1
|
Tang Y, Wang Z, Cao J, Tu Y. Bone-brain crosstalk in osteoarthritis: pathophysiology and interventions. Trends Mol Med 2025; 31:281-295. [PMID: 39438197 DOI: 10.1016/j.molmed.2024.09.006] [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: 07/28/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Osteoarthritis (OA) is a prevalent articular disorder characterized by joint degeneration and persistent pain; it imposes a significant burden on both individuals and society. While OA has traditionally been viewed as a localized peripheral disorder, recent preclinical and clinical studies have revealed the crucial interconnections between the bone and the brain, highlighting the systemic nature of OA. The neuronal pathway, molecular signaling, circadian rhythms, and genetic underpinnings within the bone-brain axis play vital roles in the complex interplay that contributes to OA initiation and progression. This review explores emerging evidence of the crosstalk between the bone and brain in OA progression, and discusses the potential contributions of the bone-brain axis to the development of effective interventions for managing OA.
Collapse
Affiliation(s)
- Yilan Tang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyan Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Cao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Yiheng Tu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Rodríguez-Palma EJ, Loya-Lopez S, Allen K, Cruz-Almeida Y, Khanna R. The contribution of clock genes BMAL1 and PER2 in osteoarthritis-associated pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2025; 17:100177. [PMID: 39850977 PMCID: PMC11754085 DOI: 10.1016/j.ynpai.2024.100177] [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: 10/03/2024] [Revised: 12/14/2024] [Accepted: 12/14/2024] [Indexed: 01/25/2025]
Abstract
Joint pain is the primary symptom of osteoarthritis (OA) and the main motivator for patients to seek medical care. OA-related pain significantly restricts joint function and diminishes quality of life. Despite the availability of various pain-relieving medications for OA, current treatment strategies often fall short in delivering adequate pain relief. Furthermore, long-term use of pain medications for OA management is frequently linked with notable side effects and toxicities, suggesting the need to explore new potential targets to treat pain in OA patients. In this context, clock genes, particularly brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1 (BMAL1) and period circadian protein homolog 2 (PER2), known for their role in circadian rhythms, represent promising opportunities for pharmacological interventions due to their involvement in both the development and maintenance of OA pain. While BMAL1 and PER2 have been extensively studied in neuropathic and inflammatory pain, their specific contributions to OA pain remain less clear, demanding further investigation. This narrative review aims to synthesize the relationship between OA pain and the BMAL1 and PER2 signaling pathways, ultimately exploring the potential therapeutic role of clock genes in addressing this challenging condition.
Collapse
Affiliation(s)
- Erick J. Rodríguez-Palma
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Santiago Loya-Lopez
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Kyle Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
- Pain Research & Intervention Center of Excellence (PRICE), University of Florida, Gainesville, FL, USA
- Pain Research and Integrated Neuroscience Center (PRINC), College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yenisel Cruz-Almeida
- Pain Research & Intervention Center of Excellence (PRICE), University of Florida, Gainesville, FL, USA
- Pain Research and Integrated Neuroscience Center (PRINC), College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rajesh Khanna
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
- Pain Research & Intervention Center of Excellence (PRICE), University of Florida, Gainesville, FL, USA
- Pain Research and Integrated Neuroscience Center (PRINC), College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Mun CJ, Youngstedt SD, Petrov ME, Pituch KA, Elliott JA, George SZ, LoVecchio F, Mardian AS, Elam KK, Winsick N, Eckert R, Sajith S, Alperin K, Lakhotia A, Kohler K, Reid MJ, Davis MC, Fillingim RB. Sleep and circadian rhythm disturbances as risk and progression factors for multiple chronic overlapping pain conditions: a protocol for a longitudinal study. Pain Rep 2024; 9:e1194. [PMID: 39465006 PMCID: PMC11512637 DOI: 10.1097/pr9.0000000000001194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/09/2024] [Accepted: 07/20/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Chronic overlapping pain conditions (COPCs), such as chronic low back pain (cLBP) and fibromyalgia, frequently cooccur and incur substantial healthcare costs. However, to date, much focus has been placed on individual anatomically based chronic pain conditions, whereas little is known about the mechanisms underlying progression to multiple (more than 1) COPCs. This study aims to address the gap by investigating the role of common and modifiable risk factors, specifically sleep and circadian rhythm disturbances, in the development of multiple COPCs. Methods The study will enroll 300 participants with cLBP, including 200 with cLBP only and 100 with cLBP plus other COPCs (ie, fibromyalgia, temporomandibular disorders, irritable bowel syndrome, and chronic headaches) and follow them up for 12 months. Sleep and circadian rhythms will be assessed using wireless sleep electroencephalography, 24-hour evaluation of the rhythm of urinary 6-sulfatoxymelatonin, actigraphy, and sleep diaries. Pain amplification using quantitative sensory testing, psychological distress using validated self-report measures, and the number of pain sites using a pain body map will also be assessed. Perspectives This research aims to (1) comprehensively characterize sleep/circadian disturbances in individuals with single and multiple COPCs using multimodal in-home assessments; (2) examine the associations between sleep/circadian disturbances, changes in pain amplification, and psychological distress; and (3) investigate the relationship among these factors and the progression in the number of pain sites, a proxy for multiple COPCs. The findings will provide insights into the mechanisms leading to multiple COPCs, potentially informing treatment and prevention strategies for these complex conditions.
Collapse
Affiliation(s)
- Chung Jung Mun
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Shawn D. Youngstedt
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Megan E. Petrov
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Keenan A. Pituch
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Jeffrey A. Elliott
- Center for Circadian Biology and Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Steven Z. George
- Departments of Orthopaedic Surgery and Population Health Sciences, Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Frank LoVecchio
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Aram S. Mardian
- Chronic Pain Wellness Center, Phoenix VA Health Care System, Phoenix, AZ, USA
- Department of Family, Community and Preventive Medicine, University of Arizona College of Medicine–Phoenix, Phoenix, AZ, USA
| | - Kit K. Elam
- Department of Applied Health Science, Indiana University, Bloomington, IN, USA
| | - Nina Winsick
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Ryan Eckert
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Surabhi Sajith
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Kate Alperin
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Ananya Lakhotia
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Kaylee Kohler
- Emergency Department, HonorHealth Deer Valley Medical Center, Phoenix, AZ, USA
| | - Matthew J. Reid
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mary C. Davis
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - Roger B. Fillingim
- Department of Community Dentistry and Behavioral Science, University of Florida, Gainesville, FL, USA
| |
Collapse
|
4
|
Bangash MA, Cubuk C, Iseppon F, Haroun R, Garcia C, Luiz AP, Arcangeletti M, Gossage SJ, Santana-Varela S, Cox JJ, Lewis MJ, Wood JN, Zhao J. Analgesic targets identified in mouse sensory neuron somata and terminal pain translatomes. Cell Rep 2024; 43:114614. [PMID: 39163201 DOI: 10.1016/j.celrep.2024.114614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/07/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
The relationship between transcription and protein expression is complex. We identified polysome-associated RNA transcripts in the somata and central terminals of mouse sensory neurons in control, painful (plus nerve growth factor), and pain-free conditions (Nav1.7-null mice). The majority (98%) of translated transcripts are shared between male and female mice in both the somata and terminals. Some transcripts are highly enriched in the somata or terminals. Changes in the translatome in painful and pain-free conditions include novel and known regulators of pain pathways. Antisense knockdown of selected somatic and terminal polysome-associated transcripts that correlate with pain states diminished pain behavior. Terminal-enriched transcripts included those encoding synaptic proteins (e.g., synaptotagmin), non-coding RNAs, transcription factors (e.g., Znf431), proteins associated with transsynaptic trafficking (HoxC9), GABA-generating enzymes (Gad1 and Gad2), and neuropeptides (Penk). Thus, central terminal translation may well be a significant regulatory locus for peripheral input from sensory neurons.
Collapse
Affiliation(s)
- M Ali Bangash
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Cankut Cubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Rayan Haroun
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Chloe Garcia
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Ana P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Manuel Arcangeletti
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Samuel J Gossage
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK.
