101
|
Delay L, Gonçalves Dos Santos G, Dias EV, Yaksh TL, Corr M. Sexual Dimorphism in the Expression of Pain Phenotype in Preclinical Models of Rheumatoid Arthritis. Rheum Dis Clin North Am 2021; 47:245-264. [PMID: 33781493 DOI: 10.1016/j.rdc.2020.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis is one of most frequent rheumatic diseases, affecting around 1% of the population worldwide. Pain impacting the quality of life for the patient with rheumatoid arthritis, is often the primary factor leading them to seek medical care. Although sex-related differences in humans and animal models of rheumatoid arthritis are described, the correlation between pain and sex in rheumatoid arthritis has only recently been directly examined. Here we review the literature and explore the mechanisms underlying the expression of the pain phenotype in females and males in preclinical models of rheumatoid arthritis.
Collapse
Affiliation(s)
- Lauriane Delay
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.
| | | | - Elayne Vieira Dias
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Tony L Yaksh
- Department of Anesthesiology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| |
Collapse
|
102
|
Forouzanfar F, Shojapour M, Aghili ZS, Asgharzade S. Growth Factors as Tools in Photoreceptor Cell Regeneration and Vision Recovery. Curr Drug Targets 2021; 21:573-581. [PMID: 31755378 DOI: 10.2174/1389450120666191121103831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/04/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023]
Abstract
Photoreceptor loss is a major cause of blindness around the world. Stem cell therapy offers a new strategy in retina degenerative disease. Retinal progenitors can be derived from embryonic stem cells (ESC) in vitro, but cannot be processed to a mature state. In addition, the adult recipient retina presents a very different environment than the photoreceptor precursor donor. It seems that modulation of the recipient environment by ectopic development regulated growth factors for transplanted cells could generate efficient putative photoreceptors. The purpose of this review article was to investigate the signaling pathway of growth factors including: insulin-like growth factors (IGFs), fibroblast growth factors (FGF), Nerve growth factor (NGF), Brain-derived neurotrophic factor (BDNF), Taurin and Retinoic acid (RA) involved in the differentiation of neuroretina cell, like; photoreceptor and retinal progenitor cells. Given the results available in the related literature, the differentiation efficacy of ESCs toward the photoreceptor and retinal neurons and the important role of growth factors in activating signaling pathways such as Akt, Ras/Raf1/ and ERKs also inhibit the ASK1/JNK apoptosis pathway. Manipulating differentiated culture, growth factors can influence photoreceptor transplantation efficiency in retinal degenerative disease.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mana Shojapour
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Zahra Sadat Aghili
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
103
|
Ma Z, Huang Z, Zhang L, Li X, Xu B, Xiao Y, Shi X, Zhang H, Liao T, Wang P. Vanillic Acid Reduces Pain-Related Behavior in Knee Osteoarthritis Rats Through the Inhibition of NLRP3 Inflammasome-Related Synovitis. Front Pharmacol 2021; 11:599022. [PMID: 33658936 PMCID: PMC7917290 DOI: 10.3389/fphar.2020.599022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives: Synovitis plays an important role in knee osteoarthritis (KOA) pain. The activation of the NOD-like receptor protein 3 (NLRP3) inflammasome in fibroblast-like synoviocytes (FLSs) promotes KOA development. In this study, we aimed to investigate whether vanillic acid (VA), a monomer derived from Chinese herbal medicines, could target NLRP3 inflammasome-related synovitis to reduce pain. Methods: Rats in the KOA and KOA + VA groups were injected with monosodium iodoacetate (MIA) in the knee to induce KOA. From day 14, the KOA + VA group was given VA at 30 mg/kg every day via gastric intubation. FLSs were collected from the synovial tissues. We examined both the protein and gene expression of caspase-1, apoptosis-associated speck-like protein with a caspase recruitment domain (ASC), NLRP3, components of the NLRP3 inflammasome, and interleukin-1β (IL-1β) and IL-18 in vivo and in vitro. Results: The upregulation of caspase-1, ASC, and NLRP3 in the KOA model were reduced by VA. VA also lowered the level of IL-1β and IL-18 in the KOA model. In addition, VA relieved pain-related behavior of KOA model rats and downregulated the pain mediators CGRP, NGF, and TrkA in FLSs. Interestingly, we also observed reduced synovial fibrosis in the animal experiments. Conclusion: Our research showed that VA reduces synovitis and pain-related behaviors in a rat model of KOA, which provides the basis for further investigations into the potential therapeutic impact of VA in KOA.
Collapse
Affiliation(s)
- Zhenyuan Ma
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengquan Huang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Li Zhang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaochen Li
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Xu
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yancheng Xiao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoqing Shi
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Haosheng Zhang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Taiyang Liao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
104
|
Werland F, Hirth M, Rukwied R, Ringkamp M, Turnquist B, Jorum E, Namer B, Schmelz M, Obreja O. Maximum axonal following frequency separates classes of cutaneous unmyelinated nociceptors in the pig. J Physiol 2021; 599:1595-1610. [DOI: 10.1113/jp280269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/17/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Fiona Werland
- Department of Experimental Pain Research, MCTN Medical Faculty Mannheim Heidelberg University Mannheim Germany
| | - Michael Hirth
- Department of Experimental Pain Research, MCTN Medical Faculty Mannheim Heidelberg University Mannheim Germany
| | - Roman Rukwied
- Department of Experimental Pain Research, MCTN Medical Faculty Mannheim Heidelberg University Mannheim Germany
| | - Matthias Ringkamp
- Department of Neurosurgery Johns Hopkins University Baltimore MD USA
| | - Brian Turnquist
- Faculty of Mathematics and Computer Science Bethel University MN USA
| | - Ellen Jorum
- Section of Clinical Neurophysiology, Department of Neurology Oslo University Hospital Oslo Norway
- Institute of Clinical Medicine University of Oslo Oslo Norway
| | - Barbara Namer
- IZKF Neuroscience Research Group, University Hospital RWTH Aachen and Department of Physiology and Pathophysiology University of Erlangen‐Nuremberg Erlangen Germany
| | - Martin Schmelz
- Department of Experimental Pain Research, MCTN Medical Faculty Mannheim Heidelberg University Mannheim Germany
| | - Otilia Obreja
- Department of Experimental Pain Research, MCTN Medical Faculty Mannheim Heidelberg University Mannheim Germany
| |
Collapse
|
105
|
Ye J, Huang B, Gong P. Nerve growth factor-chondroitin sulfate/hydroxyapatite-coating composite implant induces early osseointegration and nerve regeneration of peri-implant tissues in Beagle dogs. J Orthop Surg Res 2021; 16:51. [PMID: 33436038 PMCID: PMC7805124 DOI: 10.1186/s13018-020-02177-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/25/2020] [Indexed: 02/08/2023] Open
Abstract
Background Osseointegration is the premise of the chewing function of dental implant. Nerve growth factor (NGF), as a neurotrophic factor, can induce bone healing. However, the influence of NGF-chondroitin sulfate (CS)/hydroxyapatite (HA)-coating composite implant on the osseointegration and innervations is still not entirely clear. Materials and methods NGF-CS/HA-coating composite implants were prepared using the modified biomimetic method. The characteristics of NGF-CS/HA-coating implants were determined using a scanning electron microscope. After NGF-CS/HA-coating implants were placed in the mandible of Beagle dogs, the early osseointegration and innervation in peri-implant tissues were assessed through X-ray, Micro-CT, maximal pull-out force, double fluorescence staining, toluidine blue staining, DiI neural tracer, immunohistochemistry, and RT-qPCR assays. Results NGF-CS/HA-coating composite implants were made successfully, which presented porous mesh structures with the main components (Ti and HA). Besides, we revealed that implantation of NGF-CS/HA-coating implants significantly changed the morphology of bone tissues and elevated maximum output, MAR, BIC, and nerve fiber in the mandible of Beagle dogs. Moreover, we proved that the implantation of NGF-CS/HA-coating implants also markedly upregulated the levels of NGF, osteogenesis differentiation, and neurogenic differentiation-related genes in the mandible of Beagle dogs. Conclusion Implantation of NGF-CS/HA-coating composite implants has significant induction effects on the early osseointegration and nerve regeneration of peri-implant tissues in the mandible of Beagle dogs. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-020-02177-5.
Collapse
Affiliation(s)
- Jun Ye
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University and Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, People's Republic of China
| | - Bo Huang
- State Key Laboratory of Oral Diseases, General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, Department of Oral Implant, West China School of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
106
|
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
|
107
|
Qin L, Li J. Nerve growth factor in muscle afferent neurons of peripheral artery disease and autonomic function. Neural Regen Res 2021; 16:694-699. [PMID: 33063730 PMCID: PMC8067946 DOI: 10.4103/1673-5374.293132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In peripheral artery disease patients, the blood supply directed to the lower limbs is reduced. This results in severe limb ischemia and thereby enhances pain sensitivity in lower limbs. The painful perception is induced and exaggerate during walking, and is relieved by rest. This symptom is termed by intermittent claudication. The limb ischemia also amplifies autonomic responses during exercise. In the process of pain and autonomic responses originating exercising muscle, a number of receptors in afferent nerves sense ischemic changes and send signals to the central nervous system leading to autonomic responses. This review integrates recent study results in terms of perspectives including how nerve growth factor affects muscle sensory nerve receptors in peripheral artery disease and thereby alters responses of sympathetic nerve activity and blood pressure to active muscle. For the sensory nerve receptors, we emphasize the role played by transient receptor potential vanilloid type 1, purinergic P2X purinoceptor 3 and acid sensing ion channel subtype 3 in amplified sympathetic nerve activity responses in peripheral artery disease.
