1
|
O'Brien MS, McDougall JJ. Comparison of nociceptor properties using electrophysiology in preclinical models of osteoarthritis. Neurosci Lett 2024; 840:137950. [PMID: 39182667 DOI: 10.1016/j.neulet.2024.137950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Osteoarthritis (OA) pain originates in the joint by sensitization of articular nociceptors. While behavioural assessments provide valuable information regarding pain symptoms, the techniques are subjective and open to interpretation by the experimenter. This study used in vivo electrophysiological approaches to measure objectively joint nociceptor properties in three rodent models of OA. Single unit extracellular recordings of joint mechanosensitive afferents were carried out in male and female rats following either (1) transection of the medial meniscus (MMT: post-traumatic OA), (2) intra-articular injection of sodium monoiodoacetate (MIA: chemically-induced OA), or (3) intra-articular injection of lysophosphatidic acid (LPA: neuropathic OA). In naïve male control rats, the mechanical threshold of joint mechanonociceptors (23.5 ± 1.8 mNm) was significantly reduced with MMT (9.4 ± 1.1 mNm) and MIA (15.1 ± 1.6 mNm). In females, the mechanical threshold of naïve rats (23.2 ± 3.1 mNm) was reduced following induction of MMT (8.3 ± 1.0 mNm) and LPA (10.6 ± 2.2 mNm). Afferent firing frequency increased in male MMT (∼275 %), LPA (∼175 %), MIA (225 %), and female MMT (∼146 %), LPA (∼200 %), and MIA (∼192 %). Mechanical threshold and evoked firing were negatively correlated in all models for both sexes except LPA rats (male + female) and female MMT. These data indicate that MMT, MIA, and LPA induce peripheral sensitization of joint afferents thereby validating their use in OA pain studies.
Collapse
Affiliation(s)
- Melissa S O'Brien
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, Nova Scotia, B3H 4R2, Canada.
| | - Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, Nova Scotia, B3H 4R2, Canada.
| |
Collapse
|
2
|
Liu PW, Zhang H, Werley CA, Pichler M, Ryan SJ, Lewarch CL, Jacques J, Grooms J, Ferrante J, Li G, Zhang D, Bremmer N, Barnett A, Chantre R, Elder AE, Cohen AE, Williams LA, Dempsey GT, McManus OB. A phenotypic screening platform for chronic pain therapeutics using all-optical electrophysiology. Pain 2024; 165:922-940. [PMID: 37963235 PMCID: PMC10950549 DOI: 10.1097/j.pain.0000000000003090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 08/30/2023] [Indexed: 11/16/2023]
Abstract
ABSTRACT Chronic pain associated with osteoarthritis (OA) remains an intractable problem with few effective treatment options. New approaches are needed to model the disease biology and to drive discovery of therapeutics. We present an in vitro model of OA pain, where dorsal root ganglion (DRG) sensory neurons were sensitized by a defined mixture of disease-relevant inflammatory mediators, here called Sensitizing PAin Reagent Composition or SPARC. Osteoarthritis-SPARC components showed synergistic or additive effects when applied in combination and induced pain phenotypes in vivo. To measure the effect of OA-SPARC on neural firing in a scalable format, we used a custom system for high throughput all-optical electrophysiology. This system enabled light-based membrane voltage recordings from hundreds of neurons in parallel with single cell and single action potential resolution and a throughput of up to 500,000 neurons per day. A computational framework was developed to construct a multiparameter OA-SPARC neuronal phenotype and to quantitatively assess phenotype reversal by candidate pharmacology. We screened ∼3000 approved drugs and mechanistically focused compounds, yielding data from over 1.2 million individual neurons with detailed assessment of functional OA-SPARC phenotype rescue and orthogonal "off-target" effects. Analysis of confirmed hits revealed diverse potential analgesic mechanisms including ion channel modulators and other mechanisms including MEK inhibitors and tyrosine kinase modulators. Our results suggest that the Raf-MEK-ERK axis in DRG neurons may integrate the inputs from multiple upstream inflammatory mediators found in osteoarthritis patient joints, and MAPK pathway activation in DRG neurons may contribute to chronic pain in patients with osteoarthritis.