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK.
| |
Collapse
|
5
|
Huang ZN, Wang J, Wang ZY, Min LY, Ni HL, Han YL, Tian YY, Cui YZ, Han JX, Cheng XF. SR9009 attenuates inflammation-related NPMSC pyroptosis and IVDD through NR1D1/NLRP3/IL-1β pathway. iScience 2024; 27:109733. [PMID: 38689641 PMCID: PMC11059531 DOI: 10.1016/j.isci.2024.109733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/29/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024] Open
Abstract
Intervertebral disc is a highly rhythmical tissue. As a key factor linking biorhythm and inflammatory response, the shielding effect of NR1D1 in the process of intervertebral disc degeneration remains unclear. Here, we first confirmed that NR1D1 in the nucleus pulposus tissue presents periodic rhythmic changes and decreases in expression with intervertebral disc degeneration. Second, when NR1D1 was activated by SR9009 in vitro, NLRP3 inflammasome assembly and IL-1β production were inhibited, while ECM synthesis was increased. Finally, the vivo experiments further confirmed that the activation of NR1D1 can delay the process of disc degeneration to a certain extent. Mechanistically, we demonstrate that NR1D1 can bind to IL-1β and NLRP3 promoters, and that the NR1D1/NLRP3/IL-1β pathway is involved in this process. Our results demonstrate that the activation of NR1D1 can effectively reduce IL-1β secretion, alleviate LPS-induced NPMSC pyroptosis, and protect ECM degeneration.
Collapse
Affiliation(s)
- Ze-Nan Huang
- Department of Orthopedics, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 200072, Shandong Province, China
| | - Jing Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 200072, Shandong Province, China
| | - Ze-yu Wang
- Department of Spine Surgery, Affiliated Nanjing Jiangbei Hospital of Xinglin College, Nantong University, Nanjing 210019, Jiangsu Province, China
| | - Ling-yuan Min
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 200072, Shandong Province, China
| | - Hai-Ling Ni
- Department of Orthopedics, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 200072, Shandong Province, China
| | - Yan-Ling Han
- Medical Experimental Research Center, Yangzhou University, Yangzhou 225001, Jiangsu Province, China
| | - You-yue Tian
- Department of Pharmacy, University of South China, Hengyang 421001, Hunan Province, China
| | - Ya-Zhou Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 200072, Shandong Province, China
| | - Jing-Xiang Han
- Department of Orthopedics, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 200072, Shandong Province, China
| | - Xiao-Fei Cheng
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People’s Hospital affiliated to Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
6
|
Ko JY, Wang FS, Lian WS, Fang HC, Kuo SJ. Cartilage-specific knockout of miRNA-128a expression normalizes the expression of circadian clock genes (CCGs) and mitigates the severity of osteoarthritis. Biomed J 2024; 47:100629. [PMID: 37453588 PMCID: PMC10979161 DOI: 10.1016/j.bj.2023.100629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 06/05/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Micro-ribonucleic acids (miRNAs) are involved in osteoarthritis (OA) pathogenesis and clock-controlled genes (CCGs) regulation. However, the interaction between miRNAs and CCGs remains unclear. METHODS Human OA samples were used to assess CCGs expression. Cartilage-specific miR-128a knockout mouse model was established to investigate miR-128a's role in OA pathogenesis. Destabilization of the medial meniscus (DMM) model was employed to simulate OA. RESULTS Transcription levels of nuclear receptor subfamily 1 group D member 2 (NR1D2) were lower in both human OA samples and wild-type mice undergoing DMM compared to non-OA counterparts. MiR-128a knockout mice showed reduced disturbances in micro-computed tomographic and kinematic parameters following DMM, as well as less severe histologic cartilage loss. Immunohistochemistry staining revealed a lesser decrease in NR1D2-positive chondrocytes after DMM in miR-128a knockout mice than in wild-type mice. NR1D2 agonist rescued the suppressed expression of cartilage anabolic factors and extracellular matrix deposition caused by miR-128a precursor. CONCLUSIONS Cartilage-specific miR-128a knockout mice exhibited reduced severity, less disrupted kinematic parameters, and suppressed NR1D2 expression after DMM. NR1D2 enhanced the expression of cartilage anabolic factors and extracellular matrix deposition. These findings highlight the potential of employing miR-128a and CCG-targeted therapy for knee OA.
Collapse
Affiliation(s)
- Jih-Yang Ko
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Feng-Sheng Wang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wei-Shiung Lian
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsiao-Chi Fang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shu-Jui Kuo
- School of Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
7
|
Ma K, Pham T, Wang J, O-Sullivan I, DiCamillo A, Du S, Mwale F, Farooqui Z, Votta-Velis G, Bruce B, van Wijnen AJ, Liu Y, Im HJ. Nanoparticle-based inhibition of vascular endothelial growth factor receptors alleviates osteoarthritis pain and cartilage damage. SCIENCE ADVANCES 2024; 10:eadi5501. [PMID: 38354243 PMCID: PMC10866538 DOI: 10.1126/sciadv.adi5501] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Osteoarthritis (OA) is characterized by cartilage damage, inflammation, and pain. Vascular endothelial growth factor receptors (VEGFRs) have been associated with OA severity, suggesting that inhibitors targeting these receptors alleviate pain (via VEGFR1) or cartilage degeneration (via VEGFR2). We have developed a nanoparticle-based formulation of pazopanib (Votrient), an FDA-approved anticancer drug that targets both VEGFR1 and VEGFR2 (Nano-PAZII). We demonstrate that a single intraarticular injection of Nano-PAZII can effectively reduce joint pain for a prolonged time without substantial side effects in two different preclinical OA rodent models involving either surgical (upon partial medial meniscectomy) or nonsurgical induction (with monoiodoacetate). The injection of Nano-PAZII blocks VEGFR1 and relieves OA pain by suppressing sensory neuronal ingrowth into the knee synovium and neuronal plasticity in the dorsal root ganglia and spinal cord. Simultaneously, the inhibition of VEGFR2 reduces cartilage degeneration. These findings provide a mechanism-based disease-modifying drug strategy that addresses both pain symptoms and cartilage loss in OA.
Collapse
Affiliation(s)
- Kaige Ma
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Tiep Pham
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Jun Wang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - InSug O-Sullivan
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Amy DiCamillo
- Melior Discovery Inc., 869 Springdale Drive 500, Exton, PA 19341, USA
| | - Shiyu Du
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Fackson Mwale
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, McGill University, Montreal, Canada
| | - Zeba Farooqui
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Gina Votta-Velis
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Benjamin Bruce
- Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, IL 60612, USA
| | - Andre J. van Wijnen
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Ying Liu
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hee-Jeong Im
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, IL 60612, USA
| |
Collapse
|
8
|
Hashizume H, Motonari H, Yamamoto K, Nakamura Y, Hisaoka-Nakashima K, Morioka N. Stimulation of nuclear receptor REV-ERBs alleviates monosodium iodoacetate-induced osteoarthritis pathology of mice and the induction of inflammatory molecules expression in primary cultured chondrocytes. Int Immunopharmacol 2024; 127:111349. [PMID: 38086272 DOI: 10.1016/j.intimp.2023.111349] [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: 09/21/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
Because inflammation in chondrocytes contributes to the induction of osteoarthritis (OA), regulation of their activity is essential. A previous study showed that stimulation of the reverse erythroblastosis virus (REV-ERB) nuclear receptors in spinal glial cells elicits anti-inflammatory and antinociception effects in animal models of chronic pain. However, the involvement of REV-ERBs in chondrocyte functions and OA pathologies remains to be elucidated. In the current study, we found that pretreatment with the REV-ERB agonist SR9009 significantly blocked the increases in inflammatory molecules [(matrix metalloproteinase (MMP) 3, MMP9, and MMP13] and cytokines (interleukin-1β and tumor necrosis factor) in primary cultured chondrocytes following treatment with lipopolysaccharide. Furthermore, repeated intra-articular treatment with SR9009 significantly prevented monosodium iodoacetate-induced mechanical hypersensitivity and tended to partially reduce knee joint damage in mice. In conclusion, our findings suggest that REV-ERBs have a critical role in alleviating nociceptive hypersensitivity in OA pathologies by negatively regulating inflammation in chondrocytes.