Collapse
Affiliation(s)
- Lu Qin
- Heart & Vascular Institute, Penn State University College of Medicine, Hershey, PA, USA
| | - Jianhua Li
- Heart & Vascular Institute, Penn State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
108
|
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
|
109
|
Kendall A, Nyström S, Ekman S, Hultén LM, Lindahl A, Hansson E, Skiöldebrand E. Nerve growth factor in the equine joint. Vet J 2020; 267:105579. [PMID: 33375964 DOI: 10.1016/j.tvjl.2020.105579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/09/2020] [Accepted: 11/16/2020] [Indexed: 10/24/2022]
Abstract
Nerve growth factor (NGF) is a neurotrophin with many functions. In humans, it is involved in inflammation, nerve growth, apoptosis and pain signalling. Increased concentrations of NGF in synovial fluid has been shown in humans and dogs with osteoarthritis. Despite osteoarthritis being a common problem in horses, no studies have previously been published on NGF in the equine joint. The aim of this study was to quantify NGF in equine synovial fluid from healthy joints, acutely inflamed septic joints and joints with structural changes associated with osteoarthritis. A secondary aim was to identify the localisation of NGF and its two receptors, TrkA and p75NTR, in healthy and osteoarthritic articular cartilage. NGF concentrations in synovial fluid from osteoarthritic joints (n = 27), septic joints (n = 9) and healthy joints (n = 16) were determined by ELISA. In addition, articular cartilage from osteoarthritic and healthy joints was examined for NGF, TrkA and p75NTR using immunohistochemistry staining. NGF was present in equine synovial fluid and articular cartilage. Compared to synovial fluid from healthy joints, NGF concentration was higher in synovial fluid from joints with structural osteoarthritic changes (P = 0.032) or acute septic inflammation (P = 0.006). In articular cartilage with severe osteoarthritic changes, there was more abundant positive immunohistochemistry staining for NGF and its receptors than in normal articular cartilage. Further studies should focus on identifying precursor forms of NGF, and on receptor expression and downstream signalling of TrkA and P75NTR in health and disease.
Collapse
Affiliation(s)
- A Kendall
- Division of Pathology, Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, 750 07 Uppsala, Sweden.
| | - S Nyström
- Department of Clinical Chemistry and Transfusion Medicine, Sahlgrenska University Hospital, Bruna Stråket 16, 413 45 Gothenburg, Sweden
| | - S Ekman
- Division of Pathology, Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, 750 07 Uppsala, Sweden
| | - L M Hultén
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Wallenberg Laboratory, Bruna Stråket 16, 413 45 Gothenburg, Sweden
| | - A Lindahl
- Department of Clinical Chemistry and Transfusion Medicine, Sahlgrenska University Hospital, Bruna Stråket 16, 413 45 Gothenburg, Sweden
| | - E Hansson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 7, 413 45 Gothenburg, Sweden
| | - E Skiöldebrand
- Division of Pathology, Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Box 7028, 750 07 Uppsala, Sweden
| |
Collapse
|
110
|
Mini-review - Sodium channels and beyond in peripheral nerve disease: Modulation by cytokines and their effector protein kinases. Neurosci Lett 2020; 741:135446. [PMID: 33166641 DOI: 10.1016/j.neulet.2020.135446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022]
Abstract
Peripheral neuropathy is associated with enhanced activity of primary afferents which is often manifested as pain. Voltage-gated sodium channels (VGSCs) are critical for the initiation and propagation of action potentials and are thus essential for the transmission of the noxious stimuli from the periphery. Human peripheral sensory neurons express multiple VGSCs, including Nav1.7, Nav1.8, and Nav1.9 that are almost exclusively expressed in the peripheral nervous system. Distinct biophysical properties of Nav1.7, Nav1.8, and Nav1.9 underlie their differential contributions to finely tuned neuronal firing of nociceptors, and mutations in these channels have been associated with several inherited human pain disorders. Functional characterization of these mutations has provided additional insights into the role of these channels in electrogenesis in nociceptive neurons and pain sensation. Peripheral tissue damage activates an inflammatory response and triggers generation and release of inflammatory mediators, which can act through diverse signaling cascades to modulate expression and activity of ion channels including VGSCs, contributing to the development and maintenance of pathological pain conditions. In this review, we discuss signaling pathways that are activated by pro-nociceptive inflammatory mediators that regulate peripheral sodium channels, with a specific focus on direct phosphorylation of these channels by multiple protein kinases.
Collapse
|
111
|
Abstract
Primary nociceptors are a heterogeneous class of peripheral somatosensory neurons, responsible for detecting noxious, pruriceptive, and thermal stimuli. These neurons are further divided into several molecularly defined subtypes that correlate with their functional sensory modalities and morphological features. During development, all nociceptors arise from a common pool of embryonic precursors, and then segregate progressively into their mature specialized phenotypes. In this review, we summarize the intrinsic transcriptional programs and extrinsic trophic factor signaling mechanisms that interact to control nociceptor diversification. We also discuss how recent transcriptome profiling studies have significantly advanced the field of sensory neuron development.
Collapse
Affiliation(s)
- Suna L Cranfill
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
112
|
Vincent TL. Of mice and men: converging on a common molecular understanding of osteoarthritis. THE LANCET. RHEUMATOLOGY 2020; 2:e633-e645. [PMID: 32989436 PMCID: PMC7511206 DOI: 10.1016/s2665-9913(20)30279-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite an increasing burden of osteoarthritis in developed societies, target discovery has been slow and there are currently no approved disease-modifying osteoarthritis drugs. This lack of progress is due in part to a series of misconceptions over the years: that osteoarthritis is an inevitable consequence of ageing, that damaged articular cartilage cannot heal itself, and that osteoarthritis is driven by synovial inflammation similar to that seen in rheumatoid arthritis. Molecular interrogation of disease through ex-vivo tissue analysis, in-vitro studies, and preclinical models have radically reshaped the knowledge landscape. Inflammation in osteoarthritis appears to be distinct from that seen in rheumatoid arthritis. Recent randomised controlled trials, using treatments repurposed from rheumatoid arthritis, have largely been unsuccessful. Genome-wide studies point to defects in repair pathways, which accords well with recent promise using growth factor therapies or Wnt pathway antagonism. Nerve growth factor has emerged as a robust target in osteoarthritis pain in phase 2-3 trials. These studies, both positive and negative, align well with those in preclinical surgical models of osteoarthritis, indicating that pathogenic mechanisms identified in mice can lead researchers to valid human targets. Several novel candidate pathways are emerging from preclinical studies that offer hope of future translational impact. Enhancing trust between industry, basic, and clinical scientists will optimise our collective chance of success.
Collapse
Affiliation(s)
- Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis, Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
113
|
Squillace S, Spiegel S, Salvemini D. Targeting the Sphingosine-1-Phosphate Axis for Developing Non-narcotic Pain Therapeutics. Trends Pharmacol Sci 2020; 41:851-867. [PMID: 33010954 DOI: 10.1016/j.tips.2020.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023]
Abstract
Chronic pain is a life-altering condition affecting millions of people. Current treatments are inadequate and prolonged therapies come with severe side effects, especially dependence and addiction to opiates. Identification of non-narcotic analgesics is of paramount importance. Preclinical and clinical studies suggest that sphingolipid metabolism alterations contribute to neuropathic pain development. Functional sphingosine-1-phosphate (S1P) receptor 1 (S1PR1) antagonists, such as FTY720/fingolimod, used clinically for non-pain conditions, are emerging as non-narcotic analgesics, supporting the repurposing of fingolimod for chronic pain treatment and energizing drug discovery focused on S1P signaling. Here, we summarize the role of S1P in pain to highlight the potential of targeting the S1P axis towards development of non-narcotic therapeutics, which, in turn, will hopefully help lessen misuse of opioid pain medications and address the ongoing opioid epidemic.
Collapse
Affiliation(s)
- Silvia Squillace
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| |
Collapse
|
114
|
The opposing contribution of neurotrophin-3 and nerve growth factor to orofacial heat hyperalgesia in rats. Behav Pharmacol 2020; 31:27-33. [PMID: 31577558 DOI: 10.1097/fbp.0000000000000503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
It has been proposed that neurotrophin-3 acts in a manner that is opposed to nerve growth factor, especially in the modulation of heat hyperalgesia. Injury to the constriction of the infraorbital nerve (CION) is a well-established model of trigeminal neuropathic pain that leads to robust heat, cold, and mechanical hyperalgesia. Here, we assessed the effect of local neurotrophin-3 treatment on CION-induced hyperalgesia, and we examined some mechanisms related to the effect of neurotrophin-3. Neurotrophin-3 (1 µg/50 µl) injected into the upper lip of CION rats caused a significant and long-lasting reduction of CION-induced heat hyperalgesia, but failed to affect cold and mechanical hyperalgesia. Increased levels of neurotrophin-3 were detected in the injured nerve at the time point that represents the peak of heat hyperalgesia. The anti-hyperalgesic effect of neurotrophin-3 was markedly reduced in the presence of an antagonist of TrkA receptors (K-252a, 1 μg/50 μl). Moreover, association of lower doses of neurotrophin-3 with an antibody anti-nerve growth factor resulted in a synergistic anti-hyperalgesic effect in CION rats. Local injection of nerve growth factor (3 µg/50 µl) or the TRPV1 agonist capsaicin (1 μg/50 μl), but not neurotrophin-3 injection (1 µg/50 µl), resulted in long-lasting facial heat hyperalgesia, which was both significantly reduced by previous neurotrophin-3 local treatment. In conclusion, we suggest that neurotrophin-3 is a potent modulator of facial heat hyperalgesia, which may exert an inhibitory influence on the trkA pathway. Neurotrophin-3 treatment may represent a promising approach, especially in pain conditions associated with increased levels of nerve growth factor.