Collapse
Affiliation(s)
- Pin W. Liu
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | | | | | | | | | - Guangde Li
- Quiver Bioscience, Cambridge, MA, United States
| | - Dawei Zhang
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | - Adam E. Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | | | | | | |
Collapse
|
3
|
Zhang DH, Fan YH, Zhang YQ, Cao H. Neuroendocrine and neuroimmune mechanisms underlying comorbidity of pain and obesity. Life Sci 2023; 322:121669. [PMID: 37023950 DOI: 10.1016/j.lfs.2023.121669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Pain and obesity, as well as their associated impairments, are major health concerns. Understanding the relationship between the two is the focus of a growing body of research. However, early researches attribute increased mechanical stress from excessive weight as the main factor of obesity-related pain, which not only over-simplify the association, but also fail to explain some controversial outcomes arising from clinical investigations. This review focuses on neuroendocrine and neuroimmune modulators importantly involved in both pain and obesity, analyzing nociceptive and anti-nociceptive mechanisms based on neuroendocrine pathways including galanin, ghrelin, leptin and their interactions with other neuropeptides and hormone systems which have been reported to play roles in pain and obesity. Mechanisms of immune activities and metabolic alterations are also discussed, due to their intense interactions with neuroendocrine system and crucial roles in the development and maintenance of inflammatory and neuropathic pain. These findings have implications for health given rising rates of obesity and pain-related diagnoses, by providing novel weight-control and analgesic therapies targeted on specific pathways.
Collapse
Affiliation(s)
- Dao-Han Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ying-Hui Fan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Hong Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Kitayama E, Kimura M, Ouchi T, Furusawa M, Shibukawa Y. Functional Expression of IP, 5-HT 4, D 1, A 2A, and VIP Receptors in Human Odontoblast Cell Line. Biomolecules 2023; 13:879. [PMID: 37371459 DOI: 10.3390/biom13060879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/26/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Odontoblasts are involved in sensory generation as sensory receptor cells and in dentin formation. We previously reported that an increase in intracellular cAMP levels by cannabinoid 1 receptor activation induces Ca2+ influx via transient receptor potential vanilloid subfamily member 1 channels in odontoblasts, indicating that intracellular cAMP/Ca2+ signal coupling is involved in dentinal pain generation and reactionary dentin formation. Here, intracellular cAMP dynamics in cultured human odontoblasts were investigated to understand the detailed expression patterns of the intracellular cAMP signaling pathway activated by the Gs protein-coupled receptor and to clarify its role in cellular functions. The presence of plasma membrane Gαs as well as prostaglandin I2 (IP), 5-hydroxytryptamine 5-HT4 (5-HT4), dopamine D1 (D1), adenosine A2A (A2A), and vasoactive intestinal polypeptide (VIP) receptor immunoreactivity was observed in human odontoblasts. In the presence of extracellular Ca2+, the application of agonists for the IP (beraprost), 5-HT4 (BIMU8), D1 (SKF83959), A2A (PSB0777), and VIP (VIP) receptors increased intracellular cAMP levels. This increase in cAMP levels was inhibited by the application of the adenylyl cyclase (AC) inhibitor SQ22536 and each receptor antagonist, dose-dependently. These results suggested that odontoblasts express Gs protein-coupled IP, 5-HT4, D1, A2A, and VIP receptors. In addition, activation of these receptors increased intracellular cAMP levels by activating AC in odontoblasts.