Collapse
Affiliation(s)
- Hiroki Hashizume
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Hatsune Motonari
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kenta Yamamoto
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| |
Collapse
|
9
|
Mun CJ, Winsick N, Wegener ST, Youngstedt SD, Campbell CM, Aaron RV. Medium- and Long-Term Effects of Insomnia Severity and Circadian Preference on Pain and Emotional Distress Among Individuals With Chronic Pain. THE JOURNAL OF PAIN 2023; 24:1946-1956. [PMID: 37286095 PMCID: PMC10615674 DOI: 10.1016/j.jpain.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/21/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Studies have identified insomnia as having significant influence on chronic pain. A rising body of research has also underscored the association between eveningness and chronic pain. However, co-assessment of insomnia and eveningness in the context of chronic pain adjustment has been limited. The present study sought to investigate the effects of insomnia and eveningness on pain severity, pain interference, and emotional distress (ie, depressive and anxiety symptoms) over nearly 2 years among adults with chronic pain in the U.S. Adults with chronic pain (N = 884) were surveyed 3 times via Amazon's MTurk online crowdsourcing platform: baseline, 9-month follow-up, and 21-month follow-up. Path analysis was conducted to examine the effects of baseline insomnia severity (Insomnia Severity Index) and eveningness (Morningness and Eveningness Questionnaire), as well as their moderating effects on outcomes. Controlling for select sociodemographic variables and baseline outcome levels, greater insomnia severity at baseline was associated with worsening of all of the pain-related outcomes at 9-month follow-up, and pain interreference and emotional distress at 21-month follow-up. We did not find evidence that evening types are at a higher risk of experiencing worsening pain-related outcomes over time compared to morning and intermediate types. There were also no significant insomnia severity and eveningness moderation effects on any outcome. Our findings suggest that insomnia is a more robust predictor of changes in pain-related outcomes as compared to eveningness. Treatment of insomnia can be important in chronic pain management. Future studies should evaluate the role of circadian misalignment on pain using more accurate biobehavioral makers. PERSPECTIVE: This study examined the effects of insomnia and eveningness on pain and emotional distress in a large sample of individuals with chronic pain. Insomnia severity is a stronger predictor of changes in pain and emotional distress than eveningness, highlighting insomnia as an important clinical target for chronic pain management.
Collapse
Affiliation(s)
- Chung Jung Mun
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD.
| | - Nina Winsick
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
| | - Stephen T Wegener
- Department of Physical Medicine and Rehabilitation, Johns Hopkins School of Medicine, Baltimore, MD
| | - Shawn D Youngstedt
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
| | - Claudia M Campbell
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Rachel V Aaron
- Department of Physical Medicine and Rehabilitation, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
10
|
Bumgarner JR, McCray EW, Nelson RJ. The disruptive relationship among circadian rhythms, pain, and opioids. Front Neurosci 2023; 17:1109480. [PMID: 36875657 PMCID: PMC9975345 DOI: 10.3389/fnins.2023.1109480] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
Pain behavior and the systems that mediate opioid analgesia and opioid reward processing display circadian rhythms. Moreover, the pain system and opioid processing systems, including the mesolimbic reward circuitry, reciprocally interact with the circadian system. Recent work has demonstrated the disruptive relationship among these three systems. Disruption of circadian rhythms can exacerbate pain behavior and modulate opioid processing, and pain and opioids can influence circadian rhythms. This review highlights evidence demonstrating the relationship among the circadian, pain, and opioid systems. Evidence of how disruption of one of these systems can lead to reciprocal disruptions of the other is then reviewed. Finally, we discuss the interconnected nature of these systems to emphasize the importance of their interactions in therapeutic contexts.
Collapse
Affiliation(s)
- Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Evan W McCray
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
11
|
Qian Z, Liu Z, Feng Z, Cai Z, Qiu Y, Zhu Z. Blocking circadian clock factor Rev-erbα inhibits growth plate chondrogenesis via up-regulating MAPK-ERK1/2 pathway. Cell Cycle 2023; 22:73-84. [PMID: 35938533 PMCID: PMC9769450 DOI: 10.1080/15384101.2022.2109106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence indicated circadian clock gene Rev-erbα was involved in cartilage metabolism, however the contribution of Rev-erbα to growth plate chondrogenesis remains unknown. Here, we found that Rev-erbα exhibited the spatiotemporal expression model in growth plate. Moreover, Rev-erbα antagonist SR8278 inhibited longitudinal elongation of metatarsal bone ex vivo. And morphological analysis exhibited SR8278 led to the reduced height of growth plate and hypertrophic zone. Furthermore, blocking Rev-erbα suppressed the proliferation and hypertrophic differentiation of chondrocytes in growth plate. Similarly, knock-down Rev-erbα inhibited the proliferation and differentiation of primary chondrocytes in vitro. The mechanistic study indicated that knock-down Rev-erbα up-regulated MAPK-ERK1/2 pathway in chondrocytes. However, restraint of MAPK-ERK1/2 pathway alleviated partially SR8278-inhibited longitudinal elongation of metatarsal bone and growth plate development. Therefore, our results provide evidence of the vital role of Rev-erbα on growth plate chondrogenesis.
Collapse
Affiliation(s)
- Zhuang Qian
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhen Liu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhenhua Feng
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhenning Cai
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China,Yong Qiu Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China,CONTACT Zezhang Zhu
| |
Collapse
|
12
|
Zhu Y, Liu Y, Escames G, Yang Z, Zhao H, Qian L, Xue C, Xu D, Acuña-Castroviejo D, Yang Y. Deciphering clock genes as emerging targets against aging. Ageing Res Rev 2022; 81:101725. [PMID: 36029999 DOI: 10.1016/j.arr.2022.101725] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/21/2022] [Accepted: 08/22/2022] [Indexed: 01/31/2023]
Abstract
The old people often suffer from circadian rhythm disturbances, which in turn accelerate aging. Many aging-related degenerative diseases such as Alzheimer's disease, Parkinson's disease, and osteoarthritis have an inextricable connection with circadian rhythm. In light of the predominant effects of clock genes on regulating circadian rhythm, we systematically present the elaborate network of roles that clock genes play in aging in this review. First, we briefly introduce the basic background regarding clock genes. Second, we systemically summarize the roles of clock genes in aging and aging-related degenerative diseases. Third, we discuss the relationship between clock genes polymorphisms and aging. In summary, this review is intended to clarify the indispensable roles of clock genes in aging and sheds light on developing clock genes as anti-aging targets.
Collapse
Affiliation(s)
- Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Germaine Escames
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Avda. del Conocimiento s/n, Granada, Spain; Ibs. Granada and CIBERfes, Granada, Spain; UGC of Clinical Laboratories, Universitu San Cecilio's Hospital, Granada, Spain
| | - Zhi Yang
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, 1 Xinsi Road, Xi'an, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, 1 Xinsi Road, Xi'an, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Chengxu Xue
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Danni Xu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Darío Acuña-Castroviejo
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, Avda. del Conocimiento s/n, Granada, Spain; Ibs. Granada and CIBERfes, Granada, Spain; UGC of Clinical Laboratories, Universitu San Cecilio's Hospital, Granada, Spain.
