pain. Mechanical hyperalgesia in the hindpaws of streptozotocin (STZ)-induced diabetic (DBT) rats was assessed using the electronic Von Frey test (VFT), before diabetes induction and for up to 32 days after STZ administration and intraperitoneal ATL (0.3, 1, 3, 10, or 30 ng/rat) treatment, alone or in combination with intrathecal CB1 or CB2 receptor agonists (ACEA or JWH-133, respectively; 10 or 30 μg/rat). The effect of ATL treatment on locomotor activity and anxious or depressive-like behaviors was also evaluated. In comparison to control normoglycemic rats, control DBT rats developed: 1) mechanical hyperalgesia; 2) increase in anxious and depressive-like behaviors. ATL treatment attenuated mechanical hyperalgesia in DBT rats both acutely (at 30 ng) and cumulatively (at doses of 1, 3, 10, or 30 ng), without compromising locomotor activity. The antinociceptive effect of ATL (at 1 or 3 ng) was augmented when combined with ACEA or JWH-133 treatments (only at a dose of 30 μg/rat). While ATL treatment alone reduced anxious-like behavior in DBT rats, it did not affect depressive-like behavior. These findings underscore the therapeutic potential of ATL, in diabetic complications, suggesting a possible interaction with the endocannabinoid system.
Collapse
Affiliation(s)
- Matheus Vinícius Ferreira
- Laboratory of Pharmacology of Pain, Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil.
| | | | | | - Gabrielle Oliveira
- Laboratory of Pharmacology of Pain, Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Bruno Liebl
- Laboratory of Pharmacology of Pain, Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Waldiceu Verri Junior
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Paraná, Brazil
| | - Janaína Menezes Zanoveli
- Laboratory of Pharmacology of Pain, Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Joice Maria da Cunha
- Laboratory of Pharmacology of Pain, Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| |
Collapse
|
6
|
Thomas BL, Montero‐Melendez T, Oggero S, Kaneva MK, Chambers D, Pinto AL, Nerviani A, Lucchesi D, Austin‐Williams S, Hussain MT, Pitzalis C, Allen B, Malcangio M, Dell'Accio F, Norling LV, Perretti M. Molecular Determinants of Neutrophil Extracellular Vesicles That Drive Cartilage Regeneration in Inflammatory Arthritis. Arthritis Rheumatol 2024; 76:1705-1718. [PMID: 39041647 PMCID: PMC11605269 DOI: 10.1002/art.42958] [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: 12/16/2022] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
OBJECTIVE This study was undertaken to establish the potential therapeutic profile of neutrophil-derived extracellular vesicles (EVs) in experimental inflammatory arthritis and associate pharmacological activity with specific EV components, focusing on microRNAs. METHODS Neutrophil EVs were administered intra-articularly through a prophylactic or therapeutic protocol to male C57BL/6 mice undergoing serum-transfer-induced inflammatory arthritis. Transcriptomic analysis of knees was performed on joints following EV administration, naive and arthritic mice (untreated; n = 4/group) and EV-treated diseased mice (intra-articular administration) with contralateral (vehicle-treated; n = 8/group). Comparison of healthy donor and patients with rheumatoid arthritis (RA) neutrophil EVs was performed. RESULTS EVs afforded cartilage protection with an increase in collagen-II and reduced collagen-X expression within the joint. To gain mechanistic insights, RNA sequencing of the arthritic joints was conducted. A total of 5,231 genes were differentially expressed (P < 0.05), with 257 unique to EV treatment. EVs affected key regenerative pathways involved in joint development, including Wnt and Notch signaling. This wealth of genomic alteration prompted to identify microRNAs in EVs, 10 of which are associated with RA. As a proof of concept, we focused on miR-455-3p, which was detected in both healthy donor and RA EVs. EV addition to chondrocyte cultures elevated miR-455-3p and exerted anticatabolic effects upon interleukin-1β stimulation; these effects were blocked by actinomycin or miR-455-3p antagomir. CONCLUSION Neutrophils from patients with RA yielded EVs with composition, efficacy, and miR-455-3p content similar to those of healthy volunteers, suggesting that neutrophil EVs could be developed as an autologous treatment to protect and repair joint tissue of patients affected by inflammatory arthritides.
Collapse
Affiliation(s)
| | | | - Silvia Oggero
- Queen Mary University of London and Kings College London, Guys’ CampusLondonUnited Kingdom
| | | | | | - Andreia L. Pinto
- Royal Brompton & Harefield NHS Foundation TrustLondonUnited Kingdom
| | - Alessandra Nerviani
- Queen Mary University of London, Barts Health NHS Trust, and National Institute for Health and Care Research Barts Biomedical Research CentreLondonUnited Kingdom
| | | | | | | | - Costantino Pitzalis
- Queen Mary University of London, Barts Health NHS Trust, and National Institute for Health and Care Research Barts Biomedical Research CentreLondonUnited Kingdom
| | | | | | - Francesco Dell'Accio
- Queen Mary University of London, Barts Health NHS Trust, and National Institute for Health and Care Research Barts Biomedical Research CentreLondonUnited Kingdom
| | | | | |
Collapse
|
7
|
Maliha A, Tahsin M, Fabia TZ, Rahman SM, Rahman MM. Pro-resolving metabolites: Future of the fish oil supplements. J Funct Foods 2024; 121:106439. [DOI: 10.1016/j.jff.2024.106439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
8
|
Gusakov K, Kalinkovich A, Ashkenazi S, Livshits G. Nature of the Association between Rheumatoid Arthritis and Cervical Cancer and Its Potential Therapeutic Implications. Nutrients 2024; 16:2569. [PMID: 39125448 PMCID: PMC11314534 DOI: 10.3390/nu16152569] [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: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
It is now established that patients with rheumatoid arthritis (RA) have an increased risk of developing cervical cancer (CC) or its precursor, cervical intraepithelial neoplasia (CIN). However, the underlying mechanisms of this association have not been elucidated. RA is characterized by unresolved chronic inflammation. It is suggested that human papillomavirus (HPV) infection in RA patients exacerbates inflammation, increasing the risk of CC. The tumor microenvironment in RA patients with CC is also marked by chronic inflammation, which aggravates the manifestations of both conditions. Gut and vaginal dysbiosis are also considered potential mechanisms that contribute to the chronic inflammation and aggravation of RA and CC manifestations. Numerous clinical and pre-clinical studies have demonstrated the beneficial effects of various nutritional approaches to attenuate chronic inflammation, including polyunsaturated fatty acids and their derivatives, specialized pro-resolving mediators (SPMs), probiotics, prebiotics, and certain diets. We believe that successful resolution of chronic inflammation and correction of dysbiosis, in combination with current anti-RA and anti-CC therapies, is a promising therapeutic approach for RA and CC. This approach could also reduce the risk of CC development in HPV-infected RA patients.
Collapse
Affiliation(s)
- Kirill Gusakov
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel; (K.G.); (S.A.)
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv 6905126, Israel;
| | - Shai Ashkenazi
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel; (K.G.); (S.A.)
| | - Gregory Livshits
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel; (K.G.); (S.A.)
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv 6905126, Israel;
| |
Collapse
|
9
|
Nakhleh-Francis Y, Awad-Igbaria Y, Sakas R, Bang S, Abu-Ata S, Palzur E, Lowenstein L, Bornstein J. Exploring Localized Provoked Vulvodynia: Insights from Animal Model Research. Int J Mol Sci 2024; 25:4261. [PMID: 38673846 PMCID: PMC11050705 DOI: 10.3390/ijms25084261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Provoked vulvodynia represents a challenging chronic pain condition, characterized by its multifactorial origins. The inherent complexities of human-based studies have necessitated the use of animal models to enrich our understanding of vulvodynia's pathophysiology. This review aims to provide an exhaustive examination of the various animal models employed in this research domain. A comprehensive search was conducted on PubMed, utilizing keywords such as "vulvodynia", "chronic vulvar pain", "vulvodynia induction", and "animal models of vulvodynia" to identify pertinent studies. The search yielded three primary animal models for vulvodynia: inflammation-induced, allergy-induced, and hormone-induced. Additionally, six agents capable of triggering the condition through diverse pathways were identified, including factors contributing to hyperinnervation, mast cell proliferation, involvement of other immune cells, inflammatory cytokines, and neurotransmitters. This review systematically outlines the various animal models developed to study the pathogenesis of provoked vulvodynia. Understanding these models is crucial for the exploration of preventative measures, the development of novel treatments, and the overall advancement of research within the field.
