1
|
Gupta A, Mishra SK, Lascelles BDX. Emerging evidence of artemin/GFRα3 signaling in musculoskeletal pain. Osteoarthritis Cartilage 2025; 33:196-206. [PMID: 39374825 PMCID: PMC11757073 DOI: 10.1016/j.joca.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
Chronic musculoskeletal pain is highly prevalent and poses a significant personal, societal, and economic burden. Management of chronic musculoskeletal pain remains a challenge. Long-term use of common analgesic medications such as nonsteroidal anti-inflammatory drugs and opioids is associated with adverse events, and in the case of opioids, drug addiction. Additionally, many individuals do not experience sufficient pain relief with these therapeutic approaches. Thus, there is an urgent need to develop clinically efficacious and safe therapeutics for musculoskeletal pain. Recent advances in our understanding of musculoskeletal pain neurobiology have helped identify the role of neurotrophic factors, specifically, the glial cell line-derived neurotrophic factor (GDNF) family of ligands (GFL) and their associated signaling pathways. This review outlines our current understanding of the GFL signaling systems, discusses their role in inflammatory and chronic musculoskeletal pain and sensitivity, and comments on the analgesic therapeutic potential of targeting the GFL signaling system.
Collapse
Affiliation(s)
- Ankita Gupta
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Santosh K Mishra
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| | - B Duncan X Lascelles
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, USA; Thurston Arthritis Center, UNC School of Medicine, Chapel Hill, NC, USA; Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, NC, USA.
| |
Collapse
|
2
|
Wei X, Li M, You J, Luo J, Zhai J, Zhang J, Feng J, Wang H, Zhou Y. A Procedural Overview of the Involvement of Small Molecules in the Nervous System in the Regulation of Bone Healing. Int J Nanomedicine 2025; 20:1263-1284. [PMID: 39906525 PMCID: PMC11792627 DOI: 10.2147/ijn.s505677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Clinically, a multitude of factors can contribute to the development of bone defects. In the process of bone healing, the nervous system plays a vital role in bone regeneration. Small molecules from the nervous system, such as neurotrophic factors and neuropeptides, have been found to stimulate osteoblast proliferation and differentiation by activating signaling pathways associated with bone calcification and angiogenesis. These small molecules play a crucial regulatory role at various stages of bone healing. The systematic release mechanism of small molecules within the nervous system through diverse bone tissue engineering materials holds significant clinical implications for the controlled regulation of the bone healing process. This review provides an overview of the involvement of various nervous system small molecules at different stages of bone healing and discusses their regulatory mechanisms, aiming to establish a theoretical foundation for programmed regulation in bone regeneration and design of replacement materials in bone tissue engineering.
Collapse
Affiliation(s)
- Xuyan Wei
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Mucong Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiaqian You
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiaxin Luo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jingjie Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiameng Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jian Feng
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Hanchi Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
3
|
Wu TS, Hsiao TH, Chen CH, Li HN, Hung MN, Jhan PP, Tsai JR, Teng CLJ. ARTN and CCL23 predicted chemosensitivity in acute myeloid leukemia: an Olink ® proteomics approach. Clin Proteomics 2025; 22:3. [PMID: 39833682 PMCID: PMC11749431 DOI: 10.1186/s12014-025-09527-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The standard "7 + 3" induction results in 30% of de novo acute myeloid leukemia (AML) patients not achieving complete remission (CR). We aimed to utilize the Olink® platform to compare the bone marrow plasma proteomic profiles of newly diagnosed de novo AML patients who did and did not achieve CR following "7 + 3" induction treatment. METHODS This prospective study included 43 untreated AML patients, stratified into CR (n = 29) and non-CR (n = 14) groups based on their response to "7 + 3" induction therapy. We employed the Olink® Explore-384 Inflammation platform for proteomic analysis to investigate differences in bone marrow plasma protein levels between the CR and non-CR groups. RESULTS Proteomic analysis demonstrated that the CR group exhibited significantly higher bone marrow plasma levels of ARTN and CCL23 than did the non-CR group. Immunohistochemical staining confirmed a higher proportion of tissue samples with intense staining for ARTN (25.40% vs. 7.05%, p = 0.013) and CCL23 (24.14% vs. 14.29%, p = 0.039) in the CR group. These findings were corroborated by bulk-RNA-seq, which indicated significantly elevated mRNA expression levels of ARTN (1.93 vs. -0.09; p = 0.003) and CCL23 (1.50 vs. 0.12; p = 0.021) in the CR group. The Human Protein Atlas provided external support for our findings. CONCLUSIONS The results suggest that ARTN and CCL23 may serve as biomarkers for predicting responsiveness to the "7 + 3" induction in untreated AML. Using an enzyme-linked immunosorbent assay to identify the roles of ARTN and CCL23 in predicting AML chemosensitivity may enhance clinical applicability in the future.
Collapse
Affiliation(s)
- Ting-Shuan Wu
- Department of Biomedical Sciences, Chung Shan Medical University, No. 110, Sect. 1, Jiangou North Rd, Taichung, 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Sect. 1, Jiangou North Rd, Taichung, 40201, Taiwan
| | - Tzu-Hung Hsiao
- Department of Medical Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan
- Research Center for Biomedical Science and Engineering, National Tsing Hua University, Sect 2, Kuang-Fu Rd, Hsinchu, 101, 300044, Taiwan
- Department of Public Health, Fu Jen Catholic University, No. 510, Zhongzheng Rd, New Taipei City, 242062, Taiwan
- Institute of Genomics and Bioinformatics, National Chung Hsing University, No. 145 Xingda Rd., South Dist, Taichung, 402202, Taiwan
| | - Chung-Hsing Chen
- Department of Mathematics, University of Taipei, No. 1 Ai-Guo West Rd, Taipei, 100234, Taiwan
| | - Hsin-Ni Li
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, No. 145 Xingda Rd., South Dist, Taichung, 402202, Taiwan
| | - Miao-Neng Hung
- Department of Biomedical Sciences, Chung Shan Medical University, No. 110, Sect. 1, Jiangou North Rd, Taichung, 40201, Taiwan
| | - Pei-Pei Jhan
- Department of Medical Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan
| | - Jia-Rung Tsai
- Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan
| | - Chieh-Lin Jerry Teng
- Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, No. 145 Xingda Rd., South Dist, Taichung, 402202, Taiwan.
- Department of Life Science, Tunghai University, No. 1727, Sect. 4, Taiwan boulevard, Taichung, 407224, Taiwan.
- School of Medicine, Chung Shan Medical University, No. 110, Sect. 1, Jiangou North Rd, Taichung, 40201, Taiwan.
- Ph.D. Program in Translational Medicine, National Chung Hsing University, No. 145 Xingda Rd., South Dist, Taichung, 402202, Taiwan.
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, No. 145 Xingda Rd., South Dist, Taichung, 402202, Taiwan.
| |
Collapse
|
4
|
Pierre-Jerome C. The peripheral nervous system: peripheral neuropathies in the diabetic foot. MYOPATHIES AND TENDINOPATHIES OF THE DIABETIC FOOT 2025:451-482. [DOI: 10.1016/b978-0-443-13328-2.00022-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Morgan M, Nazemian V, Thai J, Lin I, Northfield S, Ivanusic JJ. BDNF sensitizes bone and joint afferent neurons at different stages of MIA-induced osteoarthritis. Bone 2024; 189:117260. [PMID: 39299629 DOI: 10.1016/j.bone.2024.117260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
There is emerging evidence that Brain Derived Neurotrophic Factor (BDNF), and one of its receptors TrkB, play important roles in the pathogenesis of osteoarthritis (OA) pain. Whilst these studies clearly highlight the potential for targeting BDNF/TrkB signaling to treat OA pain, the mechanism for how BDNF/TrkB signaling contributes to OA pain remains unclear. In this study, we used an animal model of mono-iodoacetate (MIA)-induced OA, in combination with electrophysiology, behavioral testing, Western blot analysis, and retrograde tracing and immunohistochemistry, to identify roles for BDNF/TrkB signaling in the pathogenesis of OA pain. We found that: 1) TrkB is expressed in myelinated medium diameter neurons that innervate the knee joint and bone in naïve animals; 2) peripheral application of BDNF increases the sensitivity of Aδ, but not C knee joint and bone afferent neurons, in response to mechanical stimulation, in naïve animals; 3) BDNF expression increases in synovial tissue in early MIA-induced OA, when pathology is confined to the joint, and in the subchondral bone in late MIA-induced OA, when there is additional damage to the surrounding bone; and 4) TrkB inhibition reverses MIA-induced changes in the sensitivity of Aδ but not C knee joint afferent neurons early in MIA-induced OA, and Aδ but not C bone afferent neurons late in MIA-induced OA. Our findings suggest that BDNF/TrkB signaling may have a role to play in the pathogenesis of OA pain, through effects on knee joint afferent neurons early in disease when there is inflammation confined to the joint, and bone afferent neurons late in disease when there is involvement of damage to subchondral bone. Targeted manipulation of BDNF/TrkB signaling may provide therapeutic benefit for the management of OA pain.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Vida Nazemian
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Irene Lin
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria, Australia
| | - Susan Northfield
- Department of Biochemistry and Pharmacology, University of Melbourne, Victoria, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
6
|
Wang Y, Zhou W, Zhang F, Wei J, Wang S, Min K, Chen Y, Yang H, Lv X. Exploring the bidirectional causal associations between pain and circulating inflammatory proteins: A Mendelian randomization study. Clin Exp Pharmacol Physiol 2024; 51:e13905. [PMID: 38965671 DOI: 10.1111/1440-1681.13905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024]
Abstract
Multisite chronic pain (MCP) and site-specific chronic pain (SSCP) may be influenced by circulating inflammatory proteins, but the causal relationship remains unknown. To overcome this limitation, two-sample bidirectional Mendelian randomization (MR) analysis was used to analyse data for 91 circulating inflammatory proteins, MCP and SSCP encompassing headache, back pain, shoulder pain, hip pain, knee pain, stomach abdominal pain and facial pain. The primary MR method used was inverse variance weighting, sensitivity analyses included weighted median, MR pleiotropy residual sum and outlier and the Egger intercept method. Heterogeneity was also detected using Cochrane's Q test and leave-one-out analyses. Finally, a causal relationship between 29 circulating inflammatory proteins and chronic pain was identified. Among these proteins, 14 exhibited a protective effect, including MCP (T-cell surface glycoprotein cluster of differentiation 5), headache (4E-binding protein 1 [4EBP1], cluster of differentiation 40, cluster of differentiation 6 and C-X-C motif chemokine [CXCL] 11), back pain (leukaemia inhibitory factor), shoulder pain (fibroblast growth factor [FGF]-5 and interleukin [IL]-18R1), stomach abdominal pain (tumour necrosis factor [TNF]-α), hip pain (CXCL1, IL-20 and signalling lymphocytic activation molecule 1) and knee pain (IL-7 and TNF-β). Additionally, 15 proteins were identified as risk factors for MCP and SSCP: MCP (colony-stimulating factor 1, human glial cell line-derived neurotrophic factor and IL-17C), headache (fms-related tyrosine kinase 3 ligand, IL-20 receptor subunit α [IL-20RA], neurotrophin-3 and tumour necrosis factor receptor superfamily member 9), facial pain (CXCL1), back pain (TNF), shoulder pain (IL-17C and matrix metalloproteinase-10), stomach abdominal pain (IL-20RA), hip pain (C-C motif chemokine 11/eotaxin-1 and tumour necrosis factor ligand superfamily member 12) and knee pain (4EBP1). Importantly, in the opposite direction, MCP and SSCP did not exhibit a significant causal impact on circulating inflammatory proteins. Our study identified potential causal influences of various circulating inflammatory proteins on MCP and SSCP and provided promising treatments for the clinical management of MCP and SSCP.