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
13
|
Hosokawa H, Akagi R, Watanabe S, Horii M, Shinohara M, Mikami Y, Toguchi K, Kimura S, Yamaguchi S, Ohtori S, Sasho T. Nuclear receptor subfamily 1 group D member 1 in the pathology of obesity-induced osteoarthritis progression. J Orthop Res 2022; 41:930-941. [PMID: 36102152 DOI: 10.1002/jor.25440] [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: 04/10/2022] [Revised: 07/28/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
Mechanical overload and chemical factors are both related to obesity-induced progression of knee osteoarthritis. The circadian rhythm is related to the development of metabolic syndrome and the progression of osteoarthritis, and the core clock genes nuclear receptor subfamily 1 group D member 1 (NR1D1) and brain and muscle arnt-like protein 1 (BMAL1) are dysregulated in cartilage from patients with osteoarthritis. Here, we focused on NR1D1 and investigated osteoarthritis-related changes and gene expression in a mouse model of diet-induced obesity. A high-fat diet was provided to C57BL6/J mice, and changes in body weight, blood lipids, and gene expression were investigated. Destabilization of the medial meniscus or sham surgery was performed on mice fed a high-fat diet or normal diet, and histological osteoarthritis-related changes and NR1D1 expression were investigated. The effects of the NR1D1 agonist SR9009 were also assessed. Mice fed a high-fat diet developed significant obesity and dyslipidemia. Nr1d1 and Bmal1 gene expression levels decreased in the liver and knee joints. Moreover, increased osteoarthritis progression and decreased NR1D1 protein expression were observed in high-fat diet-fed mice after surgical osteoarthritis induction. SR9009 decreased the progression of obesity, dyslipidemia, and osteoarthritis. Overall, obesity and dyslipidemia induced by the high-fat diet led to osteoarthritis progression and decreased NR1D1 expression. Thus, NR1D1 may play an important role in obesity-induced osteoarthritis.
Collapse
Affiliation(s)
- Hiroaki Hosokawa
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan.,Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| | - Ryuichiro Akagi
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Shotaro Watanabe
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan.,Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| | - Manato Horii
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Masashi Shinohara
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Yukio Mikami
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Kaoru Toguchi
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Seiji Kimura
- Department of Orthopaedic Surgery, Center for Advanced Joint Function and Reconstructive Spine Surgery Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Yamaguchi
- Graduate School of Global and Transdisciplinary Studies, Chiba University, Chiba, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Takahisa Sasho
- Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| |
Collapse
|
14
|
Mun CJ, Burgess HJ, Sears DD, Parthasarathy S, James D, Altamirano U, Sajith S, Lakhotia A, Fillingim RB, Youngstedt SD. Circadian Rhythm and Pain: a Review of Current Research and Future Implications. CURRENT SLEEP MEDICINE REPORTS 2022. [DOI: 10.1007/s40675-022-00228-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
15
|
He T, Pang S, Wang H, Yun H, Hao X, Jia L, Liu H, Wang D, Wang D, Xu H, Jie Q, Yang L, Zheng C. Drugging the circadian clock feedback cycle to ameliorate cartilage degeneration. FEBS J 2022; 289:6643-6658. [DOI: 10.1111/febs.16601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Ting He
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research Northwestern Polytechnical University Xi'an China
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Siyi Pang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research Northwestern Polytechnical University Xi'an China
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Huanbo Wang
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Haitao Yun
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Xue Hao
- Department of Pediatric Orthopedic, Honghui Hospital, Xi'an Jiaotong University College of Medicine Xi'an China
| | - Liyuan Jia
- Laboratory of Tissue Engineering, College of Life Science Northwest University Xi'an China
| | - He Liu
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Di Wang
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Dong Wang
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Huiyun Xu
- School of Life Sciences Northwestern Polytechnical University Xi'an China
| | - Qiang Jie
- Department of Pediatric Orthopedic, Honghui Hospital, Xi'an Jiaotong University College of Medicine Xi'an China
| | - Liu Yang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research Northwestern Polytechnical University Xi'an China
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| | - Chao Zheng
- Institute of Orthopedic Surgery, Xijing Hospital Fourth Military Medical University Xi'an China
| |
Collapse
|
16
|
Kaur S, Hickman TM, Lopez-Ramirez A, McDonald H, Lockhart LM, Darwish O, Averitt DL. Estrogen modulation of the pronociceptive effects of serotonin on female rat trigeminal sensory neurons is timing dependent and dosage dependent and requires estrogen receptor alpha. Pain 2022; 163:e899-e916. [PMID: 35121697 PMCID: PMC9288423 DOI: 10.1097/j.pain.0000000000002604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/28/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT The role of the major estrogen estradiol (E2) on orofacial pain conditions remains controversial with studies reporting both a pronociceptive and antinociceptive role of E2. E2 modulation of peripheral serotonergic activity may be one mechanism underlying the female prevalence of orofacial pain disorders. We recently reported that female rats in proestrus and estrus exhibit greater serotonin (5HT)-evoked orofacial nocifensive behaviors compared with diestrus and male rats. Further coexpression of 5HT 2A receptor mRNA in nociceptive trigeminal sensory neurons that express transient receptor potential vanilloid 1 ion channels contributes to pain sensitization. E2 may exacerbate orofacial pain through 5HT-sensitive trigeminal nociceptors, but whether low or high E2 contributes to orofacial pain and by what mechanism remains unclear. We hypothesized that steady-state exposure to a proestrus level of E2 exacerbates 5HT-evoked orofacial nocifensive behaviors in female rats, explored the transcriptome of E2-treated female rats, and determined which E2 receptor contributes to sensitization of female trigeminal sensory neurons. We report that a diestrus level of E2 is protective against 5HT-evoked orofacial pain behaviors, which increase with increasing E2 concentrations, and that E2 differentially alters several pain genes in the trigeminal ganglia. Furthermore, E2 receptors coexpressed with 5HT 2A and transient receptor potential vanilloid 1 and enhanced capsaicin-evoked signaling in the trigeminal ganglia through estrogen receptor α. Overall, our data indicate that low, but not high, physiological levels of E2 protect against orofacial pain, and we provide evidence that estrogen receptor α receptor activation, but not others, contributes to sensitization of nociceptive signaling in trigeminal sensory neurons.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Department of Biology, Texas Woman’s University, Denton, TX 76204
| | | | | | - Hanna McDonald
- Department of Biology, Texas Woman’s University, Denton, TX 76204
| | | | - Omar Darwish
- Department of Mathematics and Computer Science, Texas Woman’s University, Denton, TX 76204
| | | |
Collapse
|
17
|
Song X, Zhao M, Tang J, Ma T, Bai H, Wang X, Liu L, Li T, Xu X, Sheng X, Zhao B, Wang Y, Wang T, Guo Y, Zhang X, Gao L. Dark-light cycle disrupts bone metabolism and suppresses joint deterioration in osteoarthritic rats. Arthritis Res Ther 2022; 24:158. [PMID: 35765090 PMCID: PMC9238010 DOI: 10.1186/s13075-022-02832-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/05/2022] [Indexed: 01/03/2023] Open
Abstract
Background Light alteration affects the internal environment and metabolic homeostasis of the body through circadian rhythm disorders (CRD). CRD is one of the factors that induce and accelerate osteoarthritis (OA). Therefore, the aim of this study was to evaluate the effects of continuous dark-light (DL) cycle on joint inflammation, bone structure, and metabolism in normal and OA Sprague-Dawley (SD) rats. Methods Interleukin (IL)-1β, IL-6, inducible nitric oxide synthase (iNOS), and tumor necrosis factor (TNF)-α were used to evaluate the systemic inflammation in rats. The pathological changes and inflammatory reactions of the cartilage and synovium of the knee joint in rats were evaluated by Safranin O-fast green and immunological staining. Bone turnover was assessed by histomorphometry and μCT scanning, as well as bone metabolism markers and proteins. The expression changes of clock proteins BMAL1, NR1D1, PER3, and CRY1 in representative tissues were detected by western blotting. Results DL cycle significantly inhibited body weight gain in normal and OA rats. The levels of proinflammatory factors in the peripheral blood circulation and degradation enzymes in the cartilage were significantly decreased in OA+DL rats. DL cycle significantly destroyed the structure of subchondral bone in hindlimbs of OA rats and reduced trabecular bone numbers. The decrease of bone mineral density (BMD), percent bone volume with respect to total bone volume (BV/TV), trabecular number (TB.N), osteoclast number, and mineralization could also be found. The ratio of the receptor activator of nuclear factor-kappa B ligand/osteoprotegerin (RANKL/OPG) in the bone marrow of OA rats was markedly increased under DL, along with the activation of the mononuclear/phagocyte system. The expression of representative clock proteins and genes BMAL1, PER3, and CRY1 were markedly changed in the tissues of OA+DL rats. Conclusions These results suggested that DL cycle dampened the arthritis and promoted bone resorption and bone mass loss. Graphical abstract DL cycle affects bone turnover by regulating osteoclast production in osteoarthritic rats.![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02832-8.