Collapse
Affiliation(s)
- Yara Nakhleh-Francis
- Department of Obstetrics and Gynecology, Galilee Medical Center, Nahariya 2210001, Israel; (S.B.); (L.L.); (J.B.)
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Yaseen Awad-Igbaria
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Reem Sakas
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Sarina Bang
- Department of Obstetrics and Gynecology, Galilee Medical Center, Nahariya 2210001, Israel; (S.B.); (L.L.); (J.B.)
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Saher Abu-Ata
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Eilam Palzur
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Lior Lowenstein
- Department of Obstetrics and Gynecology, Galilee Medical Center, Nahariya 2210001, Israel; (S.B.); (L.L.); (J.B.)
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Jacob Bornstein
- Department of Obstetrics and Gynecology, Galilee Medical Center, Nahariya 2210001, Israel; (S.B.); (L.L.); (J.B.)
- Research Institute of Galilee Medical Center, Nahariya 2210001, Israel; (Y.A.-I.); (R.S.); (S.A.-A.); (E.P.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
10
|
Bekauri T, Fischer S, Honn KV, Maddipati KR, Love T, Little C, Wood RW, Bonham AD, Linder MA, Yule DI, Emanuelle C, Falsetta ML. Inflammation, lipid dysregulation, and transient receptor potential cation channel subfamily V member 4 signaling perpetuate chronic vulvar pain. Pain 2024; 165:820-837. [PMID: 37889581 PMCID: PMC10949218 DOI: 10.1097/j.pain.0000000000003088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/19/2023] [Accepted: 08/23/2023] [Indexed: 10/29/2023]
Abstract
ABSTRACT Localized provoked vulvodynia is characterized by chronic vulvar pain that disrupts every aspect of the patient's life. Pain is localized to the vulvar vestibule, a specialized ring of tissue immediately surrounding the vaginal opening involved in immune defense. In this article, we show inflammation is the critical first step necessary for the generation of pain signals in the vulva. Inflammatory stimuli alone or combined with the transient receptor potential cation channel subfamily V member 4 (TRPV4) agonist 4α-phorbol 12,13-didecanoate stimulate calcium flux into vulvar fibroblast cells. Activity is blocked by the TRPV4 antagonist HC067047, denoting specificity to TRPV4. Using lipidomics, we found pro-resolving lipids in the vulvar vestibule were dysregulated, characterized by a reduction in pro-resolving mediators and heightened production of inflammatory mediators. We demonstrate specialized pro-resolving mediators represent a potential new therapy for vulvar pain, acting on 2 key parts of the disease mechanism by limiting inflammation and acutely inhibiting TRPV4 signaling.
Collapse
Affiliation(s)
- Tamari Bekauri
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Sarah Fischer
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Kenneth V. Honn
- Pathology Department, Wayne State University, Detroit, MI, United States
- Lipidomics Core Facility and Bioactive Lipids Research Program, Wayne State University, Detroit, MI, United States
| | - Krishna Rao Maddipati
- Pathology Department, Wayne State University, Detroit, MI, United States
- Lipidomics Core Facility and Bioactive Lipids Research Program, Wayne State University, Detroit, MI, United States
| | - Tanzy Love
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Chantelle Little
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Ronald W. Wood
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Adrienne D. Bonham
- OB/GYN Department, Oregon Health Sciences University, Portland, OR, United States
| | - Mitchell A. Linder
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - David I. Yule
- Pharmacology and Physiology Department, University of Rochester, Rochester, NY, United States
| | - Chrysilla Emanuelle
- Pharmacology and Physiology Department, University of Rochester, Rochester, NY, United States
| | - Megan L. Falsetta
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
- Pharmacology and Physiology Department, University of Rochester, Rochester, NY, United States
| |
Collapse
|
11
|
Bang S, Jiang C, Xu J, Chandra S, McGinnis A, Luo X, He Q, Li Y, Wang Z, Ao X, Parisien M, Fernandes de Araujo LO, Jahangiri Esfahani S, Zhang Q, Tonello R, Berta T, Diatchenko L, Ji RR. Satellite glial GPR37L1 and its ligand maresin 1 regulate potassium channel signaling and pain homeostasis. J Clin Invest 2024; 134:e173537. [PMID: 38530364 PMCID: PMC11060744 DOI: 10.1172/jci173537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
G protein-coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR with largely unknown functions. Here, we report that Gpr37l1/GRP37L1 ranks among the most highly expressed GPCR transcripts in mouse and human dorsal root ganglia (DRGs) and is selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy induced by streptozotoxin (STZ) and paclitaxel (PTX) led to reduced GPR37L1 expression on the plasma membrane in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptoms following PTX- and STZ-induced pain, whereas overexpression of Gpr37l1 in mouse DRGs reversed pain. GPR37L1 is coexpressed with potassium channels, including KCNJ10 (Kir4.1) in mouse SGCs and both KCNJ3 (Kir3.1) and KCNJ10 in human SGCs. GPR37L1 regulates the surface expression and function of the potassium channels. Notably, the proresolving lipid mediator maresin 1 (MaR1) serves as a ligand of GPR37L1 and enhances KCNJ10- or KCNJ3-mediated potassium influx in SGCs through GPR37L1. Chemotherapy suppressed KCNJ10 expression and function in SGCs, which MaR1 rescued through GPR37L1. Finally, genetic analysis revealed that the GPR37L1-E296K variant increased chronic pain risk by destabilizing the protein and impairing the protein's function. Thus, GPR37L1 in SGCs offers a therapeutic target for the protection of neuropathy and chronic pain.
Collapse
Affiliation(s)
- Sangsu Bang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sharat Chandra
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Qianru He
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yize Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Zilong Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xiang Ao
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Marc Parisien
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Lorenna Oliveira Fernandes de Araujo
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Sahel Jahangiri Esfahani
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Qin Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Luda Diatchenko
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurobiology and
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
12
|
Natami M, Hosseini SM, Khaleel RA, Addulrahman TS, Zarei M, Asadi S, Gholami S, Mehrvar A. The role of specialized pro-resolving mediators (SPMs) in inflammatory arthritis: A therapeutic strategy. Prostaglandins Other Lipid Mediat 2024; 170:106798. [PMID: 37977352 DOI: 10.1016/j.prostaglandins.2023.106798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/28/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Rheumatoid arthritis (RA) is classified as a persistent inflammatory autoimmune disorder leading to the subsequent erosion of articular cartilage and bone tissue originating from the synovium. The fundamental objective of therapeutic interventions in RA has been the suppression of inflammation. Nevertheless, conventional medicines that lack target specificity may exhibit unpredictable effects on cell metabolism. In recent times, there has been evidence suggesting that specialized pro-resolving mediators (SPMs), which are lipid metabolites, have a role in facilitating the resolution of inflammation and the reestablishment of tissue homeostasis. SPMs are synthesized by immune cells through the enzymatic conversion of omega-3 fatty acids. In the context of RA, there is a possibility of dysregulation in the production of these SPMs. In this review, we delve into the present comprehension of the endogenous functions of SPMs in RA as lipids that exhibit pro-resolutive, protective, and immunoresolvent properties.