Collapse
|
115
|
Local anaesthesia decreases nerve growth factor induced masseter hyperalgesia. Sci Rep 2020; 10:15458. [PMID: 32963266 PMCID: PMC7508842 DOI: 10.1038/s41598-020-71620-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/13/2020] [Indexed: 11/08/2022] Open
Abstract
The aim of this investigation was to evaluate the effects of local anaesthesia on nerve growth factor (NGF) induced masseter hyperalgesia. Healthy participants randomly received an injection into the right masseter muscle of either isotonic saline (IS) given as a single injection (n = 15) or an injection of NGF (n = 30) followed by a second injection of lidocaine (NGF + lidocaine; n = 15) or IS (NGF + IS; n = 15) in the same muscle 48 h later. Mechanical sensitivity scores of the right and left masseter, referred sensations and jaw pain intensity and jaw function were assessed at baseline, 48 h after the first injection, 5 min after the second injection and 72 h after the first injection. NGF caused significant jaw pain evoked by chewing at 48 and 72 h after the first injection when compared to the IS group, but without significant differences between the NGF + lidocaine and NGF + IS groups. However, the mechanical sensitivity of the right masseter 5 min after the second injection in the NGF + lidocaine group was significantly lower than the second injection in the NGF + IS and was similar to the IS group. There were no significant differences for the referred sensations. Local anaesthetics may provide relevant information regarding the contribution of peripheral mechanisms in the maintenance of persistent musculoskeletal pain.
Collapse
|
116
|
Nerve growth factor antibody for the treatment of osteoarthritis pain and chronic low-back pain: mechanism of action in the context of efficacy and safety. Pain 2020; 160:2210-2220. [PMID: 31145219 PMCID: PMC6756297 DOI: 10.1097/j.pain.0000000000001625] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chronic pain continues to be a significant global burden despite the availability of a variety of nonpharmacologic and pharmacologic treatment options. Thus, there is a need for new analgesics with novel mechanisms of action. In this regard, antibodies directed against nerve growth factor (NGF-Abs) are a new class of agents in development for the treatment of chronic pain conditions such as osteoarthritis and chronic low-back pain. This comprehensive narrative review summarizes evidence supporting pronociceptive functions for NGF that include contributing to peripheral and central sensitization through tropomyosin receptor kinase A activation and stimulation of local neuronal sprouting. The potential role of NGF in osteoarthritis and chronic low-back pain signaling is also examined to provide a mechanistic basis for the observed efficacy of NGF-Abs in clinical trials of these particular pain states. Finally, the safety profile of NGF-Abs in terms of common adverse events, joint safety, and nerve structure/function is discussed.
Collapse
|
117
|
Mai L, Huang F, Zhu X, He H, Fan W. Role of Nerve Growth Factor in Orofacial Pain. J Pain Res 2020; 13:1875-1882. [PMID: 32801845 PMCID: PMC7399448 DOI: 10.2147/jpr.s250030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Some chronic pain conditions in the orofacial region are common and the mechanisms underlying orofacial pain are unresolved. Nerve growth factor (NGF) is a member of a family of neurotrophins and regulates the growth, maintenance and development of neurons. Increasing evidence suggests that NGF plays a crucial role in the generation of pain and hyperalgesia in different pain states. This review investigates the role of NGF in orofacial pain and their underlying cellular mechanisms, which may provide essential guidance to drug-discovery programmes. A systemic literature search was conducted in Pubmed focusing on NGF and orofacial pain. Articles were reviewed, and those discussing in vitro studies, animal evidence, clinical course, and possible mechanisms were summarized. We found a hyperalgesic effect of NGF in peripheral sensitization in orofacial pain models. We also summarize the current knowledge regarding NGF-dependent pain mechanism, which is initiated by retrograde transport of the ligand-receptor complex, ensuing transcriptional regulation of many important nociceptor genes involved in nociceptive processing. Phase III trials suggest that anti-NGF drug is endorsed with anti-inflammatory and pain-relieving effects with good tolerance in a variety of pain conditions, including pain associated with osteoarthritis and chronic lower back pain. Based on the data reviewed herein, NGF is believed to be an important hyperalgesic mediator in orofacial pain. The identification of underlying mechanisms and pathways of orofacial pain opens new frontiers for pain management.
Collapse
Affiliation(s)
- Lijia Mai
- Department of Anesthesiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, People's Republic of China
| | - Fang Huang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, People's Republic of China
| | - Xiao Zhu
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, People's Republic of China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, People's Republic of China
| | - Wenguo Fan
- Department of Anesthesiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510080, People's Republic of China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, People's Republic of China
| |
Collapse
|
118
|
Smaila BD, Holland SD, Babaeijandaghi F, Henderson HG, Rossi FMV, Ramer MS. Systemic hypoxia mimicry enhances axonal regeneration and functional recovery following peripheral nerve injury. Exp Neurol 2020; 334:113436. [PMID: 32814068 DOI: 10.1016/j.expneurol.2020.113436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/28/2020] [Accepted: 08/13/2020] [Indexed: 01/15/2023]
Abstract
Despite the ability of peripheral nerves to regenerate after injury, failure occurs due to an inability of supporting cells to maintain growth, resulting in long-term consequences such as sensorimotor dysfunction and neuropathic pain. Here, we investigate the potential of engaging the cellular adaptive response to hypoxia, via inhibiting its negative regulators, to enhance the regenerative process. Under normoxic conditions, prolyl hydroxylase domain (PHD) proteins 1, 2, and 3 hydroxylate the key metabolic regulator hypoxia inducible factor 1α (HIF1α), marking it for subsequent proteasomal degradation. We inhibited PHD protein function systemically via either individual genetic deletion or pharmacological pan-PHD inhibition using dimethyloxalylglycine (DMOG). We show enhanced axonal regeneration after sciatic nerve crush injury in PHD1-/- mice, PHD3-/- mice, and in DMOG-treated mice, and in PHD1-/- and DMOG-treated mice a reduction in hypersensitivity to cooling after permanent sciatic ligation. Electromyographically, PHD1-/- and PHD3-/- mice showed an increased CMAP amplitude one-month post-injury, probably due to protection against denervation induced muscle atrophy, while DMOG-treated and PHD2+/- mice showed reduced latencies, indicating improved motor axon function. DMOG treatment did not affect the growth of dorsal root ganglion neurites in vitro, suggesting a lack of direct effects of DMOG on axonal regrowth. Enhanced regeneration in vivo was concurrent with an increase in macrophage density, and a shift in macrophage polarization state ratios (from M1-like toward M2-like) in DMOG-treated animals. These results indicate PHD proteins as a novel therapeutic target to improve regenerative and functional outcomes after peripheral nerve injury without manipulating molecular O2.
Collapse
Affiliation(s)
- Brittney D Smaila
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada
| | - Seth D Holland
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada
| | - Farshad Babaeijandaghi
- The Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada
| | - Holly G Henderson
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada
| | - Fabio M V Rossi
- The Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada
| | - Matt S Ramer
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada.
| |
Collapse
|
119
|
Therapeutic Manipulation of Macrophages Using Nanotechnological Approaches for the Treatment of Osteoarthritis. NANOMATERIALS 2020; 10:nano10081562. [PMID: 32784839 PMCID: PMC7466380 DOI: 10.3390/nano10081562] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is the most common joint pathology causing severe pain and disability. Macrophages play a central role in the pathogenesis of OA. In the joint microenvironment, macrophages with an M1-like pro-inflammatory phenotype induce chronic inflammation and joint destruction, and they have been correlated with the development and progression of the disease, while the M2-like anti-inflammatory macrophages support the recovery of the disease, promoting tissue repair and the resolution of inflammation. Nowadays, the treatment of OA in the clinic relies on systemic and/or intra-articular administration of anti-inflammatory and pain relief drugs, as well as surgical interventions for the severe cases (i.e., meniscectomy). The disadvantages of the pharmacological therapy are related to the chronic nature of the disease, requiring prolonged treatments, and to the particular location of the pathology in joint tissues, which are separated anatomical compartments with difficult access for the drugs. To overcome these challenges, nanotechnological approaches have been investigated to improve the delivery of drugs toward macrophages into the diseased joint. This strategy may offer advantages by reducing off-target toxicities and improving long-term therapeutic efficacy. In this review, we describe the nanomaterial-based approaches designed so far to directly or indirectly manipulate macrophages for the treatment of osteoarthritis.
Collapse
|
120
|
Kościelniak-Merak B, Batko I, Kobylarz K, Sztefko K, Tomasik PJ. Intravenous, Perioperatively Administered Lidocaine Regulates Serum Pain Modulators’ Concentrations in Children Undergoing Spinal Surgery. PAIN MEDICINE 2020; 21:1464-1473. [DOI: 10.1093/pm/pnz212] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
AbstractObjectivesWe analyzed the influence of perioperative, intravenous (i.v.) lidocaine infusion as a part of multimodal anesthesia on concentrations of selected pain modulators.DesignAn observational study.SettingUniversity Children’s Hospital in Cracow, Poland, from May 2015 to May 2018.SubjectsForty-four children undergoing extensive spinal surgery, divided into two groups after surgery: the study group (N = 23), anesthetized generally with lidocaine as a co-analgesic, and the control group (N = 22), anesthetized generally without lidocaine.MethodsWe assessed proinflammatory mediators like neuron growth factor (NGF), high mobility group box 1 (HMGB1), interleukin 6 (IL-6), and FOS protein before, immediately after, six hours and 12–15 hours after surgery. We evaluated pain intensity at corresponding time points using a 10-point numerical/graphical scale.ResultsWe observed that children in the lidocaine group had reduced pain intensity in the resting state and during movement until six hours after surgery when compared with controls. We found lower NGF concentrations in the lidocaine group vs controls only at six hours after surgery. Mean HMGB1 concentrations during the postoperative period in the study group were relatively stable, whereas we observed significant increases at six hours after surgery and a slight decrease at 12–15 hours after surgery in the control group. IL-6 concentrations at six hours were lower in lidocaine patients when compared with controls. We noted a negative correlation between HMGB1, NGF, Il-6, and lidocaine concentrations after surgery. We did not find any differences in FOS protein concentrations between the groups.ConclusionsOur findings suggest that intraoperative and postoperative i.v. lidocaine administration as a part of multimodal anesthesia may reduce inflammatory-dependent postoperative pain intensity.