Collapse
|
18
|
Lactobacillus acidophilus Mitigates Osteoarthritis-Associated Pain, Cartilage Disintegration and Gut Microbiota Dysbiosis in an Experimental Murine OA Model. Biomedicines 2022; 10:biomedicines10061298. [PMID: 35740320 PMCID: PMC9220766 DOI: 10.3390/biomedicines10061298] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
To test probiotic therapy for osteoarthritis (OA), we administered Lactobacillus acidophilus (LA) by oral gavage (2×/week) after induction of OA by partial medial meniscectomy (PMM). Pain was assessed by von Frey filament and hot plate testing. Joint pathology and pain markers were comprehensively analyzed in knee joints, spinal cords, dorsal root ganglia and distal colon by Safranin O/fast green staining, immunofluorescence microscopy and RT-qPCR. LA acutely reduced inflammatory knee joint pain and prevented further OA progression. The therapeutic efficacy of LA was supported by a significant reduction of cartilage-degrading enzymes, pain markers and inflammatory factors in the tissues we examined. This finding suggests a likely clinical effect of LA on OA. The effect of LA treatment on the fecal microbiome was assessed by 16S rRNA gene amplicon sequencing analysis. LA significantly altered the fecal microbiota compared to vehicle-treated mice (PERMANOVA p < 0.009). Our pre-clinical OA animal model revealed significant OA disease modifying effects of LA as reflected by rapid joint pain reduction, cartilage protection, and reversal of dysbiosis. Our findings suggest that LA treatment has beneficial systemic effects that can potentially be developed as a safe OA disease-modifying drug (OADMD).
Collapse
|
19
|
Peng Y, Zhang Q, Cheng H, Yan G, Xing C. Upregulation of ubiquitin conjugating enzyme E2B (Ube2b) ameliorates neuropathic pain by regulating Kcna2 (potassium voltage-gated channel subfamily A member 2) in primary afferent neurons. Bioengineered 2021; 12:7470-7480. [PMID: 34632937 PMCID: PMC8806678 DOI: 10.1080/21655979.2021.1976895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Neuropathic pain is a kind of pain caused by damage to somatosensory nervous system. Currently, neuropathic pain is still a medical problem for clinicians. Ubiquitin conjugating enzyme E2B (Ube2b) is validated to be implicated with nerve function, but whether Ube2b can play a role in neuropathic pain is still elusive. In this work, we constructed chronic constriction injury (CCI) rat model by ligating the left sciatic nerve, Ube2b protein expression was confirmed to be decreased in spinal cord tissues of CCI rats via Western blot analysis and immunofluorescence (IF) staining. Moreover, Ube2b elevation alleviated the thermal hyperalgesia and mechanical hyperalgesia in CCI rats according to paw withdrawal thermal latency (PWTL) and paw withdrawal mechanic threshold (PWMT). In addition, Hematoxylin-eosin staining revealed that Ube2b elevation suppressed chronic sciatic nerve injury. All these data suggested that Ube2b could ameliorate neuropathic pain in CCI rats. Mechanically, Ube2b upregulation elevated the protein level of Kcna2 (potassium voltage-gated channel subfamily A member 2) and decreased the protein level of DNMT3a (DNA methyltransferase 3 alpha). Ube2b elevation could increase Kcna2 expression via suppressing DNMT3a. Rescue assays unveiled that Ube2b overexpression modulated-mechanical hyperalgesia and thermal hyperalgesia were reversed by Kcna2 depletion, indicating that Ube2b alleviated neuropathic pain via mediating Kcna2 via the regulation of DNMT3a. In summary, we found that Ube2b elevation ameliorated neuropathic pain through regulating Kcna2, which might offer a novel biomarker for the therapies of neuropathic pain.
Collapse
Affiliation(s)
- Yuanzhi Peng
- Department of Anesthesiology and SICU, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai,China
| | - Qingqing Zhang
- Department of Anesthesiology and SICU, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai,China
| | - Hao Cheng
- Department of Cardiovascular Surgery, Changhai Hospital, the Navy Medical University, Shanghai, China
| | - Guizhen Yan
- Department of Neurology, People's Hospital of Lixia District of Jinan, Jinan, Shandong, China
| | - Chunli Xing
- Department of Neurology, People's Hospital of Lixia District of Jinan, Jinan, Shandong, China
| |
Collapse
|
20
|
Bumgarner JR, Walker WH, Nelson RJ. Circadian rhythms and pain. Neurosci Biobehav Rev 2021; 129:296-306. [PMID: 34375675 PMCID: PMC8429267 DOI: 10.1016/j.neubiorev.2021.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 06/30/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
The goal of this review is to provide a perspective on the nature and importance of the relationship between the circadian and pain systems. We provide: 1) An overview of the circadian and pain systems, 2) a review of direct and correlative evidence that demonstrates diurnal and circadian rhythms within the pain system; 3) a perspective highlighting the need to consider the role of a proposed feedback loop of circadian rhythm disruption and maladaptive pain; 4) a perspective on the nature of the relationship between circadian rhythms and pain. In summary, we propose that there is no single locus responsible for producing the circadian rhythms of the pain system. Instead, circadian rhythms of pain are a complex result of the distributed rhythms present throughout the pain system, especially those of the descending pain modulatory system, and the rhythms of the systems with which it interacts, including the opioid, endocrine, and immune systems.
Collapse
Affiliation(s)
- Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA.
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
21
|
Bumgarner JR, Nelson RJ. Implications of Disrupted Circadian Rhythms on Pain. JOURNAL OF PSYCHIATRY AND BRAIN SCIENCE 2021; 6:e210014. [PMID: 39324149 PMCID: PMC11423940 DOI: 10.20900/jpbs.20210014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Pain is regulated by circadian rhythms. Daily fluctuations in pain thresholds are observed in health and disease. Disruptions to the circadian and pain systems may initiate a detrimental feedback loop between the two systems. The relationship between the pain and circadian systems is briefly reviewed to highlight a perspective on the need to consider disrupted circadian rhythms in the treatment of pain.
Collapse
Affiliation(s)
- Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
22
|
Song X, Bai H, Meng X, Xiao J, Gao L. Drivers of phenotypic variation in cartilage: Circadian clock genes. J Cell Mol Med 2021; 25:7593-7601. [PMID: 34213828 PMCID: PMC8358851 DOI: 10.1111/jcmm.16768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/07/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Endogenous homeostasis and peripheral tissue metabolism are disrupted by irregular fluctuations in activation, movement, feeding and temperature, which can accelerate negative biological processes and lead to immune reactions, such as rheumatoid arthritis (RA) and osteoarthritis (OA). This review summarizes abnormal phenotypes in articular joint components such as cartilage, bone and the synovium, attributed to the deletion or overexpression of clock genes in cartilage or chondrocytes. Understanding the functional mechanisms of different genes, the differentiation of mouse phenotypes and the prevention of joint ageing and disease will facilitate future research.