Collapse
Affiliation(s)
- Yu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenyu Zhou
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Faqiang Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Juan Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Sheng Wang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keting Min
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanli Chen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Yang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
7
|
Dong Q, Wu J, Zhang H, Luo L, Wu W. The causal role of circulating inflammatory markers in osteoporosis: a bidirectional Mendelian randomized study. Front Immunol 2024; 15:1412298. [PMID: 39091505 PMCID: PMC11291241 DOI: 10.3389/fimmu.2024.1412298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Background Osteoporosis (OP) associated with aging exerts substantial clinical and fiscal strains on societal structures. An increasing number of research studies have suggested a bidirectional relationship between circulating inflammatory markers (CIMs) and OP. However, observational studies are susceptible to perturbations in confounding variables. In contrast, Mendelian randomization (MR) offers a robust methodological framework to circumvent such confounders, facilitating a more accurate assessment of causality. Our study aimed to evaluate the causal relationships between CIMs and OP, identifying new approaches and strategies for the prevention, diagnosis and treatment of OP. Methods We analyzed publicly available GWAS summary statistics to investigate the causal relationships between CIMs and OP. Causal estimates were calculated via a systematic analytical framework, including bidirectional MR analysis and Bayesian colocalization analysis. Results Genetically determined levels of CXCL11 (OR = 0.91, 95% CI = 0.85-0.98, P = 0.008, PFDR = 0.119), IL-18 (OR = 0.88, 95% CI = 0.83-0.94, P = 8.66×10-5, PFDR = 0.008), and LIF (OR = 0.86, 95% CI = 0.76-0.96, P = 0.008, PFDR = 0.119) were linked to a reduced risk of OP. Conversely, higher levels of ARTN (OR = 1.11, 95% CI = 1.02-1.20, P = 0.012, PFDR = 0.119) and IFNG (OR = 1.16, 95% CI = 1.03-1.30, P = 0.013, PFDR = 0.119) were associated with an increased risk of OP. Bayesian colocalization analysis revealed no evidence of shared causal variants. Conclusion Despite finding no overall association between CIMs and OP, five CIMs demonstrated a potentially significant association with OP. These findings could pave the way for future mechanistic studies aimed at discovering new treatments for this disease. Additionally, we are the first to suggest a unidirectional causal relationship between ARTN and OP. This novel insight introduces new avenues for research into diagnostic and therapeutic strategies for OP.
Collapse
Affiliation(s)
- Qiu Dong
- Department of Bone and Joint Surgery, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jiayang Wu
- Medical Imaging Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Huaguo Zhang
- Department of Ultrasonography, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Liangping Luo
- Medical Imaging Centre, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Medical Imaging Center, The Fifth Affiliated Hospital of Jinan University, Heyuan, Guangdong, China
| | - Wenrui Wu
- Department of Bone and Joint Surgery, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Department of Orthopedics, Chaoshan Hospital, The First Affiliated Hospital of Jinan University, Chaozhou, Guangdong, China
| |
Collapse
|
8
|
Saito S, Shahbaz S, Osman M, Redmond D, Bozorgmehr N, Rosychuk RJ, Lam G, Sligl W, Cohen Tervaert JW, Elahi S. Diverse immunological dysregulation, chronic inflammation, and impaired erythropoiesis in long COVID patients with chronic fatigue syndrome. J Autoimmun 2024; 147:103267. [PMID: 38797051 DOI: 10.1016/j.jaut.2024.103267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
A substantial number of patients recovering from acute SARS-CoV-2 infection present serious lingering symptoms, often referred to as long COVID (LC). However, a subset of these patients exhibits the most debilitating symptoms characterized by ongoing myalgic encephalomyelitis or chronic fatigue syndrome (ME/CFS). We specifically identified and studied ME/CFS patients from two independent LC cohorts, at least 12 months post the onset of acute disease, and compared them to the recovered group (R). ME/CFS patients had relatively increased neutrophils and monocytes but reduced lymphocytes. Selective T cell exhaustion with reduced naïve but increased terminal effector T cells was observed in these patients. LC was associated with elevated levels of plasma pro-inflammatory cytokines, chemokines, Galectin-9 (Gal-9), and artemin (ARTN). A defined threshold of Gal-9 and ARTN concentrations had a strong association with LC. The expansion of immunosuppressive CD71+ erythroid cells (CECs) was noted. These cells may modulate the immune response and contribute to increased ARTN concentration, which correlated with pain and cognitive impairment. Serology revealed an elevation in a variety of autoantibodies in LC. Intriguingly, we found that the frequency of 2B4+CD160+ and TIM3+CD160+ CD8+ T cells completely separated LC patients from the R group. Our further analyses using a multiple regression model revealed that the elevated frequency/levels of CD4 terminal effector, ARTN, CEC, Gal-9, CD8 terminal effector, and MCP1 but lower frequency/levels of TGF-β and MAIT cells can distinguish LC from the R group. Our findings provide a new paradigm in the pathogenesis of ME/CFS to identify strategies for its prevention and treatment.
Collapse
Affiliation(s)
- Suguru Saito
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Shima Shahbaz
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Mohammed Osman
- Department of Medicine, Division of Rheumatology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Desiree Redmond
- Department of Medicine, Division of Rheumatology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Najmeh Bozorgmehr
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Rhonda J Rosychuk
- Department of Pediatrics, Division of Infectious Disease, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Grace Lam
- Department of Medicine, Division of Pulmonary Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Wendy Sligl
- Department of Critical Care Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada; Department of Medicine, Division of Infectious Diseases, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Jan Willem Cohen Tervaert
- Department of Medicine, Division of Rheumatology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, Division of Foundational Sciences, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada; Department of Oncology, University of Alberta, Edmonton, T6G 2E1, AB, Canada; Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada; Li Ka Shing Institute of Virology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, T6G 2E1, AB, Canada.
| |
Collapse
|
9
|
Bencze N, Scheich B, Szőke É, Wilhelm I, Körmöndi S, Botz B, Helyes Z. Osteosarcoma-Induced Pain Is Mediated by Glial Cell Activation in the Spinal Dorsal Horn, but Not Capsaicin-Sensitive Nociceptive Neurons: A Complex Functional and Morphological Characterization in Mice. Cancers (Basel) 2024; 16:1788. [PMID: 38791867 PMCID: PMC11120600 DOI: 10.3390/cancers16101788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Bone cancer and its related chronic pain are huge clinical problems since the available drugs are often ineffective or cannot be used long term due to a broad range of side effects. The mechanisms, mediators and targets need to be identified to determine potential novel therapies. Here, we characterize a mouse bone cancer model induced by intratibial injection of K7M2 osteosarcoma cells using an integrative approach and investigate the role of capsaicin-sensitive peptidergic sensory nerves. The mechanical pain threshold was assessed by dynamic plantar aesthesiometry, limb loading by dynamic weight bearing, spontaneous pain-related behaviors via observation, knee diameter with a digital caliper, and structural changes by micro-CT and glia cell activation by immunohistochemistry in BALB/c mice of both sexes. Capsaicin-sensitive peptidergic sensory neurons were defunctionalized by systemic pretreatment with a high dose of the transient receptor potential vanilloid 1 (TRPV1) agonist resiniferatoxin (RTX). During the 14- and 28-day experiments, weight bearing on the affected limb and the paw mechanonociceptive thresholds significantly decreased, demonstrating secondary mechanical hyperalgesia. Signs of spontaneous pain and osteoplastic bone remodeling were detected both in male and female mice without any sex differences. Microglia activation was shown by the increased ionized calcium-binding adapter molecule 1 (Iba1) immunopositivity on day 14 and astrocyte activation by the enhanced glial fibrillary acidic protein (GFAP)-positive cell density on day 28 in the ipsilateral spinal dorsal horn. Interestingly, defunctionalization of the capsaicin-sensitive afferents representing approximately 2/3 of the nociceptive fibers did not alter any functional parameters. Here, we provide the first complex functional and morphological characterization of the K7M2 mouse osteosarcoma model. Bone-cancer-related chronic pain and hyperalgesia are likely to be mediated by central sensitization involving neuroinflammation via glial cell activation in the spinal dorsal horn, but not the capsaicin-sensitive sensory neuronal system.
Collapse
Affiliation(s)
- Noémi Bencze
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
| | - Bálint Scheich
- Department of Pathology and Experimental Cancer Research, Faculty of Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary;
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN-PTE), 7624 Pécs, Hungary
| | - Imola Wilhelm
- Institute of Biophysics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary;
| | - Sándor Körmöndi
- Department of Traumatology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Bálint Botz
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- Department of Medical Imaging, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, 7624 Pécs, Hungary; (N.B.); (É.S.); (B.B.)
- National Laboratory for Drug Research and Development, Magyar Tudósok Krt. 2, 1117 Budapest, Hungary
- Hungarian Research Network, Chronic Pain Research Group (HUN-REN-PTE), 7624 Pécs, Hungary
- PharmInVivo Ltd., Szondy György Str. 10, 7629 Pécs, Hungary
| |
Collapse
|
10
|
Wang Y, Jiang T. Recent research advances in pain mechanisms in McCune-Albright syndrome thinking about the pain mechanism of FD/MAS. J Orthop Surg Res 2024; 19:196. [PMID: 38515135 PMCID: PMC10956191 DOI: 10.1186/s13018-024-04687-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/16/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND The lack of effective understanding of the pain mechanism of McCune-Albright syndrome (MAS) has made the treatment of pain in this disease a difficult clinical challenge, and new therapeutic targets are urgently needed to address this dilemma. OBJECTIVE This paper summarizes the novel mechanisms, targets, and treatments that may produce pain in MAS and fibrous dysplasia (polyfibrous dysplasia, or FD). METHODS We conducted a systematic search in the PubMed database, Web of Science, China Knowledge Network (CNKI) with the following keywords: "McCune-Albright syndrome (MAS); polyfibrous dysplasia (FD); bone pain; bone remodeling; G protein coupled receptors; GDNF family receptors; purinergic receptors and glycogen synthase kinase", as well as other keywords were systematically searched. Papers published between January 2018 and May 2023 were selected for finding. Initial screening was performed by reading the titles and abstracts, and available literature was screened against the inclusion and exclusion criteria. RESULTS In this review, we systematically analyzed the cutting-edge advances in this disease, synthesized the findings, and discussed the differences. With regard to the complete mechanistic understanding of the pain condition in FD/MAS, in particular, we collated new findings on new pathways, neurotrophic factor receptors, purinergic receptors, interferon-stimulating factors, potassium channels, protein kinases, and corresponding hormonal modulation and their respective strengths and weaknesses. CONCLUSION This paper focuses on basic research to explore FD/MAS pain mechanisms. New nonneuronal and molecular mechanisms, mechanically loaded responsive neurons, and new targets for potential clinical interventions are future research directions, and a large number of animal experiments, tissue engineering techniques, and clinical trials are still needed to verify the effectiveness of the targets in the future.
Collapse
Affiliation(s)
- Yong Wang
- Orthopedics Department, Changzhou Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Changzhou, 213000, Jangsu Province, China
| | - Tao Jiang
- Orthopedics Department, Changzhou Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Changzhou, 213000, Jangsu Province, China.
| |
Collapse
|
11
|
Torres HM, Arnold KM, Oviedo M, Westendorf JJ, Weaver SR. Inflammatory Processes Affecting Bone Health and Repair. Curr Osteoporos Rep 2023; 21:842-853. [PMID: 37759135 PMCID: PMC10842967 DOI: 10.1007/s11914-023-00824-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing. RECENT FINDINGS Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells. Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.
Collapse
Affiliation(s)
- Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Biomedical Engineering and Physiology Track/Regenerative Sciences Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuela Oviedo
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
12
|
Iwasaki T, Akeda K, Kawaguchi K, Yamada J, Hasegawa T, Takegami N, Fujiwara T, Sudo A. Expression of Glial-Cell-Line-Derived Neurotrophic Factor Family Ligands in Human Intervertebral Discs. Int J Mol Sci 2023; 24:15874. [PMID: 37958856 PMCID: PMC10649213 DOI: 10.3390/ijms242115874] [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: 09/05/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Glial-cell-line-derived neurotrophic factor (GDNF) family ligands (GFLs) contribute to the sensitization of primary afferents and are involved in the pathogenesis of inflammatory pain. The purpose of this preliminary study was to examine the expression of other GFLs (neurturin (NRTN), artemin (ARTN), persephin (PSPN)) and receptors in human IVD cells and tissues exhibiting early and advanced stages of degeneration. Human IVD cells were cultured as a monolayer after isolation from the nucleus pulposus (NP) and anulus fibrosus (AF) tissues. The mRNA expression of NRTN, ARTN, PSPN, and their receptors (GFRA2-GFRA4) was quantified using real-time PCR. Protein expression was evaluated using immunohistochemistry and Western blotting. The expression of NRTN, ARTN, PSPN, and their co-receptors (GFRA2-GFRA4) was identified in human IVD cells at both mRNA and protein levels. A trend was noted wherein the mRNA expression of ARTN, PSPN, and GFRA2 was upregulated by IL-1β treatment in a dose-dependent manner. The percentages of immunopositive cells in the advanced degenerate stage of ARTN, PSPN, and GFRA2 were significantly higher than those in the early degenerate stage. Their expression was enhanced in advanced tissue degeneration, which suggests that GFLs (ARTN and PSPN) may be involved in the pathogenesis of discogenic pain.