Collapse
Affiliation(s)
- Barbara Kościelniak-Merak
- Department of Clinical Biochemistry, Pediatrics Institute, Jagiellonian University Medical College, Cracow, Poland
| | - Ilona Batko
- Intensive Care Unit, University Children’s Hospital, Cracow, Poland
| | - Krzysztof Kobylarz
- Intensive Care Unit, University Children’s Hospital, Cracow, Poland
- Department of Anesthesiology and Intensive Care, Jagiellonian University Medical College, Cracow, Poland
| | - Krystyna Sztefko
- Department of Clinical Biochemistry, Pediatrics Institute, Jagiellonian University Medical College, Cracow, Poland
| | - Przemysław J Tomasik
- Department of Clinical Biochemistry, Pediatrics Institute, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
121
|
López-Leal R, Díaz-Viraqué F, Catalán RJ, Saquel C, Enright A, Iraola G, Court FA. Schwann cell reprogramming into repair cells increases miRNA-21 expression in exosomes promoting axonal growth. J Cell Sci 2020; 133:jcs.239004. [PMID: 32409566 DOI: 10.1242/jcs.239004] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/24/2020] [Indexed: 12/23/2022] Open
Abstract
Functional recovery after peripheral nerve damage is dependent on the reprogramming of differentiated Schwann cells (dSCs) into repair Schwann cells (rSCs), which promotes axonal regeneration and tissue homeostasis. Transition into a repair phenotype requires expression of c-Jun and Sox2, which transcriptionally mediates inhibition of the dSC program of myelination and activates a non-cell-autonomous repair program, characterized by the secretion of neuronal survival and regenerative molecules, formation of a cellular scaffold to guide regenerating axons and activation of an innate immune response. Moreover, rSCs release exosomes that are internalized by peripheral neurons, promoting axonal regeneration. Here, we demonstrate that reprogramming of Schwann cells (SCs) is accompanied by a shift in the capacity of their secreted exosomes to promote neurite growth, which is dependent on the expression of c-Jun (also known as Jun) and Sox2 by rSCs. Furthermore, increased expression of miRNA-21 is responsible for the pro-regenerative capacity of rSC exosomes, which is associated with PTEN downregulation and PI3-kinase activation in neurons. We propose that modification of exosomal cargo constitutes another important feature of the repair program of SCs, contributing to axonal regeneration and functional recovery after nerve injury.
Collapse
Affiliation(s)
- Rodrigo López-Leal
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago 7800003, Chile
| | - Florencia Díaz-Viraqué
- Laboratorio de Interacciones Hospedero Patógeno - Unidad de Biología Molecular, Institut de Pasteur Montevideo, Mataojo 2020, Montevideo 11400, Uruguay
| | - Romina J Catalán
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago 7800003, Chile
| | - Cristian Saquel
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago 7800003, Chile
| | - Anton Enright
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Gregorio Iraola
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile.,Microbial Genomics Laboratory, Institut Pasteur de Montevideo, Mataojo 2020, Montevideo 11400, Uruguay
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile .,Fondap Geroscience Center for Brain Health and Metabolism, Santiago 7800003, Chile.,Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
122
|
Albo C, Kumar S, Pope M, Kidwell KM, Xu H, Bowman L, Wells L, Barrett N, Fields S, Bora P, Patel N, Kutlar A. Characteristics and potential biomarkers of adult sickle cell patients with chronic pain. Eur J Haematol 2020; 105:419-425. [PMID: 32497305 DOI: 10.1111/ejh.13461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES In this study, we investigated the evolution of chronic pain in sickle cell patients (SCD) as an age-dependent phenomenon and studied the frequency of vaso-occlusive episode frequency, opioid use, quantitative sensory testing (QST), and biomarkers of chronic pain (CP). METHODS We undertook a cross-sectional study of the evolution of CP in SCD. A total of 72 subjects (age 15-66) were enrolled. VOE frequency, presence of CP hydroxyurea (HU) therapy, opioid use, and laboratory parameters were collected. QST was performed, and plasma tryptase, substance P, and NGF (Nerve Growth Factor) levels were assayed. RESULTS There was an age-dependent increase in frequency of CP, VOEs, opioid use, and Von Frey monofilament values. CP patients had significantly higher opioid use (daily morphine equivalents) (52.8 mg vs 6.94 mg, P = .009), suggesting a correlation between opioid use and hyperalgesia. NGF levels were also significantly higher (P = .051). Our results confirm previous observations of an age-dependent increase in the proportion of patients with CP and support the contributing role of mast cell activation and neurogenic inflammation. CONCLUSIONS This is the first study of NGF as a possible biomarker of CP in SCD. If confirmed, this could provide a diagnostic marker and therapeutic target for CP in SCD.
Collapse
Affiliation(s)
- Camila Albo
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Sanjiv Kumar
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Michael Pope
- Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | - Hongyan Xu
- Department of Population Health Sciences, Augusta University, Augusta, GA, USA
| | - Latanya Bowman
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Leigh Wells
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Nadine Barrett
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Sabine Fields
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Pritam Bora
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Niren Patel
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Abdullah Kutlar
- Center for Blood Disorders Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
123
|
Patel F, Hess DK, Maher DP. Anti-nerve growth factor antibodies for the treatment of low back pain. Expert Rev Clin Pharmacol 2020; 13:631-639. [PMID: 32436473 DOI: 10.1080/17512433.2020.1772052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION The treatment of chronic low back pain (cLBP) often involves multimodal pharmacologic and non-pharmacologic strategies. There remain shortcomings with these tools with regards to both effect size and side effects. AREAS COVERED In an effort to better address cLBP, anti-nerve growth factor (NGF) monoclonal antibodies (mAbs) are nearing marketing approval. This class of medications has been primarily evaluated for osteoarthritis, but are being examined at higher doses for use in cLBP. We review the efficacy of this class in treating LBP as well as their potential side effects based on nine phase II or III published clinical trials. Five trials evaluated Tanezumab and four trials evaluated Fasinumab, with seven trials evaluating nonspecific LBP, one evaluating sciatica related cLBP, and one evaluating vertebral fracture related cLBP. EXPERT OPINION The results of available clinical trials indicate modest effectiveness with regard to reduction of pain in the low back, and improved functionality, compared to placebo in keeping with the effect size of other pharmacologic treatment modalities. Rapidly progressive osteoarthritis was infrequently reported. However, the continued observation of this serious side effects warrants careful patient selection and balancing the risks and benefits of anti-NGF mAbs in treating cLBP.
Collapse
Affiliation(s)
- Falin Patel
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine , Baltimore, MD, USA
| | - Demere K Hess
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine , Baltimore, MD, USA
| | - Dermot P Maher
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine , Baltimore, MD, USA
| |
Collapse
|
124
|
Drissi I, Woods WA, Woods CG. Understanding the genetic basis of congenital insensitivity to pain. Br Med Bull 2020; 133:65-78. [PMID: 32219415 PMCID: PMC7227775 DOI: 10.1093/bmb/ldaa003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION OR BACKGROUND Congenital insensitivity to pain (CIP) is caused by extremely rare Mendelian genetic disorders. CIP individuals demonstrate the unexpectedly severe consequences of painlessness. Although only a small number of causative conditions and genes are known, most have led to profound insights into human nociception. CIP gene discovery is catalyzing the manufacture of completely new classes of analgesics, and these are needed as alternatives to synthetic highly potent opioids. SOURCES OF DATA Pubmed.gov peer-reviewed journal articles and reviews. AREAS OF AGREEMENT The importance of nerve growth factor-tropomyosin receptor kinase A (NGF-TRKA) signalling for nociceptor genesis and subsequent pain sensing.New analgesics can be generated from knowledge of the NGF-TRKA nociceptor pathway.Increased susceptibility to Staphylococcus aureus infection is a consequence of deficient NGF-TRKA signalling.Mutations in the voltage-gated sodium channels SCN9A and SCN11A can cause congenital painlessness, and in contradistinction, other mutations can cause episodic neuropathic pain. SCN9A/Nav1.7 is an analgesic target. SCN11A/Nav1.9 is unlikely to be an analgesic target.There are further Mendelian causes of painlessness to be discovered. AREAS OF CONTROVERSY Which NGF-TRKA intracellular signalling pathways operate in nociceptor development and which in post-natal pain sensing?Why have no clinically effective Nav1.7 antagonist been generated? SCN9A-CIP causes analgesia, at least in part, through endogenous opioids.Why do all CIP phenotypes involve a complete loss of all types of nociception? AREAS TIMELY FOR DEVELOPING RESEARCH PRDM12 as an analgesic target.Discovery of the function and analgesic potential of new CIP genes.Can NGF-TRKA be used in the treatment of S. aureus?
Collapse
Affiliation(s)
- Ichrak Drissi
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| | - William Aidan Woods
- School of Medicine, David Weatherall building, University Road, Keele University, Staffordshire ST5 5BG, UK
| | - Christopher Geoffrey Woods
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| |
Collapse
|
125
|
An update on targets for treating osteoarthritis pain: NGF and TRPV1. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2020; 6:129-145. [PMID: 34178580 DOI: 10.1007/s40674-020-00146-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose of review a)Osteoarthritis (OA) is the most common form of arthritis, and pain is the primary symptom of the disease, yet analgesic options for treating OA pain remain limited. In this review, we aimed to give an update on the current clinical and preclinical studies targeting two pathways that are being investigated for treating OA pain: the nerve growth factor (NGF) pathway and the transient receptor potential vanilloid-1 (TRPV1) pathway. Recent findings b)Antibodies against NGF, small molecule inhibitors of TrkA, TRPV1 agonists, and TRPV1 antagonists are all in different stages of clinical and pre-clinical testing for the treatment of OA pain. NGF antibodies have shown efficacy in the primary endpoints tested compared to placebo, however, rapidly progressive OA has been consistently observed in a subset of patients and the cause remains unclear. TRPV1 agonists have also demonstrated reduced pain with no serious adverse events - the most common adverse events include a burning or warming sensation upon administration. Summary c)Targeting the NGF and TRPV1 pathways appear effective for reducing OA pain, but further work is needed to better understand which patients may benefit most from these treatments. The anti-NGF antibody tanezumab and the TRPV1 agonist CNTX-4975 have both received fast-track designation from the FDA for the treatment of OA pain.