Collapse
Affiliation(s)
- Mohammad Natami
- Department of Urology, Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Seyed Mehdi Hosseini
- Department of Oral and Maxillofacial surgery, School of Dentistry, Azad University of Medical Science, Shiraz, Iran
| | | | | | - Mehdi Zarei
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Sahar Asadi
- Department of Community and Family Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sepideh Gholami
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Amir Mehrvar
- Taleghani Hospital Clinical Research Development Unit, Shahid Beheshti University of Medical Science, Tehran, Iran.
| |
Collapse
|
13
|
Bang S, Jiang C, Xu J, Chandra S, McGinnis A, Luo X, He Q, Li Y, Wang Z, Ao X, Parisien M, Fernandes de Araujo LO, Esfahan SJ, Zhang Q, Tonello R, Berta T, Diatchenko L, Ji RR. Satellite glial GPR37L1 regulates maresin and potassium channel signaling for pain control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569787. [PMID: 38106084 PMCID: PMC10723316 DOI: 10.1101/2023.12.03.569787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
G protein coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR and its function remains largely unknown. Here we report that GPR37L1 transcript is highly expressed compared to all known GPCRs in mouse and human dorsal root ganglia (DRGs) and selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy following diabetes and chemotherapy by streptozotocin and paclitaxel resulted in downregulations of surface GPR37L1 in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptom (mechanical allodynia), whereas overexpression of Gpr37l1 in mouse DRGs can reverse neuropathic pain. Notably, GPR37L1 is co-expressed and coupled with potassium channels in SGCs. We found striking species differences in potassium channel expression in SGCs, with predominant expression of KCNJ10 and KCNJ3 in mouse and human SGCs, respectively. GPR37L1 regulates the surface expression and function of KCNJ10 and KCNJ3. We identified the pro-resolving lipid mediator maresin 1 (MaR1) as a GPR37L1 ligand. MaR1 increases KCNJ10/KCNJ3-mediated potassium influx in SGCs via GPR37L1. MaR1 protected chemotherapy-induced suppression of KCNJ13/KCNJ10 expression and function in SGCs. Finally, genetic analysis revealed that the GPR37L1-E296K variant is associated with increased chronic pain risk by destabilizing the protein. Thus, GPR37L1 in SGCs offers a new target for neuropathy protection and pain control.
Collapse
|
14
|
Leite CBG, Merkely G, Charles JF, Lattermann C. From Inflammation to Resolution: Specialized Pro-resolving Mediators in Posttraumatic Osteoarthritis. Curr Osteoporos Rep 2023; 21:758-770. [PMID: 37615856 DOI: 10.1007/s11914-023-00817-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/25/2023]
Abstract
PURPOSE OF REVIEW To provide a comprehensive overview of the inflammatory response following anterior cruciate ligament (ACL) injury and to highlight the relationship between specialized pro-resolving mediators (SPMs) and inflammatory joint conditions, emphasizing the therapeutic potential of modulating the post-injury resolution of inflammation to prevent posttraumatic osteoarthritis (PTOA). RECENT FINDINGS The inflammatory response triggered after joint injuries such as ACL tear plays a critical role in posttraumatic osteoarthritis development. Inflammation is a necessary process for tissue healing, but unresolved or overactivated inflammation can lead to chronic diseases. SPMs, a family of lipid molecules derived from essential fatty acids, have emerged as active players in the resolution of inflammation and tissue repair. While their role in other inflammatory conditions has been studied, their relationship with PTOA remains underexplored. Proinflammatory mediators contribute to cartilage degradation and PTOA pathogenesis, while anti-inflammatory and pro-resolving mediators may have chondroprotective effects. Therapies aimed at suppressing inflammation in PTOA have limitations, as inflammation is crucial for tissue healing. SPMs offer a pro-resolving response without causing immunosuppression, making them a promising therapeutic option. The known onset date of PTOA makes it amenable to early interventions, and activating pro-resolving pathways may provide new possibilities for preventing PTOA progression. Harnessing the pro-resolving potential of SPMs may hold promise for preventing PTOA and restoring tissue homeostasis and function after joint injuries.
Collapse
Affiliation(s)
- Chilan B G Leite
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 20 Patriot Place Foxboro, Boston, MA, 02035, USA
| | - Gergo Merkely
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 20 Patriot Place Foxboro, Boston, MA, 02035, USA
| | - Julia F Charles
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 20 Patriot Place Foxboro, Boston, MA, 02035, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Christian Lattermann
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 20 Patriot Place Foxboro, Boston, MA, 02035, USA.
| |
Collapse
|
15
|
Kurtović Z, Sandor K, ter Heegde F, Rudjito R, Svensson CI, Palada V. circRNA landscape in dorsal root ganglia from mice with collagen antibody-induced arthritis. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 14:100142. [PMID: 38099281 PMCID: PMC10719524 DOI: 10.1016/j.ynpai.2023.100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 12/17/2023]
Abstract
Circular RNAs are a novel class of RNA molecules that are covalently closed into a ring structure. They are an epigenetic regulatory mechanism, and their best-studied function is regulation of microRNA activity. As such circular RNAs may be involved in the switch from acute to chronic pain. They have previously been studied in the context of neuropathic pain models, but their importance in inflammation-induced chronic pain models is unexplored. Microarray analysis of dorsal root ganglia collected in the late phase of collagen antibody-induced arthritis (day 59) were used to elucidate the relevance of circular RNAs in the mechanical hypersensitivity caused by this model. 120 circular RNA genes were found to be significantly differentially regulated in female BALB/c mice with collagen antibody-induced arthritis. Six genes were chosen for RT-qPCR analysis in the late (day 54-60) as well as the inflammatory (day 11-12) phase of this model. This validated an increase in circNufip1 expression in the late phase of collagen antibody-induced arthritis. Additionally, it was found that circVps13 and circMicall1 are upregulated in the inflammatory phase. Interestingly, no changes were found in dorsal root ganglia from mice injected with Freund's Complete Adjuvant (day 3) nor mice with spared nerve injury (day 20), despite their similarities to inflammatory and late phase collagen antibody-induced arthritis, respectively. This study provides evidence that mild circular RNA changes occur in dorsal root ganglia of mice with collagen antibody-induced arthritis that are, bioinformatically, predicated to be involved in processes relevant to sensitization.
Collapse
Affiliation(s)
- Zerina Kurtović
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Kancera AB, Karolinska Institutet Science Park, Solna, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Freija ter Heegde
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Resti Rudjito
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Vinko Palada
- Department of Physiology and Pharmacology and Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
16
|
Liu WC, Yang YH, Wang YC, Chang WM, Wang CW. Maresin: Macrophage Mediator for Resolving Inflammation and Bridging Tissue Regeneration-A System-Based Preclinical Systematic Review. Int J Mol Sci 2023; 24:11012. [PMID: 37446190 DOI: 10.3390/ijms241311012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Maresins are lipid mediators derived from omega-3 fatty acids with anti-inflammatory and pro-resolving properties, capable of promoting tissue regeneration and potentially serving as a therapeutic agent for chronic inflammatory diseases. The aim of this review was to systematically investigate preclinical and clinical studies on maresin to inform translational research. Two independent reviewers performed comprehensive searches with the term "Maresin (NOT) Review" on PubMed. A total of 137 studies were included and categorized into 11 human organ systems. Data pertinent to clinical translation were specifically extracted, including delivery methods, optimal dose response, and specific functional efficacy. Maresins generally exhibit efficacy in treating inflammatory diseases, attenuating inflammation, protecting organs, and promoting tissue regeneration, mostly in rodent preclinical models. The nervous system has the highest number of original studies (n = 25), followed by the cardiovascular system, digestive system, and respiratory system, each having the second highest number of studies (n = 18) in the field. Most studies considered systemic delivery with an optimal dose response for mouse animal models ranging from 4 to 25 μg/kg or 2 to 200 ng via intraperitoneal or intravenous injection respectively, whereas human in vitro studies ranged between 1 and 10 nM. Although there has been no human interventional clinical trial yet, the levels of MaR1 in human tissue fluid can potentially serve as biomarkers, including salivary samples for predicting the occurrence of cardiovascular diseases and periodontal diseases; plasma and synovial fluid levels of MaR1 can be associated with treatment response and defining pathotypes of rheumatoid arthritis. Maresins exhibit great potency in resolving disease inflammation and bridging tissue regeneration in preclinical models, and future translational development is warranted.