Collapse
Affiliation(s)
| | - Koji Akeda
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu City 514-8507, Mie, Japan; (T.I.); (K.K.); (J.Y.); (T.H.); (N.T.); (T.F.); (A.S.)
| | | | | | | | | | | | | |
Collapse
|
13
|
Morgan M, Nazemian V, Ooi LS, Burger S, Thai J, Ivanusic J. Artemin sensitizes nociceptors that innervate the osteoarthritic joint to produce pain. Osteoarthritis Cartilage 2023; 31:1342-1352. [PMID: 37353141 DOI: 10.1016/j.joca.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/14/2023] [Accepted: 06/09/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVE There have been significant developments in understanding artemin/GFRα3 signaling in recent years, and there is now accumulating evidence that artemin has important roles to play in pain signaling, including that derived from joint and bone, and that associated with osteorthritis (OA). METHODS A total of 163 Sprague-Dawley rats were used in this study. We used an animal model of mono-iodoacetate (MIA)-induced OA, in combination with electrophysiology, behavioral testing, Western blot analysis, and retrograde tracing and immunohistochemistry, to identify roles for artemin/GFRα3 signaling in the pathogenesis of OA pain. RESULTS We have found that: 1) GFRα3 is expressed in a substantial proportion of knee joint afferent neurons; 2) exogenous artemin sensitizes knee joint afferent neurons in naïve rats; 3) artemin is expressed in articular tissues of the joint, but not surrounding bone, early in MIA-induced OA; 4) artemin expression increases in bone later in MIA-induced OA when pathology involves subchondral bone; and 5) sequestration of artemin reverses MIA-induced sensitization of both knee joint and bone afferent neurons late in disease when there is inflammation of knee joint tissues and damage to the subchondral bone. CONCLUSIONS Our findings show that artemin/GFRα3 signaling has a role to play in the pathogenesis of OA pain, through effects on both knee joint and bone afferent neurons, and suggest that targeted manipulation of artemin/GFRα3 signaling may provide therapeutic benefit for the management of OA pain. DATA AVAILABILITY Data are available on request of the corresponding author.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Vida Nazemian
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Li Sha Ooi
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Sarah Burger
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| | - Jason Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
14
|
Smith ESJ. A new art to treating osteoarthritis pain? Osteoarthritis Cartilage 2023; 31:1275-1277. [PMID: 37460008 DOI: 10.1016/j.joca.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/10/2023] [Indexed: 07/31/2023]
Affiliation(s)
- Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| |
Collapse
|
15
|
Somersan-Karakaya S, Turner KC, Cortes-Burgos L, Miller J, LaCroix-Fralish M, Logovinsky V, Patel Y, Torres R, Ganguly S, Breazna A, DeVeaux M, Bhore R, Gao M, Delfino FJ, Rafique A, Fairhurst JL, Hunt C, Babb R, Badithe A, Poueymirou WT, Surowitz R, Rottey S, Murphy AJ, Harari O, Macdonald LE, Croll SD. Monoclonal antibodies against GFRα3 are efficacious against evoked hyperalgesic and allodynic responses in mouse join pain models but, one of these, REGN5069, was not effective against pain in a randomized, placebo-controlled clinical trial in patients with osteoarthritis pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 14:100136. [PMID: 38099276 PMCID: PMC10719528 DOI: 10.1016/j.ynpai.2023.100136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 12/17/2023]
Abstract
The artemin-GFRα3 signaling pathway has been implicated in various painful conditions including migraine, cold allodynia, hyperalgesia, inflammatory bone pain, and mouse knees contain GFRα3-immunoreactive nerve endings. We developed high affinity mouse (REGN1967) and human (REGN5069) GFRα3-blocking monoclonal antibodies and, following in vivo evaluations in mouse models of chronic joint pain (osteoarthritic-like and inflammatory), conducted a first-in-human phase 1 pharmacokinetics (PK) and safety trial of REGN5069 (NCT03645746) in healthy volunteers, and a phase 2 randomized placebo-controlled efficacy and safety trial of REGN5069 (NCT03956550) in patients with knee osteoarthritis (OA) pain. In three commonly used mouse models of chronic joint pain (destabilization of the medial meniscus, intra-articular monoiodoacetate, or Complete Freund's Adjuvant), REGN1967 and REGN5069 attenuated evoked behaviors including tactile allodynia and thermal hyperalgesia without discernably impacting joint pathology or inflammation, prompting us to further evaluate REGN5069 in humans. In the phase 1 study in healthy subjects, the safety profiles of single doses of REGN5069 up to 3000 mg (intravenous) or 600 mg (subcutaneous) were comparable to placebo; PK were consistent with a monoclonal antibody exhibiting target-mediated disposition. In the phase 2 study in patients with OA knee pain, two doses of REGN5069 (100 mg or 1000 mg intravenous every 4 weeks) for 8 weeks failed to achieve the 12-week primary and secondary efficacy endpoints relative to placebo. In addition to possible differences in GFRα3 biology between mice and humans, we highlight here differences in experimental parameters that could have contributed to a different profile of efficacy in mouse models versus human OA pain. Additional research is required to more fully evaluate any potential role of GFRα3 in human pain.
Collapse
Affiliation(s)
| | | | | | - Jutta Miller
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | | | - Yamini Patel
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Richard Torres
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Samit Ganguly
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Aurora Breazna
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Rafia Bhore
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Min Gao
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Ashique Rafique
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Charleen Hunt
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Robert Babb
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | - Ashok Badithe
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | | | | | | | - Olivier Harari
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| | | | - Susan D. Croll
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, United States
| |
Collapse
|
16
|
Pharmacological Interventions Targeting Pain in Fibrous Dysplasia/McCune-Albright Syndrome. Int J Mol Sci 2023; 24:ijms24032550. [PMID: 36768871 PMCID: PMC9916440 DOI: 10.3390/ijms24032550] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Fibrous dysplasia (FD) is a rare, non-inherited bone disease occurring following a somatic gain-of-function R201 missense mutation of the guanine-nucleotide binding protein alpha subunit stimulating activity polypeptide 1 (GNAS) gene. The spectrum of the disease ranges from a single FD lesion to a combination with extraskeletal features; an amalgamation with café-au-lait skin hyperpigmentation, precocious puberty, and other endocrinopathies defines McCune-Albright Syndrome (MAS). Pain in FD/MAS represents one of the most prominent aspects of the disease and one of the most challenging to treat-an outcome driven by (i) the heterogeneous nature of FD/MAS, (ii) the variable presentation of pain phenotypes (i.e., craniofacial vs. musculoskeletal pain), (iii) a lack of studies probing pain mechanisms, and (iv) a lack of rigorously validated analgesic strategies in FD/MAS. At present, a range of pharmacotherapies are prescribed to patients with FD/MAS to mitigate skeletal disease activity, as well as pain. We analyze evidence guiding the current use of bisphosphonates, denosumab, and other therapies in FD/MAS, and also discuss the potential underlying pharmacological mechanisms by which pain relief may be achieved. Furthermore, we highlight the range of presentation of pain in individual cases of FD/MAS to further describe the difficulties associated with employing effective pain treatment in FD/MAS. Potential next steps toward identifying and validating effective pain treatments in FD/MAS are discussed, such as employing randomized control trials and probing new pain pathways in this rare bone disease.
Collapse
|
17
|
Tao R, Mi B, Hu Y, Lin S, Xiong Y, Lu X, Panayi AC, Li G, Liu G. Hallmarks of peripheral nerve function in bone regeneration. Bone Res 2023; 11:6. [PMID: 36599828 PMCID: PMC9813170 DOI: 10.1038/s41413-022-00240-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/27/2022] [Accepted: 11/03/2022] [Indexed: 01/06/2023] Open
Abstract
Skeletal tissue is highly innervated. Although different types of nerves have been recently identified in the bone, the crosstalk between bone and nerves remains unclear. In this review, we outline the role of the peripheral nervous system (PNS) in bone regeneration following injury. We first introduce the conserved role of nerves in tissue regeneration in species ranging from amphibians to mammals. We then present the distribution of the PNS in the skeletal system under physiological conditions, fractures, or regeneration. Furthermore, we summarize the ways in which the PNS communicates with bone-lineage cells, the vasculature, and immune cells in the bone microenvironment. Based on this comprehensive and timely review, we conclude that the PNS regulates bone regeneration through neuropeptides or neurotransmitters and cells in the peripheral nerves. An in-depth understanding of the roles of peripheral nerves in bone regeneration will inform the development of new strategies based on bone-nerve crosstalk in promoting bone repair and regeneration.
Collapse
Affiliation(s)
- Ranyang Tao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Yiqiang Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China
| | - Xuan Lu
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, 02215, MA, USA
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, 999077, P. R. China.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, P.R. China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, P. R. China.
| |
Collapse
|
18
|
Morgan M, Thai J, Nencini S, Xu J, Ivanusic JJ. Stomatin-like protein 3 modulates the responses of Aδ, but not C fiber bone afferent neurons to noxious mechanical stimulation in an animal model of acute experimental bone pain. Mol Pain 2023; 19:17448069231222407. [PMID: 38073226 PMCID: PMC10734363 DOI: 10.1177/17448069231222407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023] Open
Abstract
STOML3 is a membrane bound scaffolding protein that has been shown to facilitate the opening of mechanically sensitive ion channels and contribute to noxious mechanical sensation, allodynia and hyperalgesia. In this study, we aimed to determine the role of STOML3 in noxious mechanical sensitivity of bone afferent neurons and carrageenan-induced acute inflammation in the bone. An in vivo, electrophysiological bone-nerve preparation was used to make recordings of the activity and sensitivity of bone afferent neurons that innervate the tibial marrow cavity in anaesthetised rats, in response to noxious mechanical stimuli delivered to the marrow cavity, before and after injection of either the STOML3 oligomerisation inhibitor OB-1 or vehicle, in either naïve animals or animals with carrageenan-induced inflammation of the marrow cavity. A dynamic weight-bearing apparatus was used to measure weight bearing in response to inflammatory pain before and after injection of OB-1 or saline into the tibial marrow cavity in the presence of carrageenan-induced inflammation. Electrophysiological recordings revealed that Aδ, but not C bone afferent neurons have a reduced discharge frequency in response to mechanical stimulation, and that carrageenan-induced sensitisation of Aδ, but not C bone afferent neurons was attenuated by inhibition of STOML3 oligomerisation with OB-1. Animals treated with OB-1 spent a significantly greater amount of time on the limb injected with carrageenan than animals treated with saline. Our findings demonstrate that inhibition of STOML3 oligomerisation reduces inflammatory bone pain by reducing the sensitivity of Aδ bone afferent neurons to mechanical stimulation. Targeting STOML3 may be an effective approach to reduce pain from noxious pressure and/or painful inflammatory pathology in bone.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Sara Nencini
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - James Xu
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Yang H, Gu X, Chen H, Zeng Q, Mao Z, Jin M, Li H, Ge Y, Zha J, Martyniuk CJ. Transcriptome profiling reveals toxicity mechanisms following sertraline exposure in the brain of juvenile zebrafish (Danio rerio). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113936. [PMID: 35930839 DOI: 10.1016/j.ecoenv.2022.113936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/24/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Sertraline (SER) is one of the most commonly detected antidepressants in the aquatic environment that can negatively affect aquatic organisms at low concentrations. Despite some knowledge on its acute toxicity to fish, the effects of chronic SER exposure remain poorly understood along with any underlying mechanisms of SER-induced toxicity. To address this knowledge gap, the effects of chronic exposure to three SER concentrations from low to high were investigated in zebrafish. Juvenile zebrafish were exposed to three concentrations of 1, 10, or 100 μg/L of SER for 28 d, after which indicators of oxidative stress and neurotoxicity in the brain were measured. Superoxide dismutase (SOD) activity was significantly enhanced by SER at 1 up to 100 μg/L, and catalase (CAT) activity was significantly induced by SER at 1 or 10 μg/L. The activity of acetylcholinesterase (AChE) was significantly induced by 10 and 100 μg/L of SER, and the serotonin (5-HT) level was significantly increased by all three concentrations of SER. To ascertain mechanisms of SER-induced toxicity, transcriptomics was conducted in the brain of zebrafish following 100 μg/L SER exposure. The molecular signaling pathways connected with circadian system and the immune system were significantly altered in the zebrafish brain. Based on transcriptomic data, the expression levels of six circadian clock genes were measured, and three genes were significantly altered in relative abundance in fish from all experimental treatments with SER, including cryptochrome circadian regulator 2 (cry2), period circadian clock 2 (per2), and period circadian clock 3 (per3). We hypothesize that the circadian system may be related to SER-induced neurotoxicity and oxidative stress in the central nervous system. This study reveals potential mechanisms and key events (i.e., oxidative stress and neurotoxicity) associated with SER-induced toxicity, and improves understanding of the molecular and biochemical pathways putatively perturbed by SER.