Collapse
|
126
|
Oostinga D, Steverink JG, van Wijck AJM, Verlaan JJ. An understanding of bone pain: A narrative review. Bone 2020; 134:115272. [PMID: 32062002 DOI: 10.1016/j.bone.2020.115272] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022]
Abstract
Skeletal pathologies are often accompanied by bone pain, which has negative effects on the quality of life and functional status of patients. Bone pain can be caused by a wide variety of injuries and diseases including (poorly healed) fractures, bone cancer, osteoarthritis and also iatrogenic by skeletal interventions. Orthopedic interventions are considered to be the most painful surgical procedures overall. Two major groups of medication currently used to attenuate bone pain are NSAIDs and opioids. However, these systemic drugs frequently introduce adverse events, emphasizing the need for alternative therapies that are directed at the pathophysiological mechanisms underlying bone pain. The periosteum, cortical bone and bone marrow are mainly innervated by sensory A-delta fibers and C-fibers. These fibers are mostly present in the periosteum rendering this structure most sensitive to nociceptive stimuli. A-delta fibers and C-fibers can be activated upon mechanical distortion, acidic environment and increased intramedullary pressure. After activation, these fibers can be sensitized by inflammatory mediators, phosphorylation of acid-sensing ion channels and cytokine receptors, or by upregulation of transcription factors. This can result in a change of pain perception such that normally non-noxious stimuli are now perceived as noxious. Pathological conditions in the bone can produce neurotrophic factors that bind to receptors on A-delta fibers and C-fibers. These fibers then start to sprout and increase the innervation density of the bone, making it more sensitive to nociceptive stimuli. In addition, repetitive painful stimuli cause neurochemical and electrophysiological alterations in afferent sensory neurons in the spinal cord, which leads to central sensitization, and can contribute to chronic bone pain. Understanding the pathophysiological mechanisms underlying bone pain in different skeletal injuries and diseases is important for the development of alternative, targeted pain treatments. These pain mechanism-based alternatives have the potential to improve the quality of life of patients suffering from bone pain without introducing undesirable systemic effects.
Collapse
Affiliation(s)
- Douwe Oostinga
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Jasper G Steverink
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Albert J M van Wijck
- Department of Anesthesiology, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| | - Jorrit-Jan Verlaan
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3508 GA Utrecht, the Netherlands.
| |
Collapse
|
127
|
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
|
128
|
Abstract
The contribution of nerves to the pathogenesis of malignancies has emerged as an important component of the tumour microenvironment. Recent studies have shown that peripheral nerves (sympathetic, parasympathetic and sensory) interact with tumour and stromal cells to promote the initiation and progression of a variety of solid and haematological malignancies. Furthermore, new evidence suggests that cancers may reactivate nerve-dependent developmental and regenerative processes to promote their growth and survival. Here we review emerging concepts and discuss the therapeutic implications of manipulating nerves and neural signalling for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Ali H Zahalka
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, NY, USA.
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA.
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
129
|
Lin C, Ren Z, Yang X, Yang R, Chen Y, Liu Z, Dai Z, Zhang Y, He Y, Zhang C, Wang X, Cao W, Ji T. Nerve growth factor (NGF)-TrkA axis in head and neck squamous cell carcinoma triggers EMT and confers resistance to the EGFR inhibitor erlotinib. Cancer Lett 2020; 472:81-96. [PMID: 31838083 DOI: 10.1016/j.canlet.2019.12.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 02/08/2023]
Abstract
Understanding the molecular mechanisms regulating tumor dissemination and therapeutic resistance is of central importance for effective cancer therapies. Here, we report that nerve growth factor (NGF) and its receptor TrkA facilitate epithelial-mesenchymal transition (EMT) and EGFR inhibitor resistance via STAT3 activation in head and neck squamous cell carcinoma (HNSCC). Both NGF and TrkA expression were elevated in HNSCC, indicating poor clinical outcomes. NGF was highly expressed in cancer cells and nerves in perineural niche, whereas TrkA expression was higher in cancer cells with perineural invasion. The NGF/TrkA axis could promote HNSCC cell dissemination and trigger EMT via STAT3 activation. Moreover, we discovered that the NGF/TrkA axis conferred resistance to the EGFR inhibitor erlotinib via EMT processes in HNSCC cells. Blocking TrkA signaling markedly reversed EMT and sensitized HNSCC cells to erlotinib in both in vitro and in vivo models. Overall, our results demonstrate novel evidence that the paracrine NGF/TrkA axis favors EMT and confers EGFR-targeted therapeutic resistance in HNSCC.
Collapse
Affiliation(s)
- Chengzhong Lin
- The 2nd Dental Center, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Zhenhu Ren
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Xi Yang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Rong Yang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Yiming Chen
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Zheqi Liu
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Zhenlin Dai
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Yu Zhang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Youya He
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Chunye Zhang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xu Wang
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China.
| | - Wei Cao
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China.
| | - Tong Ji
- National Clinical Research Center for Oral Disease, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, China; Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
130
|
|
131
|
Cao P, Li Y, Tang Y, Ding C, Hunter DJ. Pharmacotherapy for knee osteoarthritis: current and emerging therapies. Expert Opin Pharmacother 2020; 21:797-809. [PMID: 32100600 DOI: 10.1080/14656566.2020.1732924] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Peihua Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yamin Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yujin Tang
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - David J. Hunter
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Rheumatology, Royal North Shore Hospital and Institute of Bone and Joint Research, Kolling Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
132
|
Conaghan PG, Cook AD, Hamilton JA, Tak PP. Therapeutic options for targeting inflammatory osteoarthritis pain. Nat Rev Rheumatol 2020; 15:355-363. [PMID: 31068673 DOI: 10.1038/s41584-019-0221-y] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pain is the major symptom of osteoarthritis (OA) and is an important factor in strategies to manage this disease. However, the current standard of care does not provide satisfactory pain relief for many patients. The pathophysiology of OA is complex, and its presentation as a clinical syndrome is associated with pathologies of multiple joint tissues. Inflammation is associated with both OA pain and disease outcome and is therefore a major treatment target for OA and OA pain. Unlike TNF inhibitors and IL-1 inhibitors, established drugs such as glucocorticoids and methotrexate can reduce OA pain. Although central nociceptive pathways contribute to OA pain, crosstalk between the immune system and nociceptive neurons is central to inflammatory pain; therefore, new therapies might target this crosstalk. Newly identified drug targets, including neurotrophins and the granulocyte-macrophage colony-stimulating factor (GM-CSF)-CC-chemokine ligand 17 (CCL17) chemokine axis, offer the hope of better results but require clinical validation.
Collapse
Affiliation(s)
- Philip G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, and National Institute of Health Research Leeds Biomedical Research Centre, Leeds, UK
| | - Andrew D Cook
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - John A Hamilton
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St Albans, Victoria, Australia
| | - Paul P Tak
- Department of Clinical Immunology & Rheumatology, Academic Medical Centre, Amsterdam University Medical Centre, Amsterdam, Netherlands. .,Department of Rheumatology, Ghent University, Ghent, Belgium. .,Department of Medicine, Cambridge University, Cambridge, UK. .,Flagship Pioneering, Cambridge, MA, USA.
| |
Collapse
|
133
|
Xu M, Bennett DLH, Querol LA, Wu LJ, Irani SR, Watson JC, Pittock SJ, Klein CJ. Pain and the immune system: emerging concepts of IgG-mediated autoimmune pain and immunotherapies. J Neurol Neurosurg Psychiatry 2020; 91:177-188. [PMID: 30224548 DOI: 10.1136/jnnp-2018-318556] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/18/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022]
Abstract
The immune system has long been recognised important in pain regulation through inflammatory cytokine modulation of peripheral nociceptive fibres. Recently, cytokine interactions in brain and spinal cord glia as well as dorsal root ganglia satellite glia have been identified important- in pain modulation. The result of these interactions is central and peripheral sensitisation of nociceptive processing. Additionally, new insights and the term 'autoimmune pain' have emerged through discovery of specific IgGs targeting the extracellular domains of antigens at nodal and synaptic structures, causing pain directly without inflammation by enhancing neuronal excitability. Other discovered IgGs heighten pain indirectly by T-cell-mediated inflammation or destruction of targets within the nociceptive pathways. Notable identified IgGs in pain include those against the components of channels and receptors involved in inhibitory or excitatory somatosensory synapses or their pathways: nodal and paranodal proteins (LGI1, CASPR1, CASPR2); glutamate detection (AMPA-R); GABA regulation and release (GAD65, amphiphysin); glycine receptors (GLY-R); water channels (AQP4). These disorders have other neurological manifestations of central/peripheral hyperexcitabability including seizures, encephalopathy, myoclonus, tremor and spasticity, with immunotherapy responsiveness. Other pain disorders, like complex regional pain disorder, have been associated with IgGs against β2-adrenergic receptor, muscarinic-2 receptors, AChR-nicotinic ganglionic α-3 receptors and calcium channels (N and P/Q types), but less consistently with immune treatment response. Here, we outline how the immune system contributes to development and regulation of pain, review specific IgG-mediated pain disorders and summarise recent development in therapy approaches. Biological agents to treat pain (anti-calcitonin gene-related peptide and anti-nerve growth factor) are also discussed.