Collapse
Affiliation(s)
- Eri Kitayama
- Department of Physiology, Tokyo Dental College, 2-9-18, Kanda-Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
- Department of Endodontics, Tokyo Dental College, 2-9-18, Kanda-Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Maki Kimura
- Department of Physiology, Tokyo Dental College, 2-9-18, Kanda-Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, 2-9-18, Kanda-Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Masahiro Furusawa
- Department of Endodontics, Tokyo Dental College, 2-9-18, Kanda-Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Yoshiyuki Shibukawa
- Department of Physiology, Tokyo Dental College, 2-9-18, Kanda-Misaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
5
|
Reid AR, Côté PD, McDougall JJ. Long-Term Blockade of Nociceptive Na v1.7 Channels Is Analgesic in Rat Models of Knee Arthritis. Biomolecules 2022; 12:1571. [PMID: 36358921 PMCID: PMC9687684 DOI: 10.3390/biom12111571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
The voltage gated sodium channels (Nav) 1.7, 1.8, and 1.9 are primarily located on nociceptors where they are involved in signalling neuropathic pain. This study examined the effect of Nav1.7 blockade on joint pain using either the small molecule inhibitor PF05089771 or an antibody directed towards the intracellular domain of the ion channel. Male Wistar rats were assigned to one of three experimental groups consisting of either intra-articular injection of 3 mg sodium monoiodoacetate (MIA-joint degeneration group), intra-articular injection of 100 μg lysophosphatidic acid (LPA-joint neuropathy group), or transection of the medial meniscus (MMT-posttraumatic osteoarthritis group). G-ratio calculations were performed to determine potential demyelination and immunohistochemistry was used to measure Nav1.7 expression on joint afferent cell bodies. Pain behaviour was evaluated over 3 h by von Frey hair algesiometry and hindlimb weight bearing before and after local administration of PF05089771 (0.1 mg/50 µL). Chronic pain behaviour was assessed over 28 days following peripheral treatment with a Nav1.7 antibody (Ab) in conjunction with the transmembrane carrier peptide Pep1. Demyelination and increased Nav1.7 channel expression were observed in MIA and LPA rats, but not with MMT. Acute secondary allodynia was diminished by PF05089771 while a single injection of Nav1.7 Ab-Pep1 reduced pain up to 28 days. This analgesia only occurred in MIA and LPA animals. Hindlimb incapacitance was not affected by any treatment. These data indicate that joint pain associated with neural demyelination can be alleviated somewhat by Nav1.7 channel blockade. Biologics that inactivate Nav1.7 channels have the potential to reduce arthritis pain over a protracted period of time.
Collapse
Affiliation(s)
- Allison R. Reid
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada
| | - Patrice D. Côté
- Department of Biology, Dalhousie University, 1355 Oxford, Halifax, NS B3H 4R2, Canada
| | - Jason J. McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
6
|
Sokolov AY, Osipchuk AV, Skiba IB, Amelin AV. The Role of Pituitary Adenylate Cyclase-Activating Polypeptide and Vasoactive Intestinal Peptide in Migraine Pathogenesis. NEUROCHEM J+ 2022. [DOI: 10.1134/s1819712422010123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
7
|
Morgan M, Thai J, Nazemian V, Song R, Ivanusic JJ. Changes to the activity and sensitivity of nerves innervating subchondral bone contribute to pain in late-stage osteoarthritis. Pain 2022; 163:390-402. [PMID: 34108432 PMCID: PMC8756348 DOI: 10.1097/j.pain.0000000000002355] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 05/07/2021] [Indexed: 11/30/2022]
Abstract
ABSTRACT Although it is clear that osteoarthritis (OA) pain involves activation and/or sensitization of nociceptors that innervate knee joint articular tissues, much less is known about the role of the innervation of surrounding bone. In this study, we used monoiodoacetate (MIA)-induced OA in male rats to test the idea that pain in OA is driven by differential contributions from nerves that innervate knee joint articular tissues vs the surrounding bone. The time-course of pain behavior was assayed using the advanced dynamic weight-bearing device, and histopathology was examined using haematoxylin and eosin histology. Extracellular electrophysiological recordings of knee joint and bone afferent neurons were made early (day 3) and late (day 28) in the pathogenesis of MIA-induced OA. We observed significant changes in the function of knee joint afferent neurons, but not bone afferent neurons, at day 3 when there was histological evidence of inflammation in the joint capsule, but no damage to the articular cartilage or subchondral bone. Changes in the function of bone afferent neurons were only observed at day 28, when there was histological evidence of damage to the articular cartilage and subchondral bone. Our findings suggest that pain early in MIA-induced OA involves activation and sensitization of nerves that innervate the joint capsule but not the underlying subchondral bone, and that pain in late MIA-induced OA involves the additional recruitment of nerves that innervate the subchondral bone. Thus, nerves that innervate bone should be considered important targets for development of mechanism-based therapies to treat pain in late OA.