Collapse
Affiliation(s)
- Huiting Yang
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohong Gu
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China.
| | - Huihui Chen
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China.
| | - Qingfei Zeng
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Zhigang Mao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Miao Jin
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongmin Li
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - You Ge
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinmiao Zha
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611 USA
| |
Collapse
|
20
|
Yuan ZL, Liu XD, Zhang ZX, Li S, Tian Y, Xi K, Cai J, Yang XM, Liu M, Xing GG. Activation of GDNF-ERK-Runx1 signaling contributes to P2X3R gene transcription and bone cancer pain. iScience 2022; 25:104936. [PMID: 36072549 PMCID: PMC9441333 DOI: 10.1016/j.isci.2022.104936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/15/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Bone cancer pain is a common symptom in cancer patients with bone metastases and its underlying mechanisms remain unknown. Here, we report that Runx1 directly upregulates the transcriptional activity of P2X3 receptor (P2X3R) gene promoter in PC12 cells. Knocking down Runx1 in dorsal root ganglion (DRG) neurons suppresses the functional upregulation of P2X3R, attenuates neuronal hyperexcitability and pain hypersensitivity in tumor-bearing rats, whereas overexpressing Runx1 promotes P2X3R gene transcription in DRG neurons, induces neuronal hyperexcitability and pain hypersensitivity in naïve rats. Activation of GDNF-GFRα1-Ret-ERK signaling is required for Runx1-mediated P2X3R gene transcription in DRG neurons, and contributes to neuronal hyperexcitability and pain hypersensitivity in tumor-bearing rats. These findings indicate that the Runx1-mediated P2X3R gene transcription resulted from activation of GDNF-GFRα1-Ret-ERK signaling contributes to the sensitization of DRG neurons and pathogenesis of bone cancer pain. Our findings identify a potentially targetable mechanism that may cause bone metastasis-associated pain in cancer patients. Runx1 directly upregulates the transcriptional activity of P2X3R gene promoter Upregulation of Runx1-mediated P2X3R gene transcription underlies bone cancer pain Involvement of GDNF-Ret-ERK signaling in Runx1-mediated P2X3R gene transcription
Collapse
|
21
|
Sun R, Bai L, Yang Y, Ding Y, Zhuang J, Cui J. Nervous System-Driven Osseointegration. Int J Mol Sci 2022; 23:ijms23168893. [PMID: 36012155 PMCID: PMC9408825 DOI: 10.3390/ijms23168893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Implants are essential therapeutic tools for treating bone fractures and joint replacements. Despite the in-depth study of osseointegration for more than fifty years, poor osseointegration caused by aseptic loosening remains one of the leading causes of late implant failures. Osseointegration is a highly sophisticated and spatiotemporal process in vivo involving the immune response, angiogenesis, and osteogenesis. It has been unraveled that the nervous system plays a pivotal role in skeletal health via manipulating neurotrophins, neuropeptides, and nerve cells. Herein, the research related to nervous system-driven osseointegration was systematically analyzed and reviewed, aiming to demonstrate the prominent role of neuromodulation in osseointegration. Additionally, it is indicated that the implant design considering the role of neuromodulation might be a promising way to prevent aseptic loosening.
Collapse
Affiliation(s)
- Ruoyue Sun
- Key Laboratory for Ultrafine Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Correspondence: (J.C.); (L.B.)
| | - Yaru Yang
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing 314001, China
| | - Yanshu Ding
- Key Laboratory for Ultrafine Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jingwen Zhuang
- Key Laboratory for Ultrafine Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jingyuan Cui
- Key Laboratory for Ultrafine Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Correspondence: (J.C.); (L.B.)
| |
Collapse
|
22
|
Jia Z, Kang B, Cai Y, Chen C, Yu Z, Li W, Zhang W. Cell-free fat extract attenuates osteoarthritis via chondrocytes regeneration and macrophages immunomodulation. Stem Cell Res Ther 2022; 13:133. [PMID: 35365233 PMCID: PMC8973552 DOI: 10.1186/s13287-022-02813-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/02/2022] [Indexed: 01/15/2023] Open
Abstract
Background The prevalence of osteoarthritis (OA) is increasing, yet clinically effective and economical treatments are unavailable. We have previously proposed a cell-free fat extract (CEFFE) containing multiple cytokines, which possessed antiapoptotic, anti-oxidative, and proliferation promotion functions, as a “cell-free” strategy. In this study, we aimed to evaluate the therapeutic effect of CEFFE in vivo and in vitro. Methods In vivo study, sodium iodoacetate-induced OA rats were treated with CEFFE by intra-articular injections for 8 weeks. Behavioral experiments were performed every two weeks. Histological analyses, anti-type II collagen, and toluidine staining provided structural evaluation. Macrophage infiltration was assessed by anti-CD68 and anti-CD206 staining. In vitro study, the effect of CEFFE on macrophage polarization and secretory factors was evaluated by flow cytometry, immunofluorescence, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The effect of CEFFE on cartilage regeneration was accessed by cell counting kit-8 assay and qRT-PCR. The generation of reactive oxygen species (ROS) and levels of ROS-related enzymes were investigated by qRT-PCR and western blotting. Results In rat models with sodium iodoacetate (MIA)-induced OA, CEFFE increased claw retraction pressure while decreasing bipedal pressure in a dose-dependent manner. Moreover, CEFFE promoted cartilage structure restoration and increased the proportion of CD206+ macrophages in the synovium. In vitro, CEFFE decreased the proportion of CD86+ cells and reduced the expression of pro-inflammatory factors in LPS + IFN-γ induced Raw 264.7. In addition, CEFFE decreased the expression of interleukin-6 and ADAMTs-5 and promoted the expression of SOX-9 in mouse primary chondrocytes. Besides, CEFFE reduced the intracellular levels of reactive oxygen species in both in vitro models through regulating ROS-related enzymes. Conclusions CEFFE inhibits the progression of OA by promoting cartilage regeneration and limiting low-grade joint inflammation. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02813-3.
Collapse
Affiliation(s)
- Zhuoxuan Jia
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Bijun Kang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Yizuo Cai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Chingyu Chen
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China
| | - Zheyuan Yu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China.
| | - Wei Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China.
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, 639 ZhiZaoJu Road, Shanghai, 200011, China.
| |
Collapse
|
23
|
Morgan M, Thai J, Nazemian V, Song R, Ivanusic JJ. Changes to the activity and sensitivity of nerves innervating subchondral bone contribute to pain in late-stage osteoarthritis. Pain 2022; 163:390-402. [PMID: 34108432 PMCID: PMC8756348 DOI: 10.1097/j.pain.0000000000002355] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 05/07/2021] [Indexed: 11/30/2022]
Abstract
ABSTRACT Although it is clear that osteoarthritis (OA) pain involves activation and/or sensitization of nociceptors that innervate knee joint articular tissues, much less is known about the role of the innervation of surrounding bone. In this study, we used monoiodoacetate (MIA)-induced OA in male rats to test the idea that pain in OA is driven by differential contributions from nerves that innervate knee joint articular tissues vs the surrounding bone. The time-course of pain behavior was assayed using the advanced dynamic weight-bearing device, and histopathology was examined using haematoxylin and eosin histology. Extracellular electrophysiological recordings of knee joint and bone afferent neurons were made early (day 3) and late (day 28) in the pathogenesis of MIA-induced OA. We observed significant changes in the function of knee joint afferent neurons, but not bone afferent neurons, at day 3 when there was histological evidence of inflammation in the joint capsule, but no damage to the articular cartilage or subchondral bone. Changes in the function of bone afferent neurons were only observed at day 28, when there was histological evidence of damage to the articular cartilage and subchondral bone. Our findings suggest that pain early in MIA-induced OA involves activation and sensitization of nerves that innervate the joint capsule but not the underlying subchondral bone, and that pain in late MIA-induced OA involves the additional recruitment of nerves that innervate the subchondral bone. Thus, nerves that innervate bone should be considered important targets for development of mechanism-based therapies to treat pain in late OA.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Vida Nazemian
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Richard Song
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Jason J. Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Minnema L, Gupta A, Mishra SK, Lascelles BDX. Investigating the Role of Artemin and Its Cognate Receptor, GFRα3, in Osteoarthritis Pain. Front Neurosci 2022; 16:738976. [PMID: 35153665 PMCID: PMC8829392 DOI: 10.3389/fnins.2022.738976] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) associated pain (OA-pain) is a significant global problem. OA-pain limits limb use and mobility and is associated with widespread sensitivity. Therapeutic options are limited, and the available options are often associated with adverse effects. The lack of therapeutic options is partly due to a lack of understanding of clinically relevant underlying neural mechanisms of OA-pain. In previous work in naturally occurring OA-pain in dogs, we identified potential signaling molecules (artemin/GFRα3) that were upregulated. Here, we use multiple approaches, including cellular, mouse genetic, immunological suppression in a mouse model of OA, and clinically relevant measures of sensitivity and limb use to explore the functional role of artemin/GFRα3 signaling in OA-pain. We found the monoiodoacetate (MIA)-induced OA-pain in mice is associated with decreased limb use and hypersensitivity. Exogenous artemin induces mechanical, heat, and cold hypersensitivity, and systemic intraperitoneal anti-artemin monoclonal antibody administration reverses this hypersensitivity and restores limb use in mice with MIA-induced OA-pain. An artemin receptor GFRα3 expression is increased in sensory neurons in the MIA model. Our results provide a molecular basis of arthritis pain linked with artemin/GFRα3 signaling and indicate that further work is warranted to investigate the neuronal plasticity and the pathways that drive pain in OA.
Collapse
Affiliation(s)
- Laura Minnema
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Ankita Gupta
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Santosh K. Mishra
- Department of Molecular and Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, United States
- *Correspondence: Santosh K. Mishra,
| | - B. Duncan X. Lascelles
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, United States
- Thurston Arthritis Center, UNC School of Medicine, Chapel Hill, NC, United States
- B. Duncan X. Lascelles,
| |
Collapse
|
25
|
Morgan M, Nazemian V, Harrington K, Ivanusic JJ. Mini review: The role of sensory innervation to subchondral bone in osteoarthritis pain. Front Endocrinol (Lausanne) 2022; 13:1047943. [PMID: 36605943 PMCID: PMC9808033 DOI: 10.3389/fendo.2022.1047943] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/30/2022] [Indexed: 01/07/2023] Open
Abstract
Osteoarthritis pain is often thought of as a pain driven by nerves that innervate the soft tissues of the joint, but there is emerging evidence for a role for nerves that innervate the underlying bone. In this mini review we cite evidence that subchondral bone lesions are associated with pain in osteoarthritis. We explore recent studies that provide evidence that sensory neurons that innervate bone are nociceptors that signal pain and can be sensitized in osteoarthritis. Finally, we describe neuronal remodeling of sensory and sympathetic nerves in bone and discuss how these processes can contribute to osteoarthritis pain.