Collapse
Affiliation(s)
- Wen-Chun Liu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, No. 250, Wuxing St., Taipei 110310, Taiwan
| | - Yu-Hsin Yang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, No. 250, Wuxing St., Taipei 110310, Taiwan
| | - Yu-Chin Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, No. 250, Wuxing St., Taipei 110310, Taiwan
| | - Wei-Ming Chang
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Chin-Wei Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, No. 250, Wuxing St., Taipei 110310, Taiwan
- Division of Periodontics, Department of Dentistry, Taipei Medical University Hospital, Taipei 110301, Taiwan
| |
Collapse
|
17
|
Sideris-Lampretsas G, Oggero S, Zeboudj L, Silva R, Bajpai A, Dharmalingam G, Collier DA, Malcangio M. Galectin-3 activates spinal microglia to induce inflammatory nociception in wild type but not in mice modelling Alzheimer's disease. Nat Commun 2023; 14:3579. [PMID: 37349313 PMCID: PMC10287730 DOI: 10.1038/s41467-023-39077-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 05/26/2023] [Indexed: 06/24/2023] Open
Abstract
Musculoskeletal chronic pain is prevalent in individuals with Alzheimer's disease (AD); however, it remains largely untreated in these patients, raising the possibility that pain mechanisms are perturbed. Here, we utilise the TASTPM transgenic mouse model of AD with the K/BxN serum transfer model of inflammatory arthritis. We show that in male and female WT mice, inflammatory allodynia is associated with a distinct spinal cord microglial response characterised by TLR4-driven transcriptional profile and upregulation of P2Y12. Dorsal horn nociceptive afferent terminals release the TLR4 ligand galectin-3 (Gal-3), and intrathecal injection of a Gal-3 inhibitor attenuates allodynia. In contrast, TASTPM mice show reduced inflammatory allodynia, which is not affected by the Gal-3 inhibitor and correlates with the emergence of a P2Y12- TLR4- microglia subset in the dorsal horn. We suggest that sensory neuron-derived Gal-3 promotes allodynia through the TLR4-regulated release of pro-nociceptive mediators by microglia, a process that is defective in TASTPM due to the absence of TLR4 in a microglia subset.
Collapse
Affiliation(s)
| | - Silvia Oggero
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Lynda Zeboudj
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Rita Silva
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Archana Bajpai
- Eli Lilly & Company, Surrey, 8 Arlington Square West, Bracknell, RG12 1PU, United Kingdom
| | - Gopuraja Dharmalingam
- Eli Lilly & Company, Surrey, 8 Arlington Square West, Bracknell, RG12 1PU, United Kingdom
| | - David A Collier
- Eli Lilly & Company, Surrey, 8 Arlington Square West, Bracknell, RG12 1PU, United Kingdom
| | - Marzia Malcangio
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom.
| |
Collapse
|
18
|
Wang X, Botchway BOA, Zhang Y, Huang M, Liu X. Maresin1 can be a potential therapeutic target for nerve injury. Biomed Pharmacother 2023; 161:114466. [PMID: 36870281 DOI: 10.1016/j.biopha.2023.114466] [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: 02/07/2023] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Nerve injury significantly affects human motor and sensory function due to destruction of the integrity of nerve structure. In the wake of nerve injury, glial cells are activated, and synaptic integrity is destroyed, causing inflammation and pain hypersensitivity. Maresin1, an omega-3 fatty acid, is a derivative of docosahexaenoic acid. It has showed beneficial effects in several animal models of central and peripheral nerve injuries. In this review, we summarize the anti-inflammatory, neuroprotective and pain hypersensitivity effects of maresin1 in nerve injury and provide a theoretical basis for the clinical treatment of nerve injury using maresin1.
Collapse
Affiliation(s)
- Xichen Wang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China; Bupa Cromwell Hospital, London, UK
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China.
| |
Collapse
|
19
|
Kumar V, Yasmeen N, Chaudhary AA, Alawam AS, Al-Zharani M, Suliman Basher N, Harikrishnan S, Goud MD, Pandey A, Lakhawat SS, Sharma PK. Specialized pro-resolving lipid mediators regulate inflammatory macrophages: A paradigm shift from antibiotics to immunotherapy for mitigating COVID-19 pandemic. Front Mol Biosci 2023; 10:1104577. [PMID: 36825200 PMCID: PMC9942001 DOI: 10.3389/fmolb.2023.1104577] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
The most severe clinical manifestations of the horrifying COVID-19 disease, that claimed millions of lives during the pandemic time, were Acute respiratory distress syndrome (ARDS), Coagulopathies, septic shock leading eventually to death. ARDS was a consequence of Cytokine storm. The viral SARS-COV2infection lead to avalanche of cytokines and eicosanoids causing "cytokine storm" and "eicosanoid storm." Cytokine storm is one of the macrophage-derived inflammatory responses triggered by binding of virus particles to ACE2 receptors of alveolar macrophages, arise mainly due to over production of various pro-inflammatory mediators like cytokines, e.g., interleukin (IL)-1, IL-2, and tumor necrosis factor (TNF)- α, causing pulmonary edema, acute respiratory distress, and multi-organ failure. Cytokine storm was regarded as the predictor of severity of the disease and was deemed one of the causes of the high mortality rates due to the COVID-19. The basis of cytokine storm is imbalanced switching between an inflammation increasing - pro-inflammatory (M1) and an inflammation regulating-anti-inflammatory (M2) forms of alveolar macrophages which further deteriorates if opportunistic secondary bacterial infections prevail in the lungs. Lack of sufficient knowledge regarding the virus and its influence on co-morbidities, clinical treatment of the diseases included exorbitant use of antibiotics to mitigate secondary bacterial infections, which led to the unwarranted development of multidrug resistance (MDR) among the population across the globe. Antimicrobial resistance (AMR) needs to be addressed from various perspectives as it may deprive future generations of the basic health immunity. Specialized pro-resolving mediators (SPMs) are generated from the stereoselective enzymatic conversions of essential fatty acids that serve as immune resolvents in controlling acute inflammatory responses. SPMs facilitate the clearance of injured tissue and cell debris, the removal of pathogens, and augment the concentration of anti-inflammatory lipid mediators. The SPMs, e.g., lipoxins, protectins, and resolvins have been implicated in exerting inhibitory influence on with cytokine storm. Experimental evidence suggests that SPMS lower antibiotic requirement. Therefore, in this review potential roles of SPMs in enhancing macrophage polarization, triggering immunological functions, hastening inflammation resolution, subsiding cytokine storm and decreasing antibiotic requirement that can reduce AMR load are discussed.