Collapse
Affiliation(s)
- Xiaopeng Song
- College of Veterinary Medicine, Heilongjiang Key Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Hui Bai
- College of Veterinary Medicine, Heilongjiang Key Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Xinghua Meng
- College of Veterinary Medicine, Heilongjiang Key Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Jianhua Xiao
- College of Veterinary Medicine, Heilongjiang Key Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Li Gao
- College of Veterinary Medicine, Heilongjiang Key Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
23
|
Arora V, Singh G, O-Sullivan I, Ma K, Natarajan Anbazhagan A, Votta-Velis EG, Bruce B, Richard R, van Wijnen AJ, Im HJ. Gut-microbiota modulation: The impact of thegut-microbiotaon osteoarthritis. Gene 2021; 785:145619. [PMID: 33781857 DOI: 10.1016/j.gene.2021.145619] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is one of the most common medical conditions affecting > 300 million people globally which represents the formidable public health challenge. Despite its clinical and financial ramifications, there are currently no approved disease modifying OA drugs available and symptom palliation is the only alternative. Currently, the amount of data on the human intestinal microbiome is growing at a high rate, both in health and in various pathological conditions. With an increase in the amount of the accumulated data, there is an expanded understanding that the microbiome provides compelling evidence of a link between thegut microbiomeand development ofOA. The microbiota management tools of probiotics and/or prebiotics or symbiotic have been developed and indeed, commercialized over the past few decades with the expressed purpose of altering the microbiota within the gastrointestinal tract which could be a potentially novel intervention to tackle or prevent OA. However, the mechanisms how intestinal microbiota affects the OA pathogenesis are still not clear and further research targeting specific gut microbiota or its metabolites is still needed to advance OA treatment strategies from symptomatic management to individualized interventions of OA pathogenesis. This article provides an overview of the various preclinical and clinical studies using probiotics and prebiotics as plausible therapeutic options that can restore the gastrointestinal microbiota and its impact on the OA pathogenesis. May be in the near future the targeted alterations of gut microbiota may pave the way for developing new interventions to prevent and treat OA.
Collapse
Affiliation(s)
- Vipin Arora
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Gurjit Singh
- Departments of Bioengineering, the University of Illinois at Chicago, Chicago, IL, USA
| | - InSug O-Sullivan
- Departments of Medicine, the University of Illinois at Chicago, Chicago, IL, USA
| | - Kaige Ma
- Departments of Bioengineering, the University of Illinois at Chicago, Chicago, IL, USA
| | | | - E Gina Votta-Velis
- Departments of Anesthesiology, the University of Illinois at Chicago, Chicago, IL, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Benjamin Bruce
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ripper Richard
- Departments of Anesthesiology, the University of Illinois at Chicago, Chicago, IL, USA
| | | | - Hee-Jeong Im
- Departments of Bioengineering, the University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, IL, USA.
| |
Collapse
|
24
|
Andersen PAK, Petrenko V, Rose PH, Koomen M, Fischer N, Ghiasi SM, Dahlby T, Dibner C, Mandrup-Poulsen T. Proinflammatory Cytokines Perturb Mouse and Human Pancreatic Islet Circadian Rhythmicity and Induce Uncoordinated β-Cell Clock Gene Expression via Nitric Oxide, Lysine Deacetylases, and Immunoproteasomal Activity. Int J Mol Sci 2020; 22:E83. [PMID: 33374803 PMCID: PMC7795908 DOI: 10.3390/ijms22010083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic β-cell-specific clock knockout mice develop β-cell oxidative-stress and failure, as well as glucose-intolerance. How inflammatory stress affects the cellular clock is under-investigated. Real-time recording of Per2:luciferase reporter activity in murine and human pancreatic islets demonstrated that the proinflammatory cytokine interleukin-1β (IL-1β) lengthened the circadian period. qPCR-profiling of core clock gene expression in insulin-producing cells suggested that the combination of the proinflammatory cytokines IL-1β and interferon-γ (IFN-γ) caused pronounced but uncoordinated increases in mRNA levels of multiple core clock genes, in particular of reverse-erythroblastosis virus α (Rev-erbα), in a dose- and time-dependent manner. The REV-ERBα/β agonist SR9009, used to mimic cytokine-mediated Rev-erbα induction, reduced constitutive and cytokine-induced brain and muscle arnt-like 1 (Bmal1) mRNA levels in INS-1 cells as expected. SR9009 induced reactive oxygen species (ROS), reduced insulin-1/2 (Ins-1/2) mRNA and accumulated- and glucose-stimulated insulin secretion, reduced cell viability, and increased apoptosis levels, reminiscent of cytokine toxicity. In contrast, low (<5,0 μM) concentrations of SR9009 increased Ins-1 mRNA and accumulated insulin-secretion without affecting INS-1 cell viability, mirroring low-concentration IL-1β mediated β-cell stimulation. Inhibiting nitric oxide (NO) synthesis, the lysine deacetylase HDAC3 and the immunoproteasome reduced cytokine-mediated increases in clock gene expression. In conclusion, the cytokine-combination perturbed the intrinsic clocks operative in mouse and human pancreatic islets and induced uncoordinated clock gene expression in INS-1 cells, the latter effect associated with NO, HDAC3, and immunoproteasome activity.
Collapse
Affiliation(s)
- Phillip Alexander Keller Andersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark; (P.A.K.A.); (P.H.R.); (M.K.); (N.F.); (S.M.G.); (T.D.)
| | - Volodymyr Petrenko
- Division of Endocrinology, Diabetes, Nutrition and Patient Education, Department of Cell Physiology and Metabolism, Diabetes Center, Faculty of Medicine, University of Geneva, D05.2147c Rue Michel-Servet, 1 CH-1211 Geneva 4, Switzerland; (V.P.); (C.D.)
| | - Peter Horskjær Rose
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark; (P.A.K.A.); (P.H.R.); (M.K.); (N.F.); (S.M.G.); (T.D.)
| | - Melissa Koomen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark; (P.A.K.A.); (P.H.R.); (M.K.); (N.F.); (S.M.G.); (T.D.)
| | - Nico Fischer
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark; (P.A.K.A.); (P.H.R.); (M.K.); (N.F.); (S.M.G.); (T.D.)
| | - Seyed Mojtaba Ghiasi
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark; (P.A.K.A.); (P.H.R.); (M.K.); (N.F.); (S.M.G.); (T.D.)
| | - Tina Dahlby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark; (P.A.K.A.); (P.H.R.); (M.K.); (N.F.); (S.M.G.); (T.D.)
| | - Charna Dibner
- Division of Endocrinology, Diabetes, Nutrition and Patient Education, Department of Cell Physiology and Metabolism, Diabetes Center, Faculty of Medicine, University of Geneva, D05.2147c Rue Michel-Servet, 1 CH-1211 Geneva 4, Switzerland; (V.P.); (C.D.)
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark; (P.A.K.A.); (P.H.R.); (M.K.); (N.F.); (S.M.G.); (T.D.)
| |
Collapse
|
25
|
A matter of time: Circadian clocks in osteoarthritis and the potential of chronotherapy. Exp Gerontol 2020; 143:111163. [PMID: 33227402 DOI: 10.1016/j.exger.2020.111163] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/29/2020] [Accepted: 11/14/2020] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is a common and debilitating joint disease which develops and progresses with age. Despite extensive research into the disease, potent disease-modifying drugs remain elusive. Changes to the character and function of chondrocytes of the articular cartilage underly the pathogenesis of OA. A recently emerging facet of chondrocyte biology that has been implicated in OA pathogenesis is the role of circadian rhythms, and the cellular clock which governs rhythmic gene transcription. Here, we review the role of the chondrocyte's cellular clock in governing normal homeostasis, and explore the wide range of consequences that contribute to OA development when the clock is dysregulated by aging and other factors. Finally, we explore how harnessing this understanding of clock mechanics in aging and OA can be translated into novel treatment strategies, or 'chronotherapies', for patients.