Collapse
|
26
|
Micheli L, Parisio C, Lucarini E, Vona A, Toti A, Pacini A, Mello T, Boccella S, Ricciardi F, Maione S, Graziani G, Lacal PM, Failli P, Ghelardini C, Di Cesare Mannelli L. VEGF-A/VEGFR-1 signalling and chemotherapy-induced neuropathic pain: therapeutic potential of a novel anti-VEGFR-1 monoclonal antibody. J Exp Clin Cancer Res 2021; 40:320. [PMID: 34649573 PMCID: PMC8515680 DOI: 10.1186/s13046-021-02127-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/04/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Neuropathic pain is a clinically relevant adverse effect of several anticancer drugs that markedly impairs patients' quality of life and frequently leads to dose reduction or therapy discontinuation. The poor knowledge about the mechanisms involved in neuropathy development and pain chronicization, and the lack of effective therapies, make treatment of chemotherapy-induced neuropathic pain an unmet medical need. In this context, the vascular endothelial growth factor A (VEGF-A) has emerged as a candidate neuropathy hallmark and its decrease has been related to pain relief. In the present study, we have investigated the role of VEGF-A and its receptors, VEGFR-1 and VEGFR-2, in pain signalling and in chemotherapy-induced neuropathy establishment as well as the therapeutic potential of receptor blockade in the management of pain. METHODS Behavioural and electrophysiological analyses were performed in an in vivo murine model, by using selective receptor agonists, blocking monoclonal antibodies or siRNA-mediated silencing of VEGF-A and VEGFRs. Expression of VEGF-A and VEGFR-1 in astrocytes and neurons was detected by immunofluorescence staining and confocal microscopy analysis. RESULTS In mice, the intrathecal infusion of VEGF-A (VEGF165 isoforms) induced a dose-dependent noxious hypersensitivity and this effect was mediated by VEGFR-1. Consistently, electrophysiological studies indicated that VEGF-A strongly stimulated the spinal nociceptive neurons activity through VEGFR-1. In the dorsal horn of the spinal cord of animals affected by oxaliplatin-induced neuropathy, VEGF-A expression was increased in astrocytes while VEGFR-1 was mainly detected in neurons, suggesting a VEGF-A/VEGFR-1-mediated astrocyte-neuron cross-talk in neuropathic pain pathophysiology. Accordingly, the selective knockdown of astrocytic VEGF-A by intraspinal injection of shRNAmir blocked the development of oxaliplatin-induced neuropathic hyperalgesia and allodynia. Interestingly, both intrathecal and systemic administration of the novel anti-VEGFR-1 monoclonal antibody D16F7, endowed with anti-angiogenic and antitumor properties, reverted oxaliplatin-induced neuropathic pain. Besides, D16F7 effectively relieved hypersensitivity induced by other neurotoxic chemotherapeutic agents, such as paclitaxel and vincristine. CONCLUSIONS These data strongly support the role of the VEGF-A/VEGFR-1 system in mediating chemotherapy-induced neuropathic pain at the central nervous system level. Thus, treatment with the anti-VEGFR-1 mAb D16F7, besides exerting antitumor activity, might result in the additional advantage of attenuating neuropathic pain when combined with neurotoxic anticancer agents.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Alessia Vona
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine - DMSC - Anatomy and Histology Section, University of Florence, L.go Brambilla 3, 50134, Florence, Italy
| | - Tommaso Mello
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Flavia Ricciardi
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "L. Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
- I.R.C.S.S., Neuromed, 86077, Pozzilli, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
- IDI-IRCCS, Via Monti di Creta 104, 00167, Rome, Italy.
| | | | - Paola Failli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| |
Collapse
|
27
|
Morgan M, Thai J, Trinh P, Habib M, Effendi KN, Ivanusic JJ. ASIC3 inhibition modulates inflammation-induced changes in the activity and sensitivity of Aδ and C fiber sensory neurons that innervate bone. Mol Pain 2021; 16:1744806920975950. [PMID: 33280501 PMCID: PMC7724402 DOI: 10.1177/1744806920975950] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Acid Sensing Ion Channel 3 (ASIC3) is a non-selective cation channel that is
activated by acidification, and is known to have a role in regulating
inflammatory pain. It has pro-algesic roles in a range of conditions that
present with bone pain, but the mechanism for this has not yet been
demonstrated. We aimed to determine if ASIC3 is expressed in Aδ and/or C fiber
bone afferent neurons, and to explore its role in the activation and
sensitization of bone afferent neurons after acute inflammation. A combination
of retrograde tracing and immunohistochemistry was used to determine expression
of ASIC3 in the soma of bone afferent neurons. A novel, in
vivo, electrophysiological bone-nerve preparation was used to make
recordings of the activity and sensitivity of bone afferent neurons in the
presence of carrageenan-induced inflammation, with and without the selective
ASIC3 inhibitor APET×2. A substantial proportion of bone afferent neurons
express ASIC3, including unmyelinated (neurofilament poor) and small diameter
myelinated (neurofilament rich) neurons that are likely to be C and Aδ nerve
fibers respectively. Electrophysiological recordings revealed that application
of APET×2 to the marrow cavity inhibited carrageenan-induced spontaneous
activity of C and Aδ fiber bone afferent neurons. APET×2 also inhibited
carrageenan-induced sensitization of Aδ and C fiber bone afferent neurons to
mechanical stimulation, but had no effect on the sensitivity of bone afferent
neurons in the absence of inflammation. This evidence supports a role for ASIC3
in the pathogenesis of pain associated with inflammation of the bone.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Phu Trinh
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Mohamed Habib
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Kelly N Effendi
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
28
|
Nencini S, Morgan M, Thai J, Jobling AI, Mazzone SB, Ivanusic JJ. Piezo2 Knockdown Inhibits Noxious Mechanical Stimulation and NGF-Induced Sensitization in A-Delta Bone Afferent Neurons. Front Physiol 2021; 12:644929. [PMID: 34335288 PMCID: PMC8320394 DOI: 10.3389/fphys.2021.644929] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Piezo2 is a mechanically gated ion-channel that has a well-defined role in innocuous mechanical sensitivity, but recently has also been suggested to play a role in mechanically induced pain. Here we have explored a role for Piezo2 in mechanically evoked bone nociception in Sprague Dawley rats. We have used an in vivo electrophysiological bone-nerve preparation to record the activity of single Aδ bone afferent neurons in response to noxious mechanical stimulation, after Piezo2 knockdown in the dorsal root ganglia with intrathecal injections of Piezo2 antisense oligodeoxynucleotides, or in control animals that received mismatch oligodeoxynucleotides. There were no differences in the number of Aδ bone afferent neurons responding to the mechanical stimulus, or their threshold for mechanical activation, in Piezo2 knockdown animals compared to mismatch control animals. However, bone afferent neurons in Piezo2 knockdown animals had reduced discharge frequencies and took longer to recover from stimulus-evoked fatigue than those in mismatch control animals. Piezo2 knockdown also prevented nerve growth factor (NGF)-induced sensitization of bone afferent neurons, and retrograde labeled bone afferent neurons that expressed Piezo2 co-expressed TrkA, the high affinity receptor for NGF. Our findings demonstrate that Piezo2 contributes to the response of bone afferent neurons to noxious mechanical stimulation, and plays a role in processes that sensitize them to mechanical stimulation.
Collapse
Affiliation(s)
- Sara Nencini
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew I Jobling
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
29
|
Yang J, Yao L, Li Y, Gao R, Huo R, Xia L, Shen H, Lu J. Interleukin-35 Regulates Angiogenesis Through P38 Mitogen-Activated Protein Kinase Signaling Pathway in Interleukin-1β-Stimulated SW1353 Cells and Cartilage Bioinformatics Analysis. J Interferon Cytokine Res 2021; 41:164-171. [PMID: 34003680 DOI: 10.1089/jir.2021.0016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We aimed to investigate the effects of interleukin (IL)-35 on proangiogenic factors in IL-1β-pretreated chondrocyte-like SW1353 cells and screen-related genes that participated in osteoarthritis (OA) cartilage with IL-35, proangiogenic factors, and P38 mitogen-activated protein kinase (MAPK) signaling pathway. Different concentrations of IL-35 incubated with IL-1β stimulated SW1353 cells with or without SB203580 (inhibitor of P38 MAPK). Proangiogenic molecule expression was assessed by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Microarray datasets were downloaded from the Gene Expression Omnibus database of OA cartilage. Protein-protein interaction of genes was visualized by Search Tool for the Retrieval Interacting Genes and Cytoscape. Database for Annotation, Visualization, and Integrated Discovery was used to screen biological processes and pathways. IL-35 inhibited mRNA expression of proangiogenic factors in IL-1β-stimulated SW1353 cells through the P38 MAPK signaling pathway. IL-35 inhibited angiopoietin-2 secretion. We found that 8 related genes, 18 biological processes, and 6 pathways may associate with IL-35, P38 MAPK signaling pathway, and cartilage angiogenesis. IL-35 regulated the expression of proangiogenic factors through P38 MAPK signaling pathway in IL-1β-stimulated SW1353 cells. IL-35 and P38 MAPK pathway may participate in neovascularization of cartilage. Our findings may provide molecular mechanisms and possible genes target treatment for OA.
Collapse
Affiliation(s)
- Jie Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Lutian Yao
- Department of Sports Medicine and Joint Surgery, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Yuxuan Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, P.R. China
| | - Ruoxi Gao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Ran Huo
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Liping Xia
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Hui Shen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Jing Lu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
30
|
Huang HM, Huang CC, Poon LYC, Chang YC. Artemin Is Upregulated by TrkB Agonist and Protects the Immature Retina Against Hypoxic-Ischemic Injury by Suppressing Neuroinflammation and Astrogliosis. Front Mol Neurosci 2021; 14:645000. [PMID: 33912011 PMCID: PMC8072488 DOI: 10.3389/fnmol.2021.645000] [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: 12/22/2020] [Accepted: 03/17/2021] [Indexed: 11/22/2022] Open
Abstract
Hypoxic-ischemia (HI) is a major cause of acquired visual impairment in children from developed countries. Previous studies have shown that systemic administration of 7,8-dihydroxyavone (DHF), a selective tropomyosin receptor kinase B (TrkB) agonist, provides long-term neuroprotection against HI injury in an immature retina. However, the target genes and the mechanisms of the neuroprotective effects of TrkB signaling are not known. In the present study, we induced an HI retinal injury through unilateral common carotid artery ligation followed by 8% oxygen for 2 h in P7 rat pups. DHF was administered intraperitoneally 2 h before and 18 h after the HI injury. A polymerase chain reaction (PCR) array was used to identify the target genes upregulated after the DHF treatment, which was then confirmed with quantitative real-time reverse transcriptase PCR and a western blot. Effects of the downstream mediator of DHF were assessed using an intravitreal injection of neutralizing antibody 4 h after DHF administration (24 h after HI). Meanwhile, the target protein was injected into the vitreous 24 h after HI to validate its protective effect when exogenously supplemented. We found that systemic DHF treatment after HI significantly increased the expression of the artemin (ARTN) gene and protein at P8 and P10, respectively. The neuroprotective effects of DHF were inhibited after the ARTN protein blockade, with an increase in neuroinflammation and astrogliosis. ARTN treatment showed long-term protection against HI injury at both the histopathological and functional levels. The neuroprotective effects of ARTN were related to a decrease in microglial activation at P17 and attenuation of astrogliosis at P29. ARTN enhances phosphorylation of RET, ERK, and JNK, but not AKT or p38 in the immature retina. Altogether, these results suggest that the neuroprotective effect of a TrkB agonist is partially exerted through a mechanism that involves ARTN because the protective effect is ameliorated by ARTN sequestration. ARTN treatment after HI injury protects the immature retina by attenuating late neuroinflammation and astrogliosis in the immature retina relating to the ARTN/RET/JNK/ERK signaling pathway. ARTN may be a strategy by which to provide long-term protection in the immature retina against HI injury.