Collapse
Affiliation(s)
- Vikram Kumar
- Amity institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India,*Correspondence: Vikram Kumar,
| | - Nusrath Yasmeen
- Amity institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Abdullah S. Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohammed Al-Zharani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Nosiba Suliman Basher
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - S. Harikrishnan
- Amity institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | | | - Aishwarya Pandey
- INRS, Eau Terre Environnement Research Centre, Québec, QC, Canada
| | | | | |
Collapse
|
20
|
Ji RR. Specialized Pro-Resolving Mediators as Resolution Pharmacology for the Control of Pain and Itch. Annu Rev Pharmacol Toxicol 2023; 63:273-293. [PMID: 36100219 DOI: 10.1146/annurev-pharmtox-051921-084047] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Specialized pro-resolving mediators (SPMs), including resolvins, protectins, and maresins, are endogenous lipid mediators that are synthesized from omega-3 polyunsaturated fatty acids during the acute phase or resolution phase of inflammation. Synthetic SPMs possess broad safety profiles and exhibit potent actions in resolving inflammation in preclinical models. Accumulating evidence in the past decade has demonstrated powerful analgesia of exogenous SPMs in rodent models of inflammatory, neuropathic, and cancer pain. Furthermore, endogenous SPMs are produced by sham surgery and neuromodulation (e.g., vagus nerve stimulation). SPMs produce their beneficial actions through multiple G protein-coupled receptors, expressed by immune cells, glial cells, and neurons. Notably, loss of SPM receptors impairs the resolution of pain. I also highlight the emerging role of SPMs in the control of itch. Pharmacological targeting of SPMs or SPM receptors has the potential to lead to novel therapeutics for pain and itch as emerging approaches in resolution pharmacology.
Collapse
Affiliation(s)
- Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, and Departments of Neurobiology and Cell Biology, Duke University Medical Center, Durham, North Carolina, USA;
| |
Collapse
|
21
|
Inflammation and Infection in Pain and the Role of GPR37. Int J Mol Sci 2022; 23:ijms232214426. [PMID: 36430912 PMCID: PMC9692891 DOI: 10.3390/ijms232214426] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Inflammation is known to cause pain, and pain is of one of the cardinal signs of inflammation. Mounting evidence suggests that acute inflammation also resolves pain through specialized pro-resolving mediators (SPMs) and macrophage signaling. GPR37 is expressed by neurons and oligodendrocytes in the brain and has been implicated in multiple disorders, such as demyelination, Parkinson's disease, stroke, and cancer. Recent studies have demonstrated that GPR37 is expressed by macrophages and confers protection against infection by bacteria and parasites. Furthermore, GPR37 promotes the resolution of inflammatory pain and infection-induced pain, as the duration of pain after tissue injury and infection is prolonged in mice lacking Gpr37. Mechanistically, activation of GPR37 enhances macrophage phagocytosis, and Gpr37-deficient macrophages exhibit dysregulations of pro-inflammatory and anti-inflammatory cytokines, switching from M2- to M1-like phenotypes. We also discuss novel ligands of GPR37, including neuroprotectin D1 (NPD1), a SPM derived from docosahexaenoic acid (DHA), and bone-derived hormone osteocalcin (OCN), which can suppress oligodendrocyte differentiation and myelination. NPD1 stimulates macrophage phagocytosis via GPR37 and exhibits potent analgesic actions in various animal models of inflammatory and neuropathic pain. Targeting GPR37 may lead to novel therapeutics for treating inflammation, infection, pain, and neurological diseases.
Collapse
|
22
|
Oggero S, Cecconello C, Silva R, Zeboudj L, Sideris-Lampretsas G, Perretti M, Malcangio M. Dorsal root ganglia CX3CR1 expressing monocytes/macrophages contribute to arthritis pain. Brain Behav Immun 2022; 106:289-306. [PMID: 36115544 PMCID: PMC10166715 DOI: 10.1016/j.bbi.2022.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 11/18/2022] Open
Abstract
Pain is a persistent symptom of Rheumatoid Arthritis, and the K/BxN serum transfer model recapitulates both association and dissociation between pain and joint inflammation in RA. Furthermore, this model features monocyte/macrophage infiltration in joints and lumbar dorsal root ganglia (DRG), where these immune cells are close to nociceptive neurons. We focussed on CX3CR1-monocyte/macrophage trafficking and show that at peak paw swelling associated with nociception, CX3CR1 deletion altered neither swelling nor macrophage infiltration/phenotype in paws. However, acute nociception and DRG non-classical monocyte numbers were reduced in CX3CR1GFP/GFP (KO) compared to CX3CR1+/GFP (WT). Nociception that persisted despite swelling had resolved was attenuated in KO and correlated with DRG macrophages displaying M2-like phenotype. Still in the DRG, neurons up-regulated neuropeptide CGRP and olcegepant treatment reduced acute swelling, nociception, and leukocyte infiltration in paws and DRG. We delineate in-vitro a signalling pathway showing that CGRP liberates the CX3CR1 ligand fractalkine (FKN) from endothelium, and in bone marrow-derived macrophages, FKN promotes activation of intracellular kinases, polarisation towards M1-like phenotype and release of pro-nociceptive IL-6. These data implicate non-classical CX3CR1-expressing monocyte and macrophage recruitment into the DRG in initiation and maintenance of arthritis pain.
Collapse
Affiliation(s)
- Silvia Oggero
- Wolfson Centre for Age Related Diseases, King's College London, London SE1 1UL, UK
| | - Chiara Cecconello
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Rita Silva
- Wolfson Centre for Age Related Diseases, King's College London, London SE1 1UL, UK
| | - Lynda Zeboudj
- Wolfson Centre for Age Related Diseases, King's College London, London SE1 1UL, UK
| | | | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK; Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Marzia Malcangio
- Wolfson Centre for Age Related Diseases, King's College London, London SE1 1UL, UK.
| |
Collapse
|
23
|
Malange KF, Navia-Pelaez JM, Dias EV, Lemes JBP, Choi SH, Dos Santos GG, Yaksh TL, Corr M. Macrophages and glial cells: Innate immune drivers of inflammatory arthritic pain perception from peripheral joints to the central nervous system. FRONTIERS IN PAIN RESEARCH 2022; 3:1018800. [PMID: 36387416 PMCID: PMC9644179 DOI: 10.3389/fpain.2022.1018800] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Millions of people suffer from arthritis worldwide, consistently struggling with daily activities due to debilitating pain evoked by this disease. Perhaps the most intensively investigated type of inflammatory arthritis is rheumatoid arthritis (RA), where, despite considerable advances in research and clinical management, gaps regarding the neuroimmune interactions that guide inflammation and chronic pain in this disease remain to be clarified. The pain and inflammation associated with arthritis are not isolated to the joints, and inflammatory mechanisms induced by different immune and glial cells in other tissues may affect the development of chronic pain that results from the disease. This review aims to provide an overview of the state-of-the-art research on the roles that innate immune, and glial cells play in the onset and maintenance of arthritis-associated pain, reviewing nociceptive pathways from the joint through the dorsal root ganglion, spinal circuits, and different structures in the brain. We will focus on the cellular mechanisms related to neuroinflammation and pain, and treatments targeting these mechanisms from the periphery and the CNS. A comprehensive understanding of the role these cells play in peripheral inflammation and initiation of pain and the central pathways in the spinal cord and brain will facilitate identifying new targets and pathways to aide in developing therapeutic strategies to treat joint pain associated with RA.