Collapse
|
26
|
Kim HK, Lee SY, Koike N, Kim E, Wirianto M, Burish MJ, Yagita K, Lee HK, Chen Z, Chung JM, Abdi S, Yoo SH. Circadian regulation of chemotherapy-induced peripheral neuropathic pain and the underlying transcriptomic landscape. Sci Rep 2020; 10:13844. [PMID: 32796949 PMCID: PMC7427990 DOI: 10.1038/s41598-020-70757-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022] Open
Abstract
Growing evidence demonstrates circadian rhythms of pain hypersensitivity in various chronic disorders. In chemotherapy-induced peripheral neuropathy (CIPN), agents such as paclitaxel are known to elicit chronic neuropathic pain in cancer patients and seriously compromise their quality of life. Here, we report that the mechanical threshold for allodynia in paclitaxel-treated rats exhibited a robust circadian oscillation, reaching the nadir during the daytime (inactive phase). Using Per2::LucSV circadian reporter mice expressing a PER2::LUC fusion protein, we isolated dorsal root ganglia (DRG), the primary sensory cell body for peripheral nerve injury generated hypersensitivity, and monitored ex vivo reporter bioluminescence. We observed strong circadian reporter rhythms in DRG neurons which are highly entrainable by external cues. Paclitaxel treatment significantly lengthened DRG circadian periods, with little effects on the amplitude of oscillation. We further observed the core protein BMAL1 and PER2 in DRG neurons and satellite cells. Using DRG and dorsal horn (DH; another key structure for CIPN pain response) tissues from vehicle and paclitaxel treated rats, we performed RNA-sequencing and identified diurnal expression of core clock genes as well as clock-controlled genes in both sites. Interestingly, 20.1% and 30.4% of diurnal differentially expressed genes (DEGs) overlapped with paclitaxel-induced DEGs in the DRG and the DH respectively. In contrast, paclitaxel-induced DEGs displayed only a modest overlap between daytime and nighttime (Zeitgeber Time 8 and 20). Furthermore, paclitaxel treatment induced de novo diurnal DEGs, suggesting reciprocal interaction of circadian rhythms and chemotherapy. Our study therefore demonstrates a circadian oscillation of CIPN and its underlying transcriptomic landscape.
Collapse
Affiliation(s)
- Hee Kee Kim
- Division of Anesthesiology, Critical Care and Pain Medicine, Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sun-Yeul Lee
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center At Houston, 6431 Fannin St., Houston, TX, 77030, USA
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea
| | - Nobuya Koike
- Department of Physiology and Systems Bioscience, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center At Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Marvin Wirianto
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center At Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Mark J Burish
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, 6400 Fannin St., Houston, TX, 77030, USA
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hyun Kyoung Lee
- Department of Pediatrics, Baylor College of Medicine, Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center At Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Jin Mo Chung
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Salahadin Abdi
- Division of Anesthesiology, Critical Care and Pain Medicine, Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center At Houston, 6431 Fannin St., Houston, TX, 77030, USA.
| |
Collapse
|
27
|
Relationship between Pain Behavior and Changes in KCNA2 Expression in the Dorsal Root Ganglia of Rats with Osteoarthritis. Pain Res Manag 2020. [DOI: 10.1155/2020/4636838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective. To investigate the relationship between pain behavior and potassium voltage-gated channel subfamily A member 2 (KCNA2) expression in dorsal root ganglia (DRGs) of rats with osteoarthritis (OA). Methods. Male Sprague-Dawley rats were randomly divided into three groups: blank control group (group C), normal saline group (group S), and group OA. Paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL) were measured one day before injection and one, two, four, and six weeks after injection. At one, two, four, and six weeks after injection, pathological knee joint changes and activated transcription factor-3 (ATF-3) and KCNA2 expressions in DRGs were analyzed. Results. Compared with preinjection, PWMT and PWTL at two, four, and six weeks after injection were significantly decreased in the group OA (P<0.05 or 0.01). Compared with group C, PWMT and PWTL at two, four, and six weeks after injection were significantly decreased in the group OA (P<0.05 or 0.01). In the group OA, slight local articular cartilage surface destruction was found at week one. The cartilage surface destruction gradually developed, and the exacerbation of cartilage matrix reduction and bone hyperplasia were increasingly aggravated and eventually evolved into advanced OA in the second to sixth weeks. Compared with group C, ATF-3 expression was significantly increased, and KCNA2 expression was significantly decreased in the group OA at two, four, and six weeks after injection (P<0.05 or 0.01). Compared to baseline, ATF-3 expression was significantly increased, and KCNA2 expression was significantly decreased in the group OA (P<0.05 or 0.01). Conclusion. Pain behavior in OA rats was associated with decreased KCNA2 expression in DRGs.
Collapse
|
28
|
|
29
|
Modeling the daily rhythm of human pain processing in the dorsal horn. PLoS Comput Biol 2019; 15:e1007106. [PMID: 31295266 PMCID: PMC6622484 DOI: 10.1371/journal.pcbi.1007106] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/14/2019] [Indexed: 12/21/2022] Open
Abstract
Experimental studies show that human pain sensitivity varies across the 24-hour day, with the lowest sensitivity usually occurring during the afternoon. Patients suffering from neuropathic pain, or nerve damage, experience an inversion in the daily modulation of pain sensitivity, with the highest sensitivity usually occurring during the early afternoon. Processing of painful stimulation occurs in the dorsal horn (DH), an area of the spinal cord that receives input from peripheral tissues via several types of primary afferent nerve fibers. The DH circuit is composed of different populations of neurons, including excitatory and inhibitory interneurons, and projection neurons, which constitute the majority of the output from the DH to the brain. In this work, we develop a mathematical model of the dorsal horn neural circuit to investigate mechanisms for the daily modulation of pain sensitivity. The model describes average firing rates of excitatory and inhibitory interneuron populations and projection neurons, whose activity is directly correlated with experienced pain. Response in afferent fibers to peripheral stimulation is simulated by a Poisson process generating nerve fiber spike trains at variable firing rates. Model parameters for fiber response to stimulation and the excitability properties of neuronal populations are constrained by experimental results found in the literature, leading to qualitative agreement between modeled responses to pain and experimental observations. We validate our model by reproducing the wind-up of pain response to repeated stimulation. We apply the model to investigate daily modulatory effects on pain inhibition, in which response to painful stimuli is reduced by subsequent non-painful stimuli. Finally, we use the model to propose a mechanism for the observed inversion of the daily rhythmicity of pain sensation under neuropathic pain conditions. Underlying mechanisms for the shift in rhythmicity have not been identified experimentally, but our model results predict that experimentally-observed dysregulation of inhibition within the DH neural circuit may be responsible. The model provides an accessible, biophysical framework that will be valuable for experimental and clinical investigations of diverse physiological processes modulating pain processing in humans. Human pain sensitivity follows a daily (∼24 hour) rhythm. In particular, humans experience the highest sensitivity to pain in the middle of night and lowest in the afternoon. Patients suffering from neuropathy, a disease resulting from nerve damage leading to an increase in pain sensitivity, experience an approximately 12-hour shift in their rhythmicity such that the highest sensitivity occurs in the afternoon. Neuropathy is a difficult condition to treat since it is often unfeasible to locate the damaged nerve and it is also unclear how this damage causes a shift in rhythmicity and an increase in pain. Understanding the mechanism underlying the shift in rhythmicity may lead to improvements in the knowledge of the transmission of pain from the damaged nerve to the pain-processing center in the spinal cord, and thus better treatment protocols. We have built a population-based model to describe this transmission with a particular focus on daily rhythms. We show that our model reproduces experimentally-observed rhythmicity of both normal pain responses, as well as neuropathic pain. Our model predicts that a potential mechanism underlying the shift in rhythmicity for neuropathic pain is a change in the interaction of the nerve fibers from inhibition to excitation.