Collapse
Affiliation(s)
- Hsiu-Mei Huang
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Chao-Ching Huang
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Linda Yi-Chieh Poon
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| | - Ying-Chao Chang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, Taiwan
| |
Collapse
|
31
|
Wan Q, Qin W, Ma Y, Shen M, Li J, Zhang Z, Chen J, Tay FR, Niu L, Jiao K. Crosstalk between Bone and Nerves within Bone. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003390. [PMID: 33854888 PMCID: PMC8025013 DOI: 10.1002/advs.202003390] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/29/2020] [Indexed: 05/11/2023]
Abstract
For the past two decades, the function of intrabony nerves on bone has been a subject of intense research, while the function of bone on intrabony nerves is still hidden in the corner. In the present review, the possible crosstalk between bone and intrabony peripheral nerves will be comprehensively analyzed. Peripheral nerves participate in bone development and repair via a host of signals generated through the secretion of neurotransmitters, neuropeptides, axon guidance factors and neurotrophins, with additional contribution from nerve-resident cells. In return, bone contributes to this microenvironmental rendezvous by housing the nerves within its internal milieu to provide mechanical support and a protective shelf. A large ensemble of chemical, mechanical, and electrical cues works in harmony with bone marrow stromal cells in the regulation of intrabony nerves. The crosstalk between bone and nerves is not limited to the physiological state, but also involved in various bone diseases including osteoporosis, osteoarthritis, heterotopic ossification, psychological stress-related bone abnormalities, and bone related tumors. This crosstalk may be harnessed in the design of tissue engineering scaffolds for repair of bone defects or be targeted for treatment of diseases related to bone and peripheral nerves.
Collapse
Affiliation(s)
- Qian‐Qian Wan
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Wen‐Pin Qin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Yu‐Xuan Ma
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Min‐Juan Shen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Jing Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Zi‐Bin Zhang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Ji‐Hua Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Franklin R. Tay
- College of Graduate StudiesAugusta UniversityAugustaGA30912USA
| | - Li‐Na Niu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Kai Jiao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032China
| |
Collapse
|
32
|
Serum artemin is not correlated with sensitivity within dogs with naturally occurring osteoarthritis pain. Sci Rep 2021; 11:6682. [PMID: 33758254 PMCID: PMC7988108 DOI: 10.1038/s41598-021-85976-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 03/03/2021] [Indexed: 11/14/2022] Open
Abstract
Osteoarthritis (OA) pain is associated with peripheral and central sensitization in humans and results in widespread increased sensitivity across the body. Sensitization contributes to the OA-associated pain (OAP) state. We recently identified increased levels of an endogenous neurotrophic factor, artemin (ARTN), in dogs with OAP compared to healthy pain-free controls. Circulating ARTN released from damaged tissues in OA, may play a central role in widespread sensitivity and pain. However, the relationship between ARTN and somatosensory sensitivity remains unknown. The study aimed to assess the relationship between serum ARTN concentrations and measures of sensitivity in dogs with OAP using quantitative sensory testing. We hypothesized that there would be a positive association between circulating ARTN and increased sensitivity to mechanical and thermal stimuli in dogs with OAP. We used linear and logistic regression models to assess the relationship between ARTN, sensitization, and pain within a cohort of 43 dogs with spontaneous OAP. Serum ARTN was not associated with the degree of sensitization within dogs with OAP. Further, across dogs with varying OAP severity, we did not find any association between ARTN, and clinical measures of joint pain and disability. Although a relationship between ARTN and joint pain was not ruled out.
Collapse
|
33
|
Murase S, Kobayashi K, Nasu T, Kihara C, Taguchi T, Mizumura K. Synergistic interaction of nerve growth factor and glial cell‐line derived neurotrophic factor in muscular mechanical hyperalgesia in rats. J Physiol 2021; 599:1783-1798. [DOI: 10.1113/jp280683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/18/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Shiori Murase
- Department of Physical Therapy College of Life Sciences Chubu University Kasugai 487–8501 Japan
- Department of Neuroscience II Research Institute of Environmental Medicine Nagoya University Nagoya 464–8601 Japan
| | - Kimiko Kobayashi
- Department of Anatomy and Neuroscience Hyogo College of Medicine Nishinomiya 663–8501 Japan
| | - Teruaki Nasu
- Department of Physical Therapy College of Life Sciences Chubu University Kasugai 487–8501 Japan
- Department of Neuroscience II Research Institute of Environmental Medicine Nagoya University Nagoya 464–8601 Japan
| | - Chiaki Kihara
- Department of Physical Therapy College of Life Sciences Chubu University Kasugai 487–8501 Japan
| | - Toru Taguchi
- Department of Neuroscience II Research Institute of Environmental Medicine Nagoya University Nagoya 464–8601 Japan
- Department of Physical Therapy Faculty of Rehabilitation Niigata University of Health and Welfare Niigata 950–3198 Japan
- Institute for Human Movement and Medical Sciences Niigata University of Health and Welfare Niigata 950–3198 Japan
| | - Kazue Mizumura
- Department of Physical Therapy College of Life Sciences Chubu University Kasugai 487–8501 Japan
- Department of Neuroscience II Research Institute of Environmental Medicine Nagoya University Nagoya 464–8601 Japan
- Department of Physiology Nihon University School of Dentistry Tokyo 101–8310 Japan
| |
Collapse
|
34
|
Chen P, Wang C, Lin D, Li B, Ye S, Qu J, Wang W. Identification of Slc6a19os and SOX11 as Two Novel Essential Genes in Neuropathic Pain Using Integrated Bioinformatic Analysis and Experimental Verification. Front Neurosci 2021; 15:627945. [PMID: 33584192 PMCID: PMC7876402 DOI: 10.3389/fnins.2021.627945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/04/2021] [Indexed: 12/25/2022] Open
Abstract
The aim of this study was to identify critical genes associated with neuropathic pain. We also used the competing endogenous RNA (ceRNA) hypothesis to identify related long non-coding RNAs (lncRNAs) and messenger RNAs (miRNAs) with potential regulatory roles. We downloaded GSE107180 from the Gene Expression Omnibus database, screened differentially expressed genes (DEGs) using R software, performed comprehensive bioinformatic analyses, and validated the expression of lncRNA Slc6a19os, miR-125a-5p, miR-125b-5p, miR-351-5p, and Sox11 by qRT-PCR and Western blots. We identified 620 DEGs in spared nerve injury (SNI) mice compared with sham (control) mice, including 309 mRNAs and 311 non-coding RNAs. The up-regulated mRNAs were enriched primarily in several inflammation-related GO biological processes and KEGG signaling pathways. A ceRNA network was constructed that included 82 mRNAs, 4 miRNAs, and 2 lnRNAs. An ingenuity pathway analysis (IPA)-based interaction network for mRNAs differentially expressed in the ceRNA identified several biological processes, including "cellular development, connective tissue development and function, tissue development." Compared with sham mice, lncRNA Slc6a19os and Sox11 expression were significantly up-regulated in dorsal root ganglion (DRG) samples from SNI mice detected using qRT-PCR and Western blots (P < 0.05). MiR-125a-5p, miR-125b-5p, and miR-351-5p expression were down-regulated in DRG samples from SNI mice detected using qRT-PCR (P < 0.05). We concluded that Sox11 and lncRNA Slc6a19os were novel essential genes in the pathogenesis and progression of neuropathic pain and speculated that these two genes were regulated by miR-125a-5p, miR-125b-5p, and miR-351-5p.
Collapse
Affiliation(s)
- Peng Chen
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Chen Wang
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongsheng Lin
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bing Li
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shuai Ye
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Jinglian Qu
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wenjing Wang
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
35
|
Ferrini F, Salio C, Boggio EM, Merighi A. Interplay of BDNF and GDNF in the Mature Spinal Somatosensory System and Its Potential Therapeutic Relevance. Curr Neuropharmacol 2021; 19:1225-1245. [PMID: 33200712 PMCID: PMC8719296 DOI: 10.2174/1570159x18666201116143422] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 11/22/2022] Open
Abstract
The growth factors BDNF and GDNF are gaining more and more attention as modulators of synaptic transmission in the mature central nervous system (CNS). The two molecules undergo a regulated secretion in neurons and may be anterogradely transported to terminals where they can positively or negatively modulate fast synaptic transmission. There is today a wide consensus on the role of BDNF as a pro-nociceptive modulator, as the neurotrophin has an important part in the initiation and maintenance of inflammatory, chronic, and/or neuropathic pain at the peripheral and central level. At the spinal level, BDNF intervenes in the regulation of chloride equilibrium potential, decreases the excitatory synaptic drive to inhibitory neurons, with complex changes in GABAergic/glycinergic synaptic transmission, and increases excitatory transmission in the superficial dorsal horn. Differently from BDNF, the role of GDNF still remains to be unraveled in full. This review resumes the current literature on the interplay between BDNF and GDNF in the regulation of nociceptive neurotransmission in the superficial dorsal horn of the spinal cord. We will first discuss the circuitries involved in such a regulation, as well as the reciprocal interactions between the two factors in nociceptive pathways. The development of small molecules specifically targeting BDNF, GDNF and/or downstream effectors is opening new perspectives for investigating these neurotrophic factors as modulators of nociceptive transmission and chronic pain. Therefore, we will finally consider the molecules of (potential) pharmacological relevance for tackling normal and pathological pain.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- Department of Psychiatry & Neuroscience, Université Laval, Québec, Canada
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Elena M. Boggio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- National Institute of Neuroscience, Grugliasco, Italy
| |
Collapse
|
36
|
Xuan J, Zhu D, Cheng Z, Qiu Y, Shao M, Yang Y, Zhai Q, Wang F, Qin F. Crocin inhibits the activation of mouse hepatic stellate cells via the lnc-LFAR1/MTF-1/GDNF pathway. Cell Cycle 2020; 19:3480-3490. [PMID: 33295246 PMCID: PMC7781632 DOI: 10.1080/15384101.2020.1848064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 01/20/2023] Open
Abstract
Crocin is the main monomer of saffron, which is a momentous component of traditional Chinese medicine Lang Qing A Ta. Here, we tried to probe into the role of crocin in liver fibrosis. Hematoxylin-eosin staining and Sirius Red staining were used to observe the pathological changes of liver tissues. After hepatic stellate cells (HSCs) were isolated from liver tissues, lnc-LFAR1, MTF-1, GDNF, and α-SMA expressions were detected by qRT-PCR and western blot. Immunohistochemistry and immunofluorescence were used to detect α-SMA expression. Chromatin immunoprecipitation was used to analyze the binding of MTF-1 to the GDNF promoter. Moreover, the dual-luciferase reporter gene, RNA pull-down, and RNA immunoprecipitation were used to clarify the interaction between MTF-1 and GDNF, lnc-LFAR1 and MTF-1. The degree of liver fibrosis was more severe in the mice from the liver fibrosis model, while the liver fibrosis was alleviated by the injection of crocin. lnc-LFAR1, GDNF, and α-SMA were up-regulated, and MTF-1 was down-regulated in liver fibrosis tissues and cells, while these trends were reversed after the injection of crocin. Besides, lnc-LFAR1 negatively regulated MTF-1 expression, and positively regulated GDNF and α-SMA expressions, and MTF-1 was enriched in the promoter region of GDNF. Furthermore, the cellular direct interactions between MTF-1 and GDNF, lnc-LFAR1 and MTF-1 were verified. In vivo experiments confirmed the relief of crocin on liver fibrosis. Our research expounded that crocin restrained the activation of HSCs through the lnc-LFAR1/MTF-1/GDNF axis, thereby ameliorating liver fibrosis.