Collapse
Affiliation(s)
- Kaue Franco Malange
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | | | - Elayne Vieira Dias
- Department of Neurology, University of California, San Francisco, CA, United States
| | | | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, CA, United States
| | | | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|
24
|
Maresin 2 is an analgesic specialized pro-resolution lipid mediator in mice by inhibiting neutrophil and monocyte recruitment, nociceptor neuron TRPV1 and TRPA1 activation, and CGRP release. Neuropharmacology 2022; 216:109189. [PMID: 35820471 DOI: 10.1016/j.neuropharm.2022.109189] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022]
Abstract
Maresin-2 (MaR2) is a specialized pro-resolution lipid mediator (SPM) that reduces neutrophil recruitment in zymosan peritonitis. Here, we investigated the analgesic effect of MaR2 and its mechanisms in different mouse models of pain. For that, we used the lipopolysaccharide (LPS)-induced mechanical hyperalgesia (electronic version of the von Frey filaments), thermal hyperalgesia (hot plate test) and weight distribution (static weight bearing), as well as the spontaneous pain models induced by capsaicin (TRPV1 agonist) or AITC (TRPA1 agonist). Immune cell recruitment was determined by immunofluorescence and flow cytometry while changes in the pro-inflammatory mediator landscape were determined using a proteome profiler kit and ELISA after LPS injection. MaR2 treatment was also performed in cultured DRG neurons stimulated with capsaicin or AITC in the presence or absence of LPS. The effect of MaR2 on TRVP1- and TRPA1-dependent CGRP release by cultured DRG neurons was determined by EIA. MaR2 inhibited LPS-induced inflammatory pain and changes in the cytokine landscape as per cytokine array assay. MaR2 also inhibited TRPV1 and TRPA1 activation as observed by a reduction in calcium influx in cultured DRG neurons, and the number of flinches and time spent licking the paw induced by capsaicin or AITC. In corroboration, MaR2 reduced capsaicin- and AITC-induced CGRP release by cultured DRG neurons and immune cell recruitment to the paw skin close the CGRP+ fibers. In conclusion, we show that MaR2 is an analgesic SPM that acts by targeting leukocyte recruitment, nociceptor TRPV1 and TRPA1 activation, and CGRP release in mice.
Collapse
|
25
|
Lin J, Ren J, Zhu B, Dai Y, Gao DS, Xia S, Cheng Z, Huang Y, Yu L. Dimethyl Itaconate Attenuates CFA-Induced Inflammatory Pain via the NLRP3/ IL-1β Signaling Pathway. Front Pharmacol 2022; 13:938979. [PMID: 35935847 PMCID: PMC9353300 DOI: 10.3389/fphar.2022.938979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Itaconate plays a prominent role in anti-inflammatory effects and has gradually been ushered as a promising drug candidate for treating inflammatory diseases. However, its significance and underlying mechanism for inflammatory pain remain unexplored. In the current study, we investigated the effects and mechanisms of Dimethyl Itaconate (DI, a derivative of itaconate) on Complete Freund’s adjuvant (CFA)-induced inflammatory pain in a rodent model. Here, we demonstrated that DI significantly reduced mechanical allodynia and thermal hyperalgesia. The DI-attenuated neuroinflammation was evident with the amelioration of infiltrative macrophages in peripheral sites of the hind paw and the dorsal root ganglion. Concurrently, DI hindered the central microglia activation in the spinal cord. Mechanistically, DI inhibited the expression of pro-inflammatory factors interleukin (IL)-1β and tumor necrosis factor alpha (TNF-α) and upregulated anti-inflammatory factor IL-10. The analgesic mechanism of DI was related to the downregulation of the nod-like receptor protein 3 (NLRP3) inflammasome complex and IL-1β secretion. This study suggested possible novel evidence for prospective itaconate utilization in the management of inflammatory pain.
Collapse
|
26
|
Liu Y, Caterina MJ, Qu L. Sensory Neuron Expressed FcγRI Mediates Postinflammatory Arthritis Pain in Female Mice. Front Immunol 2022; 13:889286. [PMID: 35833115 PMCID: PMC9271677 DOI: 10.3389/fimmu.2022.889286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Persistent arthritis pain after resolution of joint inflammation represents a huge health burden in patients with rheumatoid arthritis (RA). However, the underling mechanisms are poorly understood. We and other groups recently revealed that FcγRI, a key immune receptor, is functionally expressed in joint nociceptors. Thus, we investigated a potential role of sensory neuron expressed FcγRI in postinflammatory arthritis pain in a mouse model of collagen antibody-induced arthritis (CAIA). Here, we show that global deletion of Fcgr1 significantly attenuated mechanical hyperalgesia in the ankle and hind paw of female mice in both inflammatory and postinflammatory phases of CAIA. No obvious differences in cartilage destruction were observed after resolution of joint inflammation between genotypes. In situ hybridization (ISH) revealed that a larger proportion of dorsal root ganglion (DRG) neurons expressed Fcgr1 mRNA signal in the late phase of CAIA. Conditional deletion of Fcgr1 in primary sensory neurons produced similar analgesic effects without affecting joint swelling. Knockdown of Fcgr1 expression within DRG in the postinflammatory phase of CAIA alleviated persistent pain. Inflammation within DRG after resolution of joint inflammation in the CAIA model was evidenced by T cell and neutrophil infiltration and upregulated mRNA expression of numerous inflammatory mediators. Yet, such changes were not altered by genetic deletion of Fcgr1. We suggest that neuroinflammation within the DRG after resolution of joint inflammation might upregulate FcγRI signaling in DRG neurons. Sensory neuron expressed FcγRI thus merits exploration as a potential target for the treatment of arthritis pain that persists in RA patients in remission.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Michael J. Caterina
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Lintao Qu
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- *Correspondence: Lintao Qu,
| |
Collapse
|
27
|
Zaninelli TH, Fattori V, Saraiva-Santos T, Badaro-Garcia S, Staurengo-Ferrari L, Andrade KC, Artero NA, Ferraz CR, Bertozzi MM, Rasquel-Oliveira F, Manchope MF, Amaral FA, Teixeira MM, Borghi SM, Rogers MS, Casagrande R, Verri WA. RvD1 disrupts nociceptor neuron and macrophage activation, and neuroimmune communication reducing pain and inflammation in gouty arthritis in mice. Br J Pharmacol 2022; 179:4500-4515. [PMID: 35716378 DOI: 10.1111/bph.15897] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 04/27/2022] [Accepted: 05/25/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Gouty arthritis is characterised by an intense inflammatory response to monosodium urate crystals (MSU), which induces severe pain. Current therapies are often ineffective in reducing gout-related pain. Resolvin D1 (RvD1) is a specialised pro-resolving lipid mediator with anti-inflammatory and analgesic proprieties. In this study, we evaluated the effects and mechanisms of action of RvD1 in an experimental mouse model of gouty arthritis, an aim that was not pursued previously in the literature. EXPERIMENTAL APPROACH Male mice were treated with RvD1 (intrathecally or intraperitoneally) before or after intraarticular stimulation with MSU. Mechanical hyperalgesia was assessed using an electronic von Frey aesthesiometer. Leukocyte recruitment was determined by knee joint wash cell counting and immunofluorescence. IL-1β production was measured by ELISA. Phosphorylated NF-kB and apoptosis-associated speck-like protein containing CARD (ASC) were detected by immunofluorescence, and mRNA expression was determined by RT-qPCR. CGRP release was determined by EIA and immunofluorescence. MSU crystal phagocytosis was evaluated by confocal microscopy. KEY RESULTS RvD1 inhibited MSU-induced mechanical hyperalgesia in a dose- and time-dependent manner by reducing leukocyte recruitment and IL-1β production in the knee joint. Intrathecal RvD1 reduced the activation of peptidergic neurons and macrophages as well as silenced nociceptor to macrophage communication and macrophage function. CGRP stimulated MSU phagocytosis and IL-1β production by macrophages. RvD1 downmodulated this phenomenon directly by acting on macrophages, and indirectly by inhibiting CGRP release and CGRP-dependent activation of macrophages. CONCLUSIONS AND IMPLICATIONS This study reveals a hitherto unknown neuro-immune axis in gouty arthritis that is targeted by RvD1.