Collapse
Affiliation(s)
- Min Xu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Neurology, Xuan wu Hospital Capital Medical University, Beijing, China
| | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Luis Antonio Querol
- Neuromuscular Diseases Unit-Neuromuscular Lab Neurology Department, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarosh R Irani
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - James C Watson
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Pain Anesthesiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sean J Pittock
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher J Klein
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA .,Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
134
|
Sun S, Diggins NH, Gunderson ZJ, Fehrenbacher JC, White FA, Kacena MA. No pain, no gain? The effects of pain-promoting neuropeptides and neurotrophins on fracture healing. Bone 2020; 131:115109. [PMID: 31715336 PMCID: PMC6934100 DOI: 10.1016/j.bone.2019.115109] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/30/2022]
Abstract
Neuropeptides and neurotrophins are key regulators of peripheral nociceptive nerves and contribute to the induction, sensitization, and maintenance of pain. It is now known that these peptides also regulate non-neuronal tissues, including bone. Here, we review the effects of numerous neuropeptides and neurotrophins on fracture healing. The neuropeptides calcitonin-gene related peptide (CGRP), substance P (SP), vasoactive intestinal peptide (VIP), and pituitary adenylate cyclase-activating peptide (PACAP) have varying effects on osteoclastic and osteoblastic activity. Ultimately, CGRP and SP both accelerate fracture healing, while VIP and PACAP seem to negatively impact healing. Unlike the aforementioned neuropeptides, the neurotrophins nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) have more uniform effects. Both factors upregulate osteoblastic activity, osteoclastic activity, and, in vivo, stimulate osteogenesis to promote fracture healing. Future research will need to clarify the exact mechanism by which the neuropeptides and neurotrophins influence fracture healing. Specifically, understanding the optimal expression patterns for these proteins in the fracture healing process may lead to therapies that can maximize their bone-healing capabilities and minimize their pain-promoting effects. Finally, further examination of protein-sequestering antibodies and/or small molecule agonists and antagonists may lead to new therapies that can decrease the rate of delayed union/nonunion outcomes and fracture-associated pain.
Collapse
Affiliation(s)
- Seungyup Sun
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Nicklaus H Diggins
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Zachary J Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Jill C Fehrenbacher
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, IN, USA
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA.
| |
Collapse
|
135
|
March B, Faulkner S, Jobling P, Steigler A, Blatt A, Denham J, Hondermarck H. Tumour innervation and neurosignalling in prostate cancer. Nat Rev Urol 2020; 17:119-130. [PMID: 31937919 DOI: 10.1038/s41585-019-0274-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2019] [Indexed: 01/06/2023]
Abstract
Prostate cancer progression has been shown to be dependent on the development of autonomic nerves into the tumour microenvironment. Sympathetic nerves activate adrenergic neurosignalling that is necessary in early stages of tumour progression and for initiating an angiogenic switch, whereas parasympathetic nerves activate cholinergic neurosignalling resulting in tumour dissemination and metastasis. The innervation of prostate cancer seems to be initiated by neurotrophic growth factors, such as the precursor to nerve growth factor secreted by tumour cells, and the contribution of brain-derived neural progenitor cells has also been reported. Current experimental, epidemiological and clinical evidence shows the stimulatory effect of tumour innervation and neurosignalling in prostate cancer. Using nerves and neurosignalling could have value in the management of prostate cancer by predicting aggressive disease, treating localized disease through denervation and relieving cancer-associated pain in bone metastases.
Collapse
Affiliation(s)
- Brayden March
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Department of Surgery, John Hunter Hospital, New Lambton Heights, NSW, Australia.,Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, Australia
| | - Sam Faulkner
- Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Phillip Jobling
- Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Allison Steigler
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Newcastle Calvary Mater Hospital, Waratah, NSW, Australia
| | - Alison Blatt
- Department of Surgery, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Jim Denham
- School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Newcastle Calvary Mater Hospital, Waratah, NSW, Australia
| | - Hubert Hondermarck
- Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, Australia. .,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
136
|
Oehler B, Brack A, Blum R, Rittner HL. Pain Control by Targeting Oxidized Phospholipids: Functions, Mechanisms, Perspectives. Front Endocrinol (Lausanne) 2020; 11:613868. [PMID: 33569042 PMCID: PMC7868524 DOI: 10.3389/fendo.2020.613868] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Within the lipidome oxidized phospholipids (OxPL) form a class of chemically highly reactive metabolites. OxPL are acutely produced in inflamed tissue and act as endogenous, proalgesic (pain-inducing) metabolites. They excite sensory, nociceptive neurons by activating transient receptor potential ion channels, specifically TRPA1 and TRPV1. Under inflammatory conditions, OxPL-mediated receptor potentials even potentiate the action potential firing rate of nociceptors. Targeting OxPL with D-4F, an apolipoprotein A-I mimetic peptide or antibodies like E06, specifically binding oxidized headgroups of phospholipids, can be used to control acute, inflammatory pain syndromes, at least in rodents. With a focus on proalgesic specificities of OxPL, this article discusses, how targeting defined substances of the epilipidome can contribute to mechanism-based therapies against primary and secondary chronic inflammatory or possibly also neuropathic pain.
Collapse
Affiliation(s)
- Beatrice Oehler
- Wolfson Center of Age-Related Diseases, IoPPN, Health and Life Science, King’s College London, London, United Kingdom
- Department of Anesthesiology, University Hospital of Heidelberg, Heidelberg, Germany
- Department of Anesthesiology, University Hospital of Würzburg, Würzburg, Germany
| | - Alexander Brack
- Department of Anesthesiology, University Hospital of Würzburg, Würzburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Heike L. Rittner
- Department of Anesthesiology, University Hospital of Würzburg, Würzburg, Germany
- *Correspondence: Heike L. Rittner,
| |
Collapse
|
137
|
Manion J, Waller MA, Clark T, Massingham JN, Neely GG. Developing Modern Pain Therapies. Front Neurosci 2019; 13:1370. [PMID: 31920521 PMCID: PMC6933609 DOI: 10.3389/fnins.2019.01370] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022] Open
Abstract
Chronic pain afflicts as much as 50% of the population at any given time but our methods to address pain remain limited, ineffective and addictive. In order to develop new therapies an understanding of the mechanisms of painful sensitization is essential. We discuss here recent progress in the understanding of mechanisms underlying pain, and how these mechanisms are being targeted to produce modern, specific therapies for pain. Finally, we make recommendations for the next generation of targeted, effective, and safe pain therapies.
Collapse
Affiliation(s)
- John Manion
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Matthew A. Waller
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Teleri Clark
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Joshua N. Massingham
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - G. Gregory Neely
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
- Genome Editing Initiative, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
138
|
Han L, Jiang J, Xue M, Qin T, Xiao Y, Wu E, Shen X, Ma Q, Ma J. Sonic hedgehog signaling pathway promotes pancreatic cancer pain via nerve growth factor. Reg Anesth Pain Med 2019; 45:137-144. [PMID: 31792027 DOI: 10.1136/rapm-2019-100991] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/30/2019] [Accepted: 11/03/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Many patients with pancreatic cancer (PC) suffer from abdominal pain and back pain. However, the cause of pain associated with PC is largely unclear. In this study, we tested the potential influence of the sonic hedgehog (sHH) signaling pathway on PC pain. METHODS Substance P (SP) and calcitonin gene-related peptide (CGRP) expression was measured in cultured PC cells and dorsal root ganglions (DRG) by real-time PCR, western blotting analysis and ELISA. Small interfering RNA transfection and plasmid constructs were used to regulate the expression of sHH in the AsPc-1 and Panc-1 cell lines. Pain-related behavior was observed in an orthotopic tumor model in nude mice. RESULTS In this study, the results show that sHH increased the expression of SP and CGRP in DRGs in a concentration and time-dependent manner. Additionally, sHH secretion from PC cells could activate the sHH signaling pathway and, in turn, increase the expression of nerve growth factor (NGF), P75, and TrkA in DRGs. Furthermore, the sHH signaling pathway and NGF/NGF receptor contributed to pain sensitivity in a nude mouse model. CONCLUSION Our results demonstrate that PC pain originates from the sHH signaling pathway, and NGF mediates the pain mechanism via regulating SP and CGRP.
Collapse
Affiliation(s)
- Liang Han
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Jie Jiang
- Department of Medical Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Mengwen Xue
- Department of Anesthesiology, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Erxi Wu
- Department of Surgery, Baylor Scott and White Health, Dallas, Texas, USA.,Department of Neurosurgery, Baylor Scott and White Health, Dallas, Texas, USA
| | - Xin Shen
- Department of Anesthesiology, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, Shaanxi, China
| | - Jiguang Ma
- Department of Anesthesiology, Xi'an Jiaotong University Medical College First Affiliated Hospital, Xi'an, China
| |
Collapse
|
139
|
The NGF R100W Mutation Specifically Impairs Nociception without Affecting Cognitive Performance in a Mouse Model of Hereditary Sensory and Autonomic Neuropathy Type V. J Neurosci 2019; 39:9702-9715. [PMID: 31685654 DOI: 10.1523/jneurosci.0688-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/01/2019] [Accepted: 10/27/2019] [Indexed: 12/22/2022] Open
Abstract
Nerve growth factor (NGF) is a key mediator of nociception, acting during the development and differentiation of dorsal root ganglion (DRG) neurons, and on adult DRG neuron sensitization to painful stimuli. NGF also has central actions in the brain, where it regulates the phenotypic maintenance of cholinergic neurons. The physiological function of NGF as a pain mediator is altered in patients with Hereditary Sensory and Autonomic Neuropathy type V (HSAN V), caused by the 661C>T transition in the Ngf gene, resulting in the R100W missense mutation in mature NGF. Homozygous HSAN V patients present with congenital pain insensitivity, but are cognitively normal. This led us to hypothesize that the R100W mutation may differentially affect the central and peripheral actions of NGF. To test this hypothesis and provide a mechanistic basis to the HSAN V phenotype, we generated transgenic mice harboring the human 661C>T mutation in the Ngf gene and studied both males and females. We demonstrate that heterozygous NGFR100W/wt mice display impaired nociception. DRG neurons of NGFR100W/wt mice are morphologically normal, with no alteration in the different DRG subpopulations, whereas skin innervation is reduced. The NGFR100W protein has reduced capability to activate pain-specific signaling, paralleling its reduced ability to induce mechanical allodynia. Surprisingly, however, NGFR100W/wt mice, unlike heterozygous mNGF+/- mice, show no learning or memory deficits, despite a reduction in secretion and brain levels of NGF. The results exclude haploinsufficiency of NGF as a mechanistic cause for heterozygous HSAN V mice and demonstrate a specific effect of the R100W mutation on nociception.SIGNIFICANCE STATEMENT The R100W mutation in nerve growth factor (NGF) causes Hereditary Sensory and Autonomic Neuropathy type V, a rare disease characterized by impaired nociception, even in apparently clinically silent heterozygotes. For the first time, we generated and characterized heterozygous knock-in mice carrying the human R100W-mutated allele (NGFR100W/wt). Mutant mice have normal nociceptor populations, which, however, display decreased activation of pain transduction pathways. NGFR100W interferes with peripheral and central NGF bioavailability, but this does not impact on CNS function, as demonstrated by normal learning and memory, in contrast with heterozygous NGF knock-out mice. Thus, a point mutation allows neurotrophic and pronociceptive functions of NGF to be split, with interesting implications for the treatment of chronic pain.