Collapse
Affiliation(s)
- Ji Xuan
- Department of Gastroenterology, Jinling Hospital, Nanjing, China
| | - Dongmei Zhu
- Department of Nursing, Jinling Hospital, Nanjing, China
| | - Zhengyuan Cheng
- Department of Gastroenterology, Jinling Hospital, Nanjing, China
| | - Yuping Qiu
- Department of Gastroenterology, Jinling Hospital, Nanjing, China
| | - Mei Shao
- Department of Gastroenterology, Jinling Hospital, Nanjing, China
| | - Ya Yang
- Department of Gastroenterology, Jinling Hospital, Nanjing, China
| | - Qi Zhai
- Department of Gastroenterology, Jinling Hospital, Nanjing, China
| | - Fangyu Wang
- Department of Gastroenterology, Jinling Hospital, Nanjing, China
| | - Feng Qin
- Jinling Hospital, Nanjing, China
| |
Collapse
|
37
|
von Loga IS, Batchelor V, Driscoll C, Burleigh A, Chia SLL, Stott B, Miotla-Zarebska J, Riley D, Dell'Accio F, Vincent TL. Does Pain at an Earlier Stage of Chondropathy Protect Female Mice Against Structural Progression After Surgically Induced Osteoarthritis? Arthritis Rheumatol 2020; 72:2083-2093. [PMID: 32602242 DOI: 10.1002/art.41421] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 06/11/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Female C57BL/6 mice exhibit less severe chondropathy than male mice. This study was undertaken to test the robustness of this observation and explore underlying mechanisms. METHODS Osteoarthritis was induced in male and female C57BL/6 or DBA/1 mice (n = 6-15 per group) by destabilization of the medial meniscus (DMM) or partial meniscectomy (PMX). Some mice were ovariectomized (OVX) (n = 30). In vivo repair after focal cartilage defect or joint immobilization (sciatic neurectomy) following DMM was assessed. Histologic analysis, evaluation of gene expression in whole knees, and behavioral analysis using Laboratory Animal Behavior Observation Registration and Analysis System (LABORAS) and Linton incapacitance testing (n = 7-10 mice per group) were performed. RESULTS Female mice displayed less severe chondropathy (20-75% reduction) across both strains and after both surgeries. Activity levels after PMX were similar for male and female mice. Some repair-associated genes were increased in female mouse joints after surgery, but no repair differences were evident in vivo. Despite reduced chondropathy, female mice developed pain-like behavior at the same time as male mice. At the time of established pain-like behavior (10 weeks after PMX), pain-associated genes were significantly up-regulated in female mice, including Gdnf (mean ± SEM fold change 2.54 ± 0.30), Nrtn (6.71 ± 1.24), Ntf3 (1.92 ± 0.27), and Ntf5 (2.89 ± 0.48) (P < 0.01, P < 0.01, P < 0.05, and P < 0.001, respectively, versus male mice). Inflammatory genes were not regulated in painful joints in mice of either sex. CONCLUSION We confirm strong structural joint protection in female mice that is not due to activity or intrinsic repair differences. Female mice develop pain at the same time as males, but induce a distinct set of neurotrophins. We speculate that heightened pain sensitivity in female mice protects the joint by preventing overuse.
Collapse
Affiliation(s)
| | - Vicky Batchelor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Clare Driscoll
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Annika Burleigh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Shi-Lu L Chia
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Bryony Stott
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - David Riley
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Tonia L Vincent
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
de Clauser L, Luiz AP, Santana-Varela S, Wood JN, Sikandar S. Sensitization of Cutaneous Primary Afferents in Bone Cancer Revealed by In Vivo Calcium Imaging. Cancers (Basel) 2020; 12:cancers12123491. [PMID: 33255209 PMCID: PMC7760605 DOI: 10.3390/cancers12123491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Cancer-induced bone pain severely impairs the quality of life of cancer patients, many of whom suffer from inadequate pain relief. The development of new analgesic therapies depends on the identification of the cells and mechanisms involved in cancer-induced bone pain. Bone marrow innervating sensory neurons have been proposed to contribute to this debilitating disease, but their role remains unexplored. Here we used in vivo calcium imaging to determine the functional role of bone innervating and skin innervating neurons in contributing to pain at an advanced stage of bone cancer. Our results indicate increased excitability of skin innervating neurons, while those innervating bone are unaffected. Our data suggests skin-innervating neurons become hyperexcitable in cancer-induced bone pain and are a potential target for pain relief. Abstract Cancer-induced bone pain (CIBP) is a complex condition, comprising components of inflammatory and neuropathic processes, but changes in the physiological response profiles of bone-innervating and cutaneous afferents remain poorly understood. We used a combination of retrograde labelling and in vivo calcium imaging of bone marrow-innervating dorsal root ganglia (DRG) neurons to determine the contribution of these cells in the maintenance of CIBP. We found a majority of femoral bone afferent cell bodies in L3 dorsal root ganglia (DRG) that also express the sodium channel subtype Nav1.8—a marker of nociceptive neurons—and lack expression of parvalbumin—a marker for proprioceptive primary afferents. Surprisingly, the response properties of bone marrow afferents to both increased intraosseous pressure and acid were unchanged by the presence of cancer. On the other hand, we found increased excitability and polymodality of cutaneous afferents innervating the ipsilateral paw in cancer bearing animals, as well as a behavioural phenotype that suggests changes at the level of the DRG contribute to secondary hypersensitivity. This study demonstrates that cutaneous afferents at distant sites from the tumour bearing tissue contribute to mechanical hypersensitivity, highlighting these cells as targets for analgesia.
Collapse
Affiliation(s)
- Larissa de Clauser
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE1 1UL, UK
| | - Ana P. Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
| | - Shafaq Sikandar
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK; (L.d.C.); (A.P.L.); (S.S.-V.); (J.N.W.)
- William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Correspondence:
| |
Collapse
|
39
|
Mahato AK, Sidorova YA. Glial cell line-derived neurotrophic factors (GFLs) and small molecules targeting RET receptor for the treatment of pain and Parkinson's disease. Cell Tissue Res 2020; 382:147-160. [PMID: 32556722 PMCID: PMC7529621 DOI: 10.1007/s00441-020-03227-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
Abstract
Rearranged during transfection (RET), in complex with glial cell line-derived (GDNF) family receptor alpha (GFRα), is the canonical signaling receptor for GDNF family ligands (GFLs) expressed in both central and peripheral parts of the nervous system and also in non-neuronal tissues. RET-dependent signaling elicited by GFLs has an important role in the development, maintenance and survival of dopamine and sensory neurons. Both Parkinson's disease and neuropathic pain are devastating disorders without an available cure, and at the moment are only treated symptomatically. GFLs have been studied extensively in animal models of Parkinson's disease and neuropathic pain with remarkable outcomes. However, clinical trials with recombinant or viral vector-encoded GFL proteins have produced inconclusive results. GFL proteins are not drug-like; they have poor pharmacokinetic properties and activate multiple receptors. Targeting RET and/or GFRα with small molecules may resolve the problems associated with using GFLs as drugs and can result in the development of therapeutics for disease-modifying treatments against Parkinson's disease and neuropathic pain.
Collapse
Affiliation(s)
- Arun Kumar Mahato
- Institute of Biotechnology, HiLIFE, University of Helsinki, Viikinkaari 5D, 00014, Helsinki, Finland
| | - Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Viikinkaari 5D, 00014, Helsinki, Finland.
| |
Collapse
|
40
|
Zhu S, Li Y, Bennett S, Chen J, Weng IZ, Huang L, Xu H, Xu J. The role of glial cell line-derived neurotrophic factor family member artemin in neurological disorders and cancers. Cell Prolif 2020; 53:e12860. [PMID: 32573073 PMCID: PMC7377943 DOI: 10.1111/cpr.12860] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/17/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Artemin (ARTN) is a member of the glial cell line‐derived neurotrophic factor (GDNF) family ligands (GFLs), which encompasses family members, GDNF, neurturin (NRTN) and persephin (PSPN). ARTN is also referred to as Enovin or Neublastin, and bears structural characteristics of the TGF‐β superfamily. ARTN contains a dibasic cleavage site (RXXR) that is predicted to be cleaved by furin to yield a carboxy‐terminal 113 amino acid mature form. ARTN binds preferentially to receptor GFRα3, coupled to a receptor tyrosine kinase RET, forming a signalling complex for the regulation of intracellular pathways that affect diverse outcomes of nervous system development and homoeostasis. Standard signalling cascades activated by GFLs via RET include the phosphorylation of mitogen‐activated protein kinase or MAPK (p‐ERK, p‐p38 and p‐JNK), PI3K‐AKT and Src. Neural cell adhesion molecule (NCAM) is an alternative signalling receptor for ARTN in the presence of GFRα1, leading to activation of Fyn and FAK. Further, ARTN also interacts with heparan sulphate proteoglycan syndecan‐3 and mediates non‐RET signalling via activation of Src kinases. This review discusses the role of ARTN in spinal cord injury, neuropathic pain and other neurological disorders. Additionally, ARTN plays a role in non‐neuron tissues, such as the formation of Peyer's patch‐like structures in the lymphoid tissue of the gut. The emerging role of ARTN in cancers and therapeutic resistance to cancers is also explored. Further research is necessary to determine the function of ARTN in a tissue‐specific manner, including its signalling mechanisms, in order to improve the therapeutic potential of ARTN in human diseases.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Yihe Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Samuel Bennett
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Junhao Chen
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Isabel Ziwai Weng
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Lin Huang
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Orthopaedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
41
|
Malfait AM, Miller RE, Block JA. Targeting neurotrophic factors: Novel approaches to musculoskeletal pain. Pharmacol Ther 2020; 211:107553. [PMID: 32311372 DOI: 10.1016/j.pharmthera.2020.107553] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022]
Abstract
Chronic pain represents a substantial unmet medical need globally. In recent years, the quest for a new generation of novel, safe, mechanism-based analgesic treatments has focused on neurotrophic factors, a large group of secreted proteins that control the growth and survival of different populations of neurons, but that postnatally are involved in the genesis and maintenance of pain, with biological activity in both the periphery and the central nervous system. In this narrative review, we discuss the two families of neurotrophic proteins that have been extensively studied for their role in pain: first, the neurotrophins, nerve growth factor (NGF) and brain-derived growth factor (BDNF), and secondly, the GDNF family of ligands (GFLs). We provide an overview of the pain pathway, and the pain-producing effects of these different proteins. We summarize accumulating preclinical and clinical findings with a focus on musculoskeletal pain, and on osteoarthritis in particular, because the musculoskeletal system is the most prevalent source of chronic pain and of disability, and clinical testing of these novel agents - often biologics- is most advanced in this area.
Collapse
Affiliation(s)
- Anne-Marie Malfait
- Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL 60612, United States of America
| | - Rachel E Miller
- Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL 60612, United States of America
| | - Joel A Block
- Division of Rheumatology, Rush University Medical Center, 1611 W Harrison Street, Suite 510, Chicago, IL 60612, United States of America.
| |
Collapse
|
42
|
Morel L, Domingues O, Zimmer J, Michel T. Revisiting the Role of Neurotrophic Factors in Inflammation. Cells 2020; 9:cells9040865. [PMID: 32252363 PMCID: PMC7226825 DOI: 10.3390/cells9040865] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The neurotrophic factors are well known for their implication in the growth and the survival of the central, sensory, enteric and parasympathetic nervous systems. Due to these properties, neurturin (NRTN) and Glial cell-derived neurotrophic factor (GDNF), which belong to the GDNF family ligands (GFLs), have been assessed in clinical trials as a treatment for neurodegenerative diseases like Parkinson’s disease. In addition, studies in favor of a functional role for GFLs outside the nervous system are accumulating. Thus, GFLs are present in several peripheral tissues, including digestive, respiratory, hematopoietic and urogenital systems, heart, blood, muscles and skin. More precisely, recent data have highlighted that different types of immune and epithelial cells (macrophages, T cells, such as, for example, mucosal-associated invariant T (MAIT) cells, innate lymphoid cells (ILC) 3, dendritic cells, mast cells, monocytes, bronchial epithelial cells, keratinocytes) have the capacity to release GFLs and express their receptors, leading to the participation in the repair of epithelial barrier damage after inflammation. Some of these mechanisms pass on to ILCs to produce cytokines (such as IL-22) that can impact gut microbiota. In addition, there are indications that NRTN could be used in the treatment of inflammatory airway diseases and it prevents the development of hyperglycemia in the diabetic rat model. On the other hand, it is suspected that the dysregulation of GFLs produces oncogenic effects. This review proposes the discussion of the biological understanding and the potential new opportunities of the GFLs, in the perspective of developing new treatments within a broad range of human diseases.