Collapse
Affiliation(s)
- Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.,Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, Boston, Massachusetts, United States
| | - Telma Saraiva-Santos
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Stephanie Badaro-Garcia
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Larissa Staurengo-Ferrari
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Ketlem C Andrade
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Nayara A Artero
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Camila R Ferraz
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Mariana M Bertozzi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Fernanda Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Marilia F Manchope
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Flávio A Amaral
- Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Sergio M Borghi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Michael S Rogers
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, Boston, Massachusetts, United States
| | - Rubia Casagrande
- Laboratory of Antioxidants and Inflammation, Department of Pharmaceutical Sciences, Centre of Health Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Centre of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| |
Collapse
|
28
|
Pistorius K, Ly L, Souza PR, Gomez EA, Koenis DS, Rodriguez AR, Foster J, Sosabowski J, Hopkinson M, Rajeeve V, Spur BW, Pitsillides A, Pitzalis C, Dalli J. MCTR3 reprograms arthritic monocytes to upregulate Arginase-1 and exert pro-resolving and tissue-protective functions in experimental arthritis. EBioMedicine 2022; 79:103974. [PMID: 35430453 PMCID: PMC9038546 DOI: 10.1016/j.ebiom.2022.103974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a progressive degenerative disorder that leads to joint destruction. Available treatments only target the inflammatory component with minimal impact on joint repair. We recently uncovered a previously unappreciated family of pro-resolving mediators, the maresin conjugate in tissue regeneration (MCTR), that display both immunoregulatory and tissue-protective activities. Thus, we queried whether the production of these autacoids is disrupted in RA patients and whether they can be useful in treating joint inflammation and promoting joint repair. METHODS Using a highly phenotyped RA cohort we evaluated plasma MCTR concentrations and correlated these to clinical markers of disease activity. To evaluate the immunoregulatory and tissue reparative activities we employed both in vivo models of arthritis and organ culture models. FINDINGS Herein, we observed that plasma MCTR3 concentrations were negatively correlated with joint disease activity and severity in RA patients. Evaluation of the mechanisms engaged by this mediator in arthritic mice demonstrated that MCTR3 reprograms monocytes to confer enduring joint protective properties. Single cell transcriptomic profiling and flow cytometric evaluation of macrophages from mice treated with MCTR3-reprogrammed monocytes revealed a role for Arginase-1 (Arg-1) in mediating their joint reparative and pro-resolving activities. Arg-1 inhibition reversed both the anti-arthritic and tissue reparative actions of MCTR3-reprogrammed monocytes. INTERPRETATION Our findings demonstrate that circulating MCTR3 levels are negatively correlated with disease in RA. When administered to mice in vivo, MCTR3 displayed both anti-inflammatory and joint reparative activities, protecting both cartilage and bone in murine arthritis. These activities were, at least in part, mediated via the reprogramming of mononuclear phagocyte responses. FUNDING This work was supported by funding from the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation programme (grant no: 677542) and the Barts Charity (grant no: MGU0343) to J.D. J.D. is also supported by a Sir Henry Dale Fellowship jointly funded by the Wellcome Trust and the Royal Society (grant 107613/Z/15/Z).
Collapse
Affiliation(s)
- Kimberly Pistorius
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Lucy Ly
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Patricia R Souza
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Esteban A Gomez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Duco S Koenis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Ana R Rodriguez
- Rowan University School of Osteopathic Medicine, Department of Cell Biology & Neuroscience, 2 Medical Centre Drive, Stratford NJ 08084, USA
| | - Julie Foster
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Jane Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Mark Hopkinson
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Vinothini Rajeeve
- Mass spectrometry Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Bernd W Spur
- Rowan University School of Osteopathic Medicine, Department of Cell Biology & Neuroscience, 2 Medical Centre Drive, Stratford NJ 08084, USA
| | - Andrew Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK; Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
29
|
Dalli J, Gomez EA, Jouvene CC. Utility of the Specialized Pro-Resolving Mediators as Diagnostic and Prognostic Biomarkers in Disease. Biomolecules 2022; 12:biom12030353. [PMID: 35327544 PMCID: PMC8945731 DOI: 10.3390/biom12030353] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 12/14/2022] Open
Abstract
A precision medicine approach is widely acknowledged to yield more effective therapeutic strategies in the treatment of patients with chronic inflammatory conditions than the prescriptive paradigm currently utilized in the management and treatment of these patients. This is because such an approach will take into consideration relevant factors including the likelihood that a patient will respond to given therapeutics based on their disease phenotype. Unfortunately, the application of this precision medicine paradigm in the daily treatment of patients has been greatly hampered by the lack of robust biomarkers, in particular biomarkers for determining early treatment responsiveness. Lipid mediators are central in the regulation of host immune responses during both the initiation and resolution of inflammation. Amongst lipid mediators, the specialized pro-resolving mediators (SPM) govern immune cells to promote the resolution of inflammation. These autacoids are produced via the stereoselective conversion of essential fatty acids to yield molecules that are dynamically regulated during inflammation and exert potent immunoregulatory activities. Furthermore, there is an increasing appreciation for the role that these mediators play in conveying the biological actions of several anti-inflammatory therapeutics, including statins and aspirin. Identification and quantitation of these mediators has traditionally been achieved using hyphenated mass spectrometric techniques, primarily liquid-chromatography tandem mass spectrometry. Recent advances in the field of chromatography and mass spectrometry have increased both the robustness and the sensitivity of this approach and its potential deployment for routine clinical diagnostics. In the present review, we explore the evidence supporting a role for specific SPM as potential biomarkers for patient stratification in distinct disease settings together with methodologies employed in the identification and quantitation of these autacoids.
Collapse
Affiliation(s)
- Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.A.G.); (C.C.J.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
- Correspondence:
| | - Esteban Alberto Gomez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.A.G.); (C.C.J.)
| | - Charlotte Camille Jouvene
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (E.A.G.); (C.C.J.)
| |
Collapse
|
30
|
Falsetta ML, Wood RW, Linder MA, Bonham AD, Honn KV, Maddipati KR, Phipps RP, Haidaris CG, Foster DC. Specialized Pro-resolving Mediators Reduce Pro-nociceptive Inflammatory Mediator Production in Models of Localized Provoked Vulvodynia. THE JOURNAL OF PAIN 2021; 22:1195-1209. [PMID: 33813057 PMCID: PMC8484336 DOI: 10.1016/j.jpain.2021.03.144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/05/2021] [Accepted: 03/24/2021] [Indexed: 12/25/2022]
Abstract
Localized provoked vulvodynia (LPV) is the most common cause of chronic dyspareunia in premenopausal women, characterized by pain with light touch to the vulvar vestibule surrounding the vaginal opening. The devastating impact of LPV includes sexual dysfunction, infertility, depression, and even suicide. Yet, its etiology is unclear. No effective medical therapy exists; surgical removal of the painful vestibule is the last resort. In LPV, the vestibule expresses a unique inflammatory profile with elevated levels of pro-nociceptive proinflammatory mediators prostaglandin E2 (PGE2) and interleukin-6 (IL-6), which are linked to lower mechanical sensitivity thresholds. Specialized pro-resolving mediators (SPMs), lipids produced endogenously within the body, hold promise as an LPV treatment by resolving inflammation without impairing host defense. Ten of 13 commercially available SPMs reduced IL-6 and PGE2 production by vulvar fibroblasts, administered either before or after inflammatory stimulation. Using a murine vulvar pain model, coupling proinflammatory mediator quantification with mechanical sensitivity threshold determination, topical treatment with the SPM, maresin 1, decreased sensitivity and suppressed PGE2 levels. Docosahexaenoic acid, a precursor of maresin 1, was also effective in reducing PGE2 in vulvar fibroblasts and rapidly restored mouse sensitivity thresholds. Overall, SPMs and their precursors may be a safe and efficacious for LPV. Perspective: Vulvodynia, like many pain conditions, is difficult to treat because disease origins are incompletely understood. Here, we applied our knowledge of more recently discovered vulvodynia disease mechanisms to screen novel therapeutics. We identified several specialized pro-resolving mediators as likely potent and safe for treating LPV with potential for broader application.