Collapse
|
30
|
Morioka N, Kodama K, Tomori M, Yoshikawa K, Saeki M, Nakamura Y, Zhang FF, Hisaoka-Nakashima K, Nakata Y. Stimulation of nuclear receptor REV-ERBs suppresses production of pronociceptive molecules in cultured spinal astrocytes and ameliorates mechanical hypersensitivity of inflammatory and neuropathic pain of mice. Brain Behav Immun 2019; 78:116-130. [PMID: 30682503 DOI: 10.1016/j.bbi.2019.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/15/2019] [Accepted: 01/19/2019] [Indexed: 11/15/2022] Open
Abstract
The orphan nuclear receptors REV-ERBα and REV-ERBβ (REV-ERBs) are crucial in the regulation of inflammatory-related gene transcription in astroglioma cells, but their role in nociceptive transduction has yet to be elaborated. Spinal dorsal horn astrocytes contribute to the maintenance of chronic pain. Treatment of cultured spinal astrocytes with specific REV-ERBs agonists SR9009 or GSK4112 significantly prevented lipopolysaccharide (LPS)-induced mRNA upregulation of pronociceptive molecules interleukin-1β (IL-1β) mRNA, interleukin-6 (IL-6) mRNA and matrix metalloprotease-9 (MMP-9) mRNA, but not CCL2 mRNA expression. Treatment with SR9009 also blocked tumor necrosis factor-induced IL-1β mRNA, IL-6 mRNA and MMP-9 mRNA. In addition, treatment with SR9009 significantly blocked LPS-induced upregulation of IL-1β protein, IL-6 protein and MMP-9 activity. The inhibitory effects of SR9009 on LPS-induced expression of pronociceptive molecules were blocked by knockdown of REV-ERBs expression with short interference RNA, confirming that SR9009 exerts its effect through REV-ERBs. Intrathecal LPS treatment in male mice induces hind paw mechanical hypersensitivity, and upregulation of IL-1β mRNA, IL-6 mRNA and glial fibrillary acidic protein (GFAP) expression in spinal dorsal horn. Intrathecal pretreatment of SR9009 prevented the onset of LPS-induced mechanical hypersensitivity, cytokine expression and GFAP expression. Intrathecal injection of SR9009 also ameliorated mechanical hypersensitivity during the maintenance phase of complete Freund's adjuvant-induced inflammatory pain and partial sciatic nerve ligation-, paclitaxel-, and streptozotocin-induced neuropathy in mice. The current findings suggest that spinal astrocytic REV-ERBs could be critical in the regulation of nociceptive transduction through downregulation of pronociceptive molecule expression. Thus, spinal REV-ERBs could be an effective therapeutic target in the treatment of chronic pain.
Collapse
Affiliation(s)
- Norimitsu Morioka
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| | - Keitaro Kodama
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Mizuki Tomori
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kanade Yoshikawa
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Munenori Saeki
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; Cellular Pathobiology Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse IRP, Triad Suite 3305, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Fang Fang Zhang
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; Institute of Pharmacology, Taishan Medical University, 619 Changcheng Road, Taian, Shandong 271016, China
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Yoshihiro Nakata
- Department of Pharmacology, Hiroshima University Graduate School of Biomedical & Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| |
Collapse
|
31
|
Koike T, Tanaka S, Hirahara Y, Oe S, Kurokawa K, Maeda M, Suga M, Kataoka Y, Yamada H. Morphological characteristics of p75 neurotrophin receptor‐positive cells define a new type of glial cell in the rat dorsal root ganglia. J Comp Neurol 2019; 527:2047-2060. [DOI: 10.1002/cne.24667] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 02/07/2019] [Accepted: 02/12/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Taro Koike
- Department of Anatomy and Cell ScienceKansai Medical University Hirakata Osaka Japan
| | - Susumu Tanaka
- Department of Anatomy and Cell ScienceKansai Medical University Hirakata Osaka Japan
| | - Yukie Hirahara
- Department of Anatomy and Cell ScienceKansai Medical University Hirakata Osaka Japan
| | - Souichi Oe
- Department of Anatomy and Cell ScienceKansai Medical University Hirakata Osaka Japan
| | - Kiyoshi Kurokawa
- Department of Human Health ScienceOsaka International University Moriguchi Osaka Japan
| | - Mitsuyo Maeda
- Multi‐Modal Microstructure Analysis UnitRIKEN‐JEOL Collaboration Center Kobe Hyogo Japan
| | - Mitsuo Suga
- Multi‐Modal Microstructure Analysis UnitRIKEN‐JEOL Collaboration Center Kobe Hyogo Japan
| | - Yosky Kataoka
- Multi‐Modal Microstructure Analysis UnitRIKEN‐JEOL Collaboration Center Kobe Hyogo Japan
- Laboratory for Cellular Function ImagingRIKEN Center for Biosystems Dynamics Research Kobe Hyogo Japan
| | - Hisao Yamada
- Department of Anatomy and Cell ScienceKansai Medical University Hirakata Osaka Japan
| |
Collapse
|
32
|
Das V, Kc R, Li X, O-Sullivan I, van Wijnen AJ, Kroin JS, Pytowski B, Applegate DT, Votta-Velis G, Ripper RL, Park TJ, Im HJ. Blockade of Vascular Endothelial Growth Factor Receptor-1 (Flt-1), Reveals a Novel Analgesic For Osteoarthritis-Induced Joint Pain. GENE REPORTS 2018; 11:94-100. [PMID: 30873504 DOI: 10.1016/j.genrep.2018.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is a painful and debilitating disease. A striking feature of OA is the dramatic increase in vascular endothelial growth factor (VEGF) levels and in new blood vessel formation in the joints, both of which correlate with the severity of OA pain. Our aim was to determine whether anti-VEGF monoclonal antibodies (mAbs) - MF-1 (mAb to VEGFR1) and DC101 (mAb to VEGFR2) - can reduce OA pain and can do so by targeting VEGF signaling pathways such as Flt-1 (VEGFR1) and Flk-1 (VEGFR2). After IACUC approval, OA was induced by partial medial meniscectomy (PMM) in C57/BL6 mice (20 g). ln the first experiment, for validation of VEGFR1 in DRG, the mouse dorsal root ganglion (DRG) was stimulated with NGF for 48 hours to find the relative gene induction for VEGFR1 vs. 18S by RT-PCR. In the second experiment, Biotin-conjugated VEGFA (1 µg/knee joint) was administered in the left knee joint of mice with advanced OA in order to characterization of VEGFR1 and VEGFR2. pVEGFR1/VEGFR2 was detected by immunostaining in DRGs. Finally, MF-1 and DC101 were administered in OA mice by both intrathecal (IT) and intraarticular (IA) injections, and the change in paw withdrawal threshold (PWT) was measured. Retrograde transport of VEGF was confirmed for detection of pVEGFR1/VEGFR2 in the DRG. PMM surgery led to development of OA and mechanical allodynia, with reduced paw withdrawal thresholds (PWT) (P<0.0001). IT injection of MF-1 led to a reduction of allodynia in advanced OA, but injection of DC101 did not. IA injection of MF-1 or DC101 at one week after PMM injury did not reduce allodynia, but when injected in advanced OA mice joints at 12 weeks, both Mabs increased PWT an indicator of analgesia. Our data show that MF-1 (VEGR1 inhibition) decreases pain in advanced OA after IT or IA injection. Activation of MF-1 or DC101 may ameliorate OA-related joint pain.
Collapse
Affiliation(s)
- Vaskar Das
- Department of Anesthesiology, Rush University Medical Center, Chicago, Illinois, USA.,Department of Biochemistry, Rush University Medical Center, Chicago, Illinois, USA
| | - Ranjan Kc
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois, USA
| | - Xin Li
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois, USA
| | - InSug O-Sullivan
- Department of Internal Medicine, the University of Illinois at Chicago (UIC), Illinois, USA
| | | | - Jeffrey S Kroin
- Department of Anesthesiology, Rush University Medical Center, Chicago, Illinois, USA
| | - Bronislaw Pytowski
- Eli Lilly and Company, Alexandria Center for Life Sciences, New York, USA
| | - Daniel T Applegate
- Department of Biological Science, the University of Illinois at Chicago (UIC), Illinois, USA
| | - Gina Votta-Velis
- Department of Anesthesiology, the University of Illinois at Chicago (UIC), Illinois, USA
| | - Richard L Ripper
- Department of Anesthesiology, the University of Illinois at Chicago (UIC), Illinois, USA
| | - Thomas J Park
- Department of Biological Science, the University of Illinois at Chicago (UIC), Illinois, USA
| | - Hee-Jeong Im
- Department of Bioengineering, the University of Illinois at Chicago (UIC), Illinois, USA.,Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, Illinois, USA
| |
Collapse
|