Collapse
|
43
|
Minnema L, Wheeler J, Enomoto M, Pitake S, Mishra SK, Lascelles BDX. Correlation of Artemin and GFRα3 With Osteoarthritis Pain: Early Evidence From Naturally Occurring Osteoarthritis-Associated Chronic Pain in Dogs. Front Neurosci 2020; 14:77. [PMID: 32116521 PMCID: PMC7031206 DOI: 10.3389/fnins.2020.00077] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Arthritis, including osteoarthritis (OA) and other musculoskeletal-associated pain, is a worldwide problem, however, effective drug options are limited. Several receptors, neurotransmitters, and endogenous mediators have been identified in rodent models, but the relevance of these molecules in disease-associated pain is not always clear. Artemin, a neurotrophic factor, and its receptor, glial-derived neurotrophic factor (GDNF) family receptor alpha-3 (GFRα3), have been identified as involved in pain in rodents. Their role in OA-associated pain is unknown. To explore a possible association, we analyzed tissue from naturally occurring OA in dogs to characterize the correlation with chronic pain. We used behavioral assessment, objective measures of limb use, and molecular tools to identify whether artemin and GFRα3 might be associated with OA pain. Our results using banked tissue from well-phenotyped dogs indicates that artemin/GFRα3 may play an important, and hitherto unrecognized, role in chronic OA-associated pain. Elevated serum levels of artemin from osteoarthritic humans compared to healthy individuals suggest translational relevance. Our data provide compelling evidence that the artemin/GFRα3 signaling pathway may be important in OA pain in both non-humans and humans and may ultimately lead to novel therapeutics.
Collapse
Affiliation(s)
- Laura Minnema
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Joshua Wheeler
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Masataka Enomoto
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Saumitra Pitake
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Santosh K Mishra
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - B Duncan X Lascelles
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Thurston Arthritis Research Center, UNC School of Medicine, Chapel Hill, NC, United States.,Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, NC, United States
| |
Collapse
|
44
|
Thai J, Kyloh M, Travis L, Spencer NJ, Ivanusic JJ. Identifying spinal afferent (sensory) nerve endings that innervate the marrow cavity and periosteum using anterograde tracing. J Comp Neurol 2020; 528:1903-1916. [PMID: 31970770 DOI: 10.1002/cne.24862] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 01/04/2023]
Abstract
While sensory and sympathetic neurons are known to innervate bone, previous studies have found it difficult to unequivocally identify and characterize only those that are of sensory origin. In this study, we have utilized an in vivo anterograde tracing technique to selectively label spinal afferent (sensory) nerve endings that innervate the periosteum and marrow cavity of murine long bones. Unilateral injections of dextran-biotin (anterograde tracer; 20% in saline, 50-100 nl) were made into L3-L5 dorsal root ganglia. After a 10-day recovery period to allow sufficient time for selective anterograde transport of the tracer to nerve terminal endings in bone, the periosteum (whole-mount) and underlying bone were collected, processed to reveal anterograde labeling, and immuno-labeled with antibodies directed against protein gene product (pan-neuronal marker; PGP9.5), tyrosine hydroxylase (sympathetic neuron marker; TH), calcitonin gene-related protein (peptidergic nociceptor marker; CGRP), and/or neurofilament 200 (myelinated axon marker; NF200). Anterograde-labeled nerve endings were dispersed throughout the periosteum and marrow cavity and could be identified in close apposition to blood vessels and at sites distant from them. The periosteum and the marrow cavity were each innervated by myelinated (NF200+) sensory neurons, and unmyelinated (NF200-) sensory neurons that were either peptidergic (CGRP+) or nonpeptidergic (CGRP-). Spinal afferent nerve endings did not express TH, and lacked the cylindrical morphology around blood vessels characteristic of sympathetic innervation. This approach to selective labeling of sensory nerve terminal endings will help to better identify how different sub-populations of sensory neurons, and their peripheral nerve terminal endings, interact with bone.
Collapse
Affiliation(s)
- Jenny Thai
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Melinda Kyloh
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Lee Travis
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Nick J Spencer
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
45
|
Peripheral nociceptive mechanisms in an experimental rat model of fibromyalgia induced by repeated cold stress. Neurosci Res 2019; 162:22-30. [PMID: 31891739 DOI: 10.1016/j.neures.2019.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/06/2019] [Accepted: 12/19/2019] [Indexed: 01/15/2023]
Abstract
Fibromyalgia (FM) is a debilitating disease characterized by generalized and persistent musculoskeletal pain. Although central mechanisms are strongly implicated in the pathogenesis of FM, the involvement of peripheral mechanisms is poorly understood. To understand the peripheral nociceptive mechanisms, we examined muscular nociceptors in an FM model, which was made by exposing rats to repeated cold stress (RCS). A single muscle C-fiber nociceptors were identified through the teased fiber technique using ex vivo muscle-nerve preparations. Response properties of C-fibers to noxious stimuli were systematically analyzed. Messenger RNA expression of neurotrophic factors and inflammatory mediators were also studied in the muscle. In the RCS group, the mechanical response threshold of C-fibers, measured using a ramp mechanical stimulus, was significantly decreased, and the response magnitude was significantly increased in the RCS group when compared with the SHAM group, where the environmental temperature was not altered. The general characteristics of C-fibers and the responsiveness to noxious cold and heat stimuli were similar between the two groups. Messenger RNAs of neurotrophic factors and inflammatory mediators were not changed in the muscle during and after RCS. These results suggest that augmentation of the mechanical response of muscle C-fiber nociceptors contributes to hyperalgesia in the RCS model.
Collapse
|
46
|
Tao L, Ma W, Wu L, Xu M, Yang Y, Zhang W, Sha W, Li H, Xu J, Feng R, Xue D, Zhang J, Dooley S, Seki E, Liu P, Liu C. Glial cell line-derived neurotrophic factor (GDNF) mediates hepatic stellate cell activation via ALK5/Smad signalling. Gut 2019; 68:2214-2227. [PMID: 31171625 PMCID: PMC6842044 DOI: 10.1136/gutjnl-2018-317872] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Although glial cell line-derived neurotrophic factor (GDNF) is a member of the transforming growth factor-β superfamily, its function in liver fibrosis has rarely been studied. Here, we investigated the role of GDNF in hepatic stellate cell (HSC) activation and liver fibrosis in humans and mice. DESIGN GDNF expression was examined in liver biopsies and sera from patients with liver fibrosis. The functional role of GDNF in liver fibrosis was examined in mice with adenoviral delivery of the GDNF gene, GDNF sgRNA CRISPR/Cas9 and the administration of GDNF-blocking antibodies. GDNF was examined on HSC activation using human and mouse primary HSCs. The binding of activin receptor-like kinase 5 (ALK5) to GDNF was determined using surface plasmon resonance (SPR), molecular docking, mutagenesis and co-immunoprecipitation. RESULTS GDNF mRNA and protein levels are significantly upregulated in patients with stage F4 fibrosis. Serum GDNF content correlates positively with α-smooth muscle actin (α-SMA) and Col1A1 mRNA in human fibrotic livers. Mice with overexpressed GDNF display aggravated liver fibrosis, while mice with silenced GDNF expression or signalling inhibition by GDNF-blocking antibodies have reduced fibrosis and HSC activation. GDNF is confined mainly to HSCs and contributes to HSC activation through ALK5 at His39 and Asp76 and through downstream signalling via Smad2/3, but not through GDNF family receptor alpha-1 (GFRα1). GDNF, ALK5 and α-SMA colocalise in human and mouse HSCs, as demonstrated by confocal microscopy. CONCLUSIONS GDNF promotes HSC activation and liver fibrosis through ALK5/Smad signalling. Inhibition of GDNF could be a novel therapeutic strategy to combat liver fibrosis.
Collapse
Affiliation(s)
- Le Tao
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenting Ma
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Wu
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingyi Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqin Yang
- Department of Pathology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Zhang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Sha
- Departmentof Endocrinology and Metabolism, PutuoHospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongshan Li
- Department of Hepatology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Jianrong Xu
- Department of Pharmacology and Chemical Biology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rilu Feng
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dongying Xue
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Zhang
- Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ekihiro Seki
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ping Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China,Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Liu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Laboratory of Liver Disease, Department of Infectious Disease, PutuoHospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Shanghai, China
| |
Collapse
|
47
|
Peters CM, Muñoz-Islas E, Ramírez-Rosas MB, Jiménez-Andrade JM. Mechanisms underlying non-malignant skeletal pain. CURRENT OPINION IN PHYSIOLOGY 2019. [DOI: 10.1016/j.cophys.2019.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
48
|
Brazill JM, Beeve AT, Craft CS, Ivanusic JJ, Scheller EL. Nerves in Bone: Evolving Concepts in Pain and Anabolism. J Bone Miner Res 2019; 34:1393-1406. [PMID: 31247122 PMCID: PMC6697229 DOI: 10.1002/jbmr.3822] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/28/2019] [Accepted: 06/18/2019] [Indexed: 12/21/2022]
Abstract
The innervation of bone has been described for centuries, and our understanding of its function has rapidly evolved over the past several decades to encompass roles of subtype-specific neurons in skeletal homeostasis. Current research has been largely focused on the distribution and function of specific neuronal populations within bone, as well as their cellular and molecular relationships with target cells in the bone microenvironment. This review provides a historical perspective of the field of skeletal neurobiology that highlights the diverse yet interconnected nature of nerves and skeletal health, particularly in the context of bone anabolism and pain. We explore what is known regarding the neuronal subtypes found in the skeleton, their distribution within bone compartments, and their central projection pathways. This neuroskeletal map then serves as a foundation for a comprehensive discussion of the neural control of skeletal development, homeostasis, repair, and bone pain. Active synthesis of this research recently led to the first biotherapeutic success story in the field. Specifically, the ongoing clinical trials of anti-nerve growth factor therapeutics have been optimized to titrated doses that effectively alleviate pain while maintaining bone and joint health. Continued collaborations between neuroscientists and bone biologists are needed to build on this progress, leading to a more complete understanding of neural regulation of the skeleton and development of novel therapeutics. © 2019 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jennifer M Brazill
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Washington University, St. Louis, MO, USA
| | - Alec T Beeve
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Washington University, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Clarissa S Craft
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Washington University, St. Louis, MO, USA.,Department of Cell Biology and Physiology, Washington University, St. Louis, MO, USA
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Erica L Scheller
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Washington University, St. Louis, MO, USA.,Department of Cell Biology and Physiology, Washington University, St. Louis, MO, USA
| |
Collapse
|
49
|
A population of nonneuronal GFRα3-expressing cells in the bone marrow resembles nonmyelinating Schwann cells. Cell Tissue Res 2019; 378:441-456. [DOI: 10.1007/s00441-019-03068-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 07/01/2019] [Indexed: 12/17/2022]
|
50
|
Morgan M, Nencini S, Thai J, Ivanusic JJ. TRPV1 activation alters the function of Aδ and C fiber sensory neurons that innervate bone. Bone 2019; 123:168-175. [PMID: 30936039 DOI: 10.1016/j.bone.2019.03.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/19/2019] [Accepted: 03/28/2019] [Indexed: 12/31/2022]
Abstract
The Transient receptor potential cation channel subfamily V member 1 (TRPV1) is a non-selective cation channel that is activated by capsaicin, low pH and noxious heat. It has been suggested to have a pro-algesic role in a range of conditions that present with bone pain, but the mechanisms by which this occurs are not yet clear. In this study we aimed to determine if TRPV1 is expressed in Aδ and/or C fiber bone afferent neurons, and to explore its role in the activation and/or sensitization of bone afferent neurons to mechanical stimulation. A combination of retrograde tracing and immunohistochemistry was used to determine expression of TRPV1 in the soma of bone afferent neurons that innervate the rat tibial marrow cavity. A novel, in vivo, electrophysiological bone-nerve preparation, recently developed in our laboratory, was used to make recordings of the activity and sensitivity of bone afferent neurons in response to application of the TRPV1 agonist capsaicin to the marrow cavity. We found that a substantial proportion of bone afferent neurons express TRPV1. These include both small-diameter myelinated (neurofilament rich) and unmyelinated (neurofilament poor) neurons that are likely to be Aδ and C fiber neurons, respectively. Electrophysiological recordings revealed that application of capsaicin to the marrow cavity increased ongoing activity of C fiber, and to a lesser extent Aδ fiber, bone afferent neurons. Capsaicin also sensitized both Aδ and C fiber bone afferent neurons to mechanical stimulation. This evidence supports a role for TRPV1 in the pathogenesis of pain associated with bone pathology or disease.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Sara Nencini
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia.
| |
Collapse
|