Collapse
Affiliation(s)
- Michael Morgan
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Jenny Thai
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Vida Nazemian
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Richard Song
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| | - Jason J. Ivanusic
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Morgan M, Nazemian V, Harrington K, Ivanusic JJ. Mini review: The role of sensory innervation to subchondral bone in osteoarthritis pain. Front Endocrinol (Lausanne) 2022; 13:1047943. [PMID: 36605943 PMCID: PMC9808033 DOI: 10.3389/fendo.2022.1047943] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/30/2022] [Indexed: 01/07/2023] Open
Abstract
Osteoarthritis pain is often thought of as a pain driven by nerves that innervate the soft tissues of the joint, but there is emerging evidence for a role for nerves that innervate the underlying bone. In this mini review we cite evidence that subchondral bone lesions are associated with pain in osteoarthritis. We explore recent studies that provide evidence that sensory neurons that innervate bone are nociceptors that signal pain and can be sensitized in osteoarthritis. Finally, we describe neuronal remodeling of sensory and sympathetic nerves in bone and discuss how these processes can contribute to osteoarthritis pain.
Collapse
|
9
|
Proteomic Analysis of Synovial Fibroblasts and Articular Chondrocytes Co-Cultures Reveals Valuable VIP-Modulated Inflammatory and Degradative Proteins in Osteoarthritis. Int J Mol Sci 2021; 22:ijms22126441. [PMID: 34208590 PMCID: PMC8235106 DOI: 10.3390/ijms22126441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disorder causing a great disability and a reduction in the quality of life. In OA, articular chondrocytes (AC) and synovial fibroblasts (SF) release innate-derived immune mediators that initiate and perpetuate inflammation, inducing cartilage extracellular matrix (ECM) degradation. Given the lack of therapies for the treatment of OA, in this study, we explore biomarkers that enable the development of new therapeutical approaches. We analyze the set of secreted proteins in AC and SF co-cultures by stable isotope labeling with amino acids (SILAC). We describe, for the first time, 115 proteins detected in SF-AC co-cultures stimulated by fibronectin fragments (Fn-fs). We also study the role of the vasoactive intestinal peptide (VIP) in this secretome, providing new proteins involved in the main events of OA, confirmed by ELISA and multiplex analyses. VIP decreases proteins involved in the inflammatory process (CHI3L1, PTX3), complement activation (C1r, C3), and cartilage ECM degradation (DCN, CTSB and MMP2), key events in the initiation and progression of OA. Our results support the anti-inflammatory and anti-catabolic properties of VIP in rheumatic diseases and provide potential new targets for OA treatment.
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Osteoarthritis is a degenerative joint disease that features pain as a hallmark symptom. This review summarises progress and obstacles in our understanding of pain mechanisms in arthritis. RECENT FINDINGS Pain phenotypes in osteoarthritis are poorly characterized in clinical studies and animal studies are largely carti-centric. Different animal models incur variable disease progression patterns and activation of distinct pain pathways, but studies reporting both structural and pain outcomes permit better translational insights. In patients, classification of osteoarthritis disease severity is only based on structural integrity of the joint, but pain outcomes do not consistently correlate with joint damage. The complexity of this relationship underlines the need for pain detection in criteria for osteoarthritis classification and patient-reported outcome measures. SUMMARY Variable inflammatory and neuropathic components and spatiotemporal evolution underlie the heterogeneity of osteoarthritis pain phenotypes, which must be considered to adequately stratify patients. Revised classification of osteoarthritis at different stages encompassing both structural and pain outcomes would significantly improve detection and diagnosis at both early and late stages of disease. These are necessary advancements in the field that would also improve trial design and provide better understanding of basic mechanisms of disease progression and pain in osteoarthritis.