Collapse
|
140
|
Obeidat AM, Miller RE, Miller RJ, Malfait AM. The nociceptive innervation of the normal and osteoarthritic mouse knee. Osteoarthritis Cartilage 2019; 27:1669-1679. [PMID: 31351964 PMCID: PMC7020657 DOI: 10.1016/j.joca.2019.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 07/01/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To document the nociceptive innervation of the normal and osteoarthritic murine knee. METHODS Knees were collected from naïve male C57BL/6 NaV1.8-tdTomato reporter mice aged 10, 26, and 52 weeks (n = 5/group). Destabilization of the medial meniscus (DMM) or sham surgeries (n = 5/group) were performed in the right knee of 10-week old male NaV1.8-tdTomato mice, and knees were harvested 16 weeks later. Twenty 20-μm frozen sections from a 400-μm mid-joint region were collected for confocal microscopy. Integrated density of the tdTomato signal was calculated using Image J by two independent observers blinded to the groups. Consecutive sections were stained with hematoxylin & eosin. C57BL/6-Pirt-GCaMP3 mice (n = 5/group) and protein gene product 9.5 (PGP9.5) immunostaining of C57BL/6 wild type (WT) mice (n = 5/group) were used to confirm innervation patterns. RESULTS In naive 10-week old mice, nociceptive innervation was most dense in bone marrow cavities, lateral synovium and at the insertions of the cruciate ligaments. By age 26 weeks, unoperated knees showed a marked decline in nociceptors in the lateral synovium and cruciate ligament insertions. No further decline was observed by age 1 year. Sixteen weeks after DMM, the medial compartment of OA knees exhibited striking changes in NaV1.8+ innervation, including increased innervation of the medial synovium and meniscus, and nociceptors in subchondral bone channels. All results were confirmed through quantification, also in Pirt-GCaMP3 and PGP9.5-immunostained WT mice. CONCLUSIONS Nociceptive innervation of the mouse knee markedly declines by age 26 weeks, before onset of spontaneous OA. Late-stage surgically induced OA is associated with striking plasticity of joint afferents in the medial compartment of the knee.
Collapse
Affiliation(s)
- Alia M. Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago IL
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago IL
| | | | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago IL
| |
Collapse
|
141
|
Miller RE, Scanzello CR, Malfait AM. An emerging role for Toll-like receptors at the neuroimmune interface in osteoarthritis. Semin Immunopathol 2019; 41:583-594. [PMID: 31612243 DOI: 10.1007/s00281-019-00762-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA) is a chronic progressive, painful disease of synovial joints, characterized by cartilage degradation, subchondral bone remodeling, osteophyte formation, and synovitis. It is now widely appreciated that the innate immune system, and in particular Toll-like receptors (TLRs), contributes to pathological changes in OA joint tissues. Furthermore, it is now also increasingly recognized that TLR signaling plays a key role in initiating and maintaining pain. Here, we reviewed the literature of the past 5 years with a focus on how TLRs may contribute to joint damage and pain in OA. We discuss biological effects of specific damage-associated molecular patterns (DAMPs) which act as TLR ligands in vitro, including direct effects on pain-sensing neurons. We then discuss the phenotype of transgenic mice that target TLR pathways, and provide evidence for a complex balance between pro- and anti-inflammatory signaling pathways activated by OA DAMPs. Finally, we summarize clinical evidence implicating TLRs in OA pathogenesis, including polymorphisms and surrogate markers of disease activity. Our review of the literature led us to propose a model where multi-directional crosstalk between connective tissue cells (chondrocytes, fibroblasts), innate immune cells, and sensory neurons in the affected joint may promote OA pathology and pain.
Collapse
Affiliation(s)
- Rachel E Miller
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1611 W Harrison Street, Chicago, IL, 60612, USA
| | - Carla R Scanzello
- Section of Rheumatology and Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center & Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Anne-Marie Malfait
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1611 W Harrison Street, Chicago, IL, 60612, USA.
| |
Collapse
|
142
|
Huang K, Cai HL, Wu LD. Potential of dehydroepiandrosterone in modulating osteoarthritis-related pain. Steroids 2019; 150:108433. [PMID: 31229511 DOI: 10.1016/j.steroids.2019.108433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023]
Abstract
Osteoarthritis (OA) is the most common form of degenerative arthropathy, and the primary symptom is chronic joint pain. Dehydroepiandrosterone (DHEA) exerts a chondroprotective effect against OA and has been reported to have potent structure-modifying effects on osteoarthritic cartilage, thereby attenuating disease progression. However, the ability of DHEA to modulate OA-related pain has not yet been verified. Recent evidence suggests that there may be a link between the pharmacological effects of DHEA and pain generation. For example, DHEA synthesized in the adrenal gland interferes directly with nerve growth factor (NGF) receptors, a major biochemical contributor to peripheral hypersensitivity. Similarly, endogenous DHEA produced in the spinal cord exerts a regulatory effect on nociception in neuropathic rats. In this short review, we discuss recent studies concerning crucial signalling cascades and molecular mechanisms involved in pain generation as well as the potential link between DHEA activity and nociception. Particular attention is given to the putative molecular mechanisms underlying the favourable efficacy of DHEA against pain generation. Elucidating the molecular mechanisms of DHEA against osteoarthritic pain may pave the way for the discovery and development of novel anti-OA drugs, as effective drugs for OA treatment are not currently available.
Collapse
Affiliation(s)
- Kai Huang
- Department of Orthopedic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, PR China.
| | - Hai-Li Cai
- Department of Ultrasound, The 903rd Hospital of PLA, Hangzhou 310012, PR China
| | - Li-Dong Wu
- Department of Orthopedic Surgery, The Second Hospital of Medical College, Zhejiang University, Hangzhou 310009, PR China
| |
Collapse
|
143
|
Dakin P, DiMartino SJ, Gao H, Maloney J, Kivitz AJ, Schnitzer TJ, Stahl N, Yancopoulos GD, Geba GP. The Efficacy, Tolerability, and Joint Safety of Fasinumab in Osteoarthritis Pain: A Phase IIb/III Double-Blind, Placebo-Controlled, Randomized Clinical Trial. Arthritis Rheumatol 2019; 71:1824-1834. [PMID: 31207169 PMCID: PMC6900077 DOI: 10.1002/art.41012] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 06/11/2019] [Indexed: 01/20/2023]
Abstract
Objective To prospectively assess the efficacy, general safety, and joint safety of fasinumab, an anti–nerve growth factor monoclonal antibody, in osteoarthritis (OA) hip and/or knee pain. Methods Patients with moderate‐to‐severe OA pain (knee or hip) and history of inadequate response or intolerance to analgesics were randomized to receive fasinumab (at 1 mg, 3 mg, 6 mg, or 9 mg) or placebo every 4 weeks over 16 weeks and were followed up to week 36. Efficacy end points were the change from baseline to week 16 in the pain and physical function subscale scores of the Western Ontario and McMaster Universities OA Index (WOMAC), and patient global assessment (PGA) of OA. Joints were monitored at scheduled assessments (by plain film radiography and magnetic resonance imaging) during treatment and follow‐up, and if prompted, at the time of active joint symptoms. Results Of the 421 patients randomized, 342 completed the 36‐week study. All doses of fasinumab yielded statistically significant and clinically important reductions in pain compared to placebo (least squares mean difference in WOMAC pain subscale scores at week 16 ranging −0.78 to −1.40), without any clear dose dependence. Physical function and PGA scores improved in parallel. Treatment‐emergent adverse event rates were 17% with fasinumab and 10% with placebo, and 4% and 1% of patients, respectively, discontinued treatment. Arthropathies (25 in total, 7% of fasinumab‐treated patients and 1% of placebo‐treated patients) occurred in a dose‐dependent manner, with 2 occurring in patients receiving the lowest dose of fasinumab and 10 in patients receiving the highest dose. Most of the arthropathies (16 of 25) were discovered with scheduled radiographs and not based on symptoms. Destructive arthropathy (in 1 of 337 treated patients) occurred in 1 patient who was receiving 6 mg fasimumab. Conclusion Fasinumab provided improvements in OA pain and function, even in those benefitting little from previous analgesics. The observed benefit‐to‐risk relationship favors further clinical development to explore the lowest doses of fasinumab in patients with knee or hip OA.
Collapse
Affiliation(s)
- Paula Dakin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Haitao Gao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Alan J Kivitz
- Altoona Center for Clinical Research, Duncansville, Pennsylvania
| | | | - Neil Stahl
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | |
Collapse
|
144
|
Nees TA, Rosshirt N, Reiner T, Schiltenwolf M, Moradi B. [Inflammation and osteoarthritis-related pain]. Schmerz 2019; 33:4-12. [PMID: 30560495 DOI: 10.1007/s00482-018-0346-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is one of the major causes of chronic pain. Although OA has long been considered a non-inflammatory "wear and tear" disease leading to loss of articular cartilage, recent findings provide convincing evidence that inflammatory mechanisms play a pivotal role in the pathophysiology of OA. In OA mononuclear cells (e. g. T‑cells and macrophages) infiltrate the synovial membrane and the levels of pro-inflammatory cytokines in peripheral blood and synovial fluid samples are elevated. Increased release of inflammatory mediators including interleukin (IL) IL-1β, IL-6, IL-8, IL-15 und tumor necrosis factor alpha (TNF‑α) induces the expression of proteolytic enzymes such as matrix metalloproteinases resulting in cartilage breakdown. Molecular and cellular interactions between the immune and nervous system are also involved in the development of OA-related pain. Inflammatory mediators including IL-6 und TNF‑α lead to peripheral sensitization of joint nociceptors and growth factors (e. g. NGF) trigger the expression of TRPV1 channels in primary afferents. Moreover, neuropeptides reduce the threshold of nociceptors of OA joints. The current review highlights the role of inflammatory mechanisms in OA-induced joint pain considering clinical signs of inflammation and major inflammatory pathways.