Collapse
Affiliation(s)
- Megan L Falsetta
- Department of Obstetrics and Gynecology, University of Rochester, School of Medicine and Dentistry Rochester, New York; Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York.
| | - Ronald W Wood
- Department of Obstetrics and Gynecology, University of Rochester, School of Medicine and Dentistry Rochester, New York
| | - Mitchell A Linder
- Department of Obstetrics and Gynecology, University of Rochester, School of Medicine and Dentistry Rochester, New York
| | - Adrienne D Bonham
- Department of Obstetrics and Gynecology, University of Rochester, School of Medicine and Dentistry Rochester, New York
| | - Kenneth V Honn
- Department of Pathology, Wayne State University, School of Medicine, Detroit, Michigan
| | - Krishna Rao Maddipati
- Department of Pathology, Wayne State University, School of Medicine, Detroit, Michigan
| | | | - Constantine G Haidaris
- Department of Microbiology and Immunology, University of Rochester, School of Medicine and Dentistry Rochester, New York
| | - David C Foster
- Department of Obstetrics and Gynecology, University of Rochester, School of Medicine and Dentistry Rochester, New York
| |
Collapse
|
31
|
Zaninelli TH, Fattori V, Verri WA. Harnessing Inflammation Resolution in Arthritis: Current Understanding of Specialized Pro-resolving Lipid Mediators' Contribution to Arthritis Physiopathology and Future Perspectives. Front Physiol 2021; 12:729134. [PMID: 34539449 PMCID: PMC8440959 DOI: 10.3389/fphys.2021.729134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/12/2021] [Indexed: 12/26/2022] Open
Abstract
The concept behind the resolution of inflammation has changed in the past decades from a passive to an active process, which reflects in novel avenues to understand and control inflammation-driven diseases. The time-dependent and active process of resolution phase is orchestrated by the endogenous biosynthesis of specialized pro-resolving lipid mediators (SPMs). Inflammation and its resolution are two forces in rheumatic diseases that affect millions of people worldwide with pain as the most common experienced symptom. The pathophysiological role of SPMs in arthritis has been demonstrated in pre-clinical and clinical studies (no clinical trials yet), which highlight their active orchestration of disease control. The endogenous roles of SPMs also give rise to the opportunity of envisaging these molecules as novel candidates to improve the life quality of rhematic diseases patients. Herein, we discuss the current understanding of SPMs endogenous roles in arthritis as pro-resolutive, protective, and immunoresolvent lipids.
Collapse
Affiliation(s)
- Tiago H Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| | - Victor Fattori
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Londrina State University, Londrina, Brazil
| |
Collapse
|
32
|
Imbalance of proresolving lipid mediators in persistent allodynia dissociated from signs of clinical arthritis: erratum. Pain 2021; 162:2456. [PMID: 34448758 DOI: 10.1097/j.pain.0000000000002435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 11/26/2022]
|
33
|
Leuti A, Fava M, Pellegrini N, Maccarrone M. Role of Specialized Pro-Resolving Mediators in Neuropathic Pain. Front Pharmacol 2021; 12:717993. [PMID: 34456731 PMCID: PMC8385637 DOI: 10.3389/fphar.2021.717993] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammation and neuroinflammation are critical mechanisms in the generation of neuropathic pain that is experienced in several chronic diseases. The aberrant inflammation that triggers this pathophysiologic process can be tracked down to an exacerbated immune response, which establishes a vicious cycle and continuously recruits inflammatory cells by inducing chronic tissue damage. Recently, impairment of the cellular and molecular machinery orchestrated by specialized pro-resolving mediators (SPMs)-i.e., endogenous lipids termed resolvins, protectins, maresins, and lipoxins that confine the inflammatory cascades in space and time during the "resolution of inflammation"-has emerged as a crucial event in the derangement of the inflammatory homeostasis and the onset of chronic inflammation and pain. Indeed, a deviant inflammatory response that is not adequately controlled by the resolution network leads to the overproduction of pro-inflammatory eicosanoids that, opposite to SPMs, lead to neuropathic pain. Interestingly, in the last two decades convincing evidence has demonstrated that SPMs antagonize the in vivo activity of pro-inflammatory eicosanoids and, overall, exert potent anti-hyperalgesic effects in a number of pain-associated paradigms of disease, such as arthritis and chemotherapy-induced peripheral neuropathy, as well as in many experimental models of pain like mechanical allodynia, chemical pain, heat hypersensitivity and phase 1 and 2 inflammatory pain. Of note, accumulated evidence supports a synergy between SPMs and other signalling pathways, such as those mediated by transient receptor potential (TRP) channels and those triggered by opioid receptors, suggesting that the cascade of events where inflammation and pain perception take part might be ways more intricated than originally expected. Here, we aim at presenting a state-of-the-art view of SPMs, their metabolism and signalling, in the context of cellular and molecular pathways associated to neuropathic pain.
Collapse
Affiliation(s)
- Alessandro Leuti
- Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy.,European Center for Brain Research/IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marina Fava
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Rome, Italy.,Faculty of Biosciences and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Niccolò Pellegrini
- Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
34
|
Abstract
Pain is a near-universal feature of rheumatoid arthritis, but peripheral joint inflammation may not suffice to explain the etiology of pain in all patients with rheumatoid arthritis. Inflammation in rheumatoid arthritis releases several algogens that may generate pain. Also, central nervous system processes may play a crucial role in the regulation and perpetuation of pain. Several methods for assessing pain in rheumatoid arthritis exist, and recently the role of assessing therapeutics in treating specific etiologies of pain has gained interest.
Collapse
Affiliation(s)
- Priyanka Iyer
- Division of Rheumatology, Department of Internal Medicine, University of California Irvine, Irvine, CA, USA.
| | | |
Collapse
|
35
|
Tao X, Lee MS, Donnelly CR, Ji RR. Neuromodulation, Specialized Proresolving Mediators, and Resolution of Pain. Neurotherapeutics 2020; 17:886-899. [PMID: 32696274 PMCID: PMC7609770 DOI: 10.1007/s13311-020-00892-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The current crises in opioid abuse and chronic pain call for the development of nonopioid and nonpharmacological therapeutics for pain relief. Neuromodulation-based approaches, such as spinal cord stimulation, dorsal root ganglion simulation, and nerve stimulation including vagus nerve stimulation, have shown efficacy in achieving pain control in preclinical and clinical studies. However, the mechanisms by which neuromodulation alleviates pain are not fully understood. Accumulating evidence suggests that neuromodulation regulates inflammation and neuroinflammation-a localized inflammation in peripheral nerves, dorsal root ganglia/trigeminal ganglia, and spinal cord/brain-through neuro-immune interactions. Specialized proresolving mediators (SPMs) such as resolvins, protectins, maresins, and lipoxins are lipid molecules produced during the resolution phase of inflammation and exhibit multiple beneficial effects in resolving inflammation in various animal models. Recent studies suggest that SPMs inhibit inflammatory pain, postoperative pain, neuropathic pain, and cancer pain in rodent models via immune, glial, and neuronal modulations. It is noteworthy that sham surgery is sufficient to elevate resolvin levels and may serve as a model of resolution. Interestingly, it has been shown that the vagus nerve produces SPMs and vagus nerve stimulation (VNS) induces SPM production in vitro. In this review, we discuss how neuromodulation such as VNS controls pain via immunomodulation and neuro-immune interactions and highlight possible involvement of SPMs. In particular, we demonstrate that VNS via auricular electroacupuncture effectively attenuates chemotherapy-induced neuropathic pain. Furthermore, auricular stimulation is able to increase resolvin levels in mice. Thus, we propose that neuromodulation may control pain and inflammation/neuroinflammatioin via SPMs. Finally, we discuss key questions that remain unanswered in our understanding of how neuromodulation-based therapies provide short-term and long-term pain relief.
Collapse
Affiliation(s)
- Xueshu Tao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Michael S Lee
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Christopher R Donnelly
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, 27710, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|