Collapse
Affiliation(s)
- T A Nees
- Klinik für Orthopädie und Unfallchirurgie, Zentrum für Orthopädie, Unfallchirurgie und Paraplegiologie, Universitätsklinikum Heidelberg, Schlierbacher Landstraße 200a, 69118, Heidelberg, Deutschland
| | - N Rosshirt
- Klinik für Orthopädie und Unfallchirurgie, Zentrum für Orthopädie, Unfallchirurgie und Paraplegiologie, Universitätsklinikum Heidelberg, Schlierbacher Landstraße 200a, 69118, Heidelberg, Deutschland
| | - T Reiner
- Klinik für Orthopädie und Unfallchirurgie, Zentrum für Orthopädie, Unfallchirurgie und Paraplegiologie, Universitätsklinikum Heidelberg, Schlierbacher Landstraße 200a, 69118, Heidelberg, Deutschland
| | - M Schiltenwolf
- Klinik für Orthopädie und Unfallchirurgie, Zentrum für Orthopädie, Unfallchirurgie und Paraplegiologie, Universitätsklinikum Heidelberg, Schlierbacher Landstraße 200a, 69118, Heidelberg, Deutschland
| | - B Moradi
- Klinik für Orthopädie und Unfallchirurgie, Zentrum für Orthopädie, Unfallchirurgie und Paraplegiologie, Universitätsklinikum Heidelberg, Schlierbacher Landstraße 200a, 69118, Heidelberg, Deutschland.
| |
Collapse
|
145
|
Tateiwa D, Yoshikawa H, Kaito T. Cartilage and Bone Destruction in Arthritis: Pathogenesis and Treatment Strategy: A Literature Review. Cells 2019; 8:cells8080818. [PMID: 31382539 PMCID: PMC6721572 DOI: 10.3390/cells8080818] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Arthritis is inflammation of the joints accompanied by osteochondral destruction. It can take many forms, including osteoarthritis, rheumatoid arthritis, and psoriatic arthritis. These diseases share one commonality—osteochondral destruction based on inflammation. The background includes a close interaction between osseous tissues and immune cells through various inflammatory cytokines. However, the tissues and cytokines that play major roles are different in each disease, and as a result, the mechanism of osteochondral destruction also differs. In recent years, there have been many findings regarding not only extracellular signaling pathways but also intracellular signaling pathways. In particular, we anticipate that the intracellular signals of osteoclasts, which play a central role in bone destruction, will become novel therapeutic targets. In this review, we have summarized the pathology of arthritis and the latest findings on the mechanism of osteochondral destruction, as well as present and future therapeutic strategies for these targets.
Collapse
Affiliation(s)
- Daisuke Tateiwa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Kaito
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
146
|
Ghouri A, Conaghan PG. Update on novel pharmacological therapies for osteoarthritis. Ther Adv Musculoskelet Dis 2019; 11:1759720X19864492. [PMID: 31384314 PMCID: PMC6651659 DOI: 10.1177/1759720x19864492] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/25/2019] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is a chronic painful arthritis with increasing global prevalence. Current management involves non-pharmacological interventions and commonly used pharmacological treatments that generally have limited analgesic efficacy and multiple side effects. New treatments are therefore required to relieve patient symptoms and disease impact. A number of existing pharmacological therapies have been recently trialled in OA. These include extended-release triamcinolone and conventional disease-modifying anti-rheumatic drugs (DMARDs) used in the management of rheumatoid arthritis; generally, DMARDs have not shown a benefit in treating OA. Novel analgesic therapies are in development, including those targeting peripheral pain pathways. Disease-modifying osteoarthritis drugs (DMOADs) target key tissues in the OA pathophysiology process and aim to prevent structural progression; a number of putative DMOADs are in phase II development. There is preliminary evidence of structural improvement with some of these therapies but without concomitant symptom improvement, raising new considerations for future DMOAD trials.
Collapse
Affiliation(s)
- Asim Ghouri
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - Philip G. Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Chapeltown Rd, Leeds LS7 4SA, UK
| |
Collapse
|
147
|
Abstract
Pain is a hallmark of tissue injury, inflammatory diseases, pathogen invasion and neuropathy. It is mediated by nociceptor sensory neurons that innervate the skin, joints, bones, muscles and mucosal tissues and protects organisms from noxious stimuli. Nociceptors are sensitized by inflammatory mediators produced by the immune system, including cytokines, lipid mediators and growth factors, and can also directly detect pathogens and their secreted products to produce pain during infection. Upon activation, nociceptors release neuropeptides from their terminals that potently shape the function of innate and adaptive immune cells. For some pathogens, neuron-immune interactions enhance host protection from infection, but for other pathogens, neuron-immune signalling pathways can be exploited to facilitate pathogen survival. Here, we discuss the role of nociceptor interactions with the immune system in pain and infection and how understanding these pathways could produce new approaches to treat infectious diseases and chronic pain.
Collapse
|
148
|
Abstract
Beyond their well-known role in embryonic development of the central and peripheral nervous system, neurotrophins, particularly nerve growth factor and brain-derived neurotrophic factor, exert an essential role in pain production and sensitization. This has mainly been studied within the framework of somatic pain, and even antibodies (tanezumab and fasinumab) have recently been developed for their use in chronic somatic painful conditions, such as osteoarthritis or low back pain. However, data suggest that neurotrophins also exert an important role in the occurrence of visceral pain and visceral sensitization. Visceral pain is a distressing symptom that prompts many consultations and is typically encountered in both 'organic' (generally inflammatory) and 'functional' (displaying no obvious structural changes in routine clinical evaluations) disorders of the gut, such as inflammatory bowel disease and irritable bowel syndrome, respectively. The present review provides a summary of neurotrophins as a molecular family and their role in pain in general and addresses recent investigations of the involvement of nerve growth factor and brain-derived neurotrophic factor in visceral pain, particularly that associated with inflammatory bowel disease and irritable bowel syndrome.
Collapse
|
149
|
Nociceptor Signalling through ion Channel Regulation via GPCRs. Int J Mol Sci 2019; 20:ijms20102488. [PMID: 31137507 PMCID: PMC6566991 DOI: 10.3390/ijms20102488] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022] Open
Abstract
The prime task of nociceptors is the transformation of noxious stimuli into action potentials that are propagated along the neurites of nociceptive neurons from the periphery to the spinal cord. This function of nociceptors relies on the coordinated operation of a variety of ion channels. In this review, we summarize how members of nine different families of ion channels expressed in sensory neurons contribute to nociception. Furthermore, data on 35 different types of G protein coupled receptors are presented, activation of which controls the gating of the aforementioned ion channels. These receptors are not only targeted by more than 20 separate endogenous modulators, but can also be affected by pharmacotherapeutic agents. Thereby, this review provides information on how ion channel modulation via G protein coupled receptors in nociceptors can be exploited to provide improved analgesic therapy.
Collapse
|
150
|
Dong Y, Li S, Yin J, Chen JDZ. Ameliorating effects of optimized gastric electrical stimulation and mechanisms involving nerve growth factor and opioids in a rodent model of gastric hypersensitivity. Neurogastroenterol Motil 2019; 31:e13551. [PMID: 30790401 DOI: 10.1111/nmo.13551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 12/09/2018] [Accepted: 12/20/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Gastric electrical stimulation (GES) has been applied to treat gastric motility disorders for decades. This study was designed to investigate the effects and mechanisms of GES for visceral hypersensitivity in a rodent model of functional dyspepsia (FD). METHODS Male Sprague-Dawley rat pups at 10-days old received 0.1% iodoacetamide (IA) daily for 6 days. The experiments were performed when the rats reached 8-11 weeks of age, and visceral hypersensitivity was established. Then, GES parameters were optimized and the chronic effects of GES on gastric hypersensitivity were assessed by electromyogram (EMG). Naloxone (3 mg/kg), D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP, 1 mg/kg), and anti-NGF (16 μg/kg) were individually intraperitoneally injected to investigate opioid and nerve growth factor (NGF) mechanisms. Tissues were analyzed for NGF expression. KEY RESULTS In the IA-treated rats, the visceromotor response to gastric distension was significantly increased, and both acute GES with optimized stimulation parameters (0.25 seconds on, 0.25 seconds off, 100 Hz, 0.25 ms, 6 mA) and chronic GES (7 days, 2 hours/day) normalized gastric hypersensitivity. The inhibitory effect of GES on gastric hypersensitivity was blocked by naloxone and CTOP. Anti-NGF normalized EMG responses in IA-treated rats. The expressions of NGF in the tissues of IA-treated rats were dramatically increased, and these increases were suppressed with GES. CONCLUSIONS AND INFERENCES GES with optimized parameters improves gastric hypersensitivity induced by neonatal treatment of IA mediated peripherally by suppressing NGF and via the opioid mechanism involving the µ receptor. GES as a potential therapy for treating visceral pain may be explored in clinical studies.
Collapse
Affiliation(s)
- Yan Dong
- Veterans Research and Education Foundation, VA Medical Center, Oklahoma City, Oklahoma.,Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shiying Li
- Veterans Research and Education Foundation, VA Medical Center, Oklahoma City, Oklahoma.,Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jieyun Yin
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jiande D Z Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|