Collapse
|
11
|
Kwok CHT, Kohro Y, Mousseau M, O'Brien MS, Matyas JR, McDougall JJ, Trang T. Role of Primary Afferents in Arthritis Induced Spinal Microglial Reactivity. Front Immunol 2021; 12:626884. [PMID: 33897685 PMCID: PMC8058457 DOI: 10.3389/fimmu.2021.626884] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/18/2021] [Indexed: 11/18/2022] Open
Abstract
Increased afferent input resulting from painful injury augments the activity of central nociceptive circuits via both neuron-neuron and neuron-glia interactions. Microglia, resident immune cells of the central nervous system (CNS), play a crucial role in the pathogenesis of chronic pain. This study provides a framework for understanding how peripheral joint injury signals the CNS to engage spinal microglial responses. During the first week of monosodium iodoacetate (MIA)-induced knee joint injury in male rats, inflammatory and neuropathic pain were characterized by increased firing of peripheral joint afferents. This increased peripheral afferent activity was accompanied by increased Iba1 immunoreactivity within the spinal dorsal horn indicating microglial activation. Pharmacological silencing of C and A afferents with co-injections of QX-314 and bupivacaine, capsaicin, or flagellin prevented the development of mechanical allodynia and spinal microglial activity after MIA injection. Elevated levels of ATP in the cerebrospinal fluid (CSF) and increased expression of the ATP transporter vesicular nucleotide transporter (VNUT) in the ipsilateral spinal dorsal horn were also observed after MIA injections. Selective silencing of primary joint afferents subsequently inhibited ATP release into the CSF. Furthermore, increased spinal microglial reactivity, and alleviation of MIA-induced arthralgia with co-administration of QX-314 with bupivacaine were recapitulated in female rats. Our results demonstrate that early peripheral joint injury activates joint nociceptors, which triggers a central spinal microglial response. Elevation of ATP in the CSF, and spinal expression of VNUT suggest ATP signaling may modulate communication between sensory neurons and spinal microglia at 2 weeks of joint degeneration.
Collapse
Affiliation(s)
- Charlie H T Kwok
- Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Yuta Kohro
- Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Michael Mousseau
- Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Melissa S O'Brien
- Departments of Pharmacology and Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - John R Matyas
- Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
| | - Jason J McDougall
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tuan Trang
- Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
Kanemitsu M, Nakasa T, Shirakawa Y, Ishikawa M, Miyaki S, Adachi N. Role of vasoactive intestinal peptide in the progression of osteoarthritis through bone sclerosis and angiogenesis in subchondral bone. J Orthop Sci 2020; 25:897-906. [PMID: 31928851 DOI: 10.1016/j.jos.2019.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/25/2019] [Accepted: 11/18/2019] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Osteoarthritis (OA) is a progressive joint disorder, with abnormal remodeling of subchondral bone linked to the disruption of cartilage metabolism. Nerves also play an important role in bone remodeling in OA progression, and vasoactive intestinal peptide (VIP), one of the neuropeptides, plays an important role in bone metabolism. The aim of this study was to analyze the expression pattern of VIP in subchondral bone, and its potential as a therapeutic target for OA progression. DESIGN The pattern of VIP expression in the human tibia was histologically evaluated. The effect of VIP on angiogenesis was investigated using human umbilical vein endothelial cells (HUVECs). Knee OA was induced by the resection of the medial meniscotibial ligament in C57BL/6 mice. A VIP receptor antagonist was intraperitoneally administered postoperatively, and therapeutic effects were analyzed at 4 and 8 weeks. RESULTS VIP expression in the subchondral bone increased as OA progressed in human tibia. VIP was also expressed in the vascular channels into the cartilage layer. The total length and branch points were significantly increased, due to the VIP receptor agonist in HUVECs. In OA mice, the VIP receptor antagonist could prevent cartilage degeneration and subchondral bone sclerosis. The Osteoarthritis Research Society International score in the VIP receptor antagonist group was significantly lower than in the control group. CONCLUSION VIP is involved in the progression of OA through its effect on subchondral bone sclerosis and angiogenesis. Inhibition of VIP signaling has the potential to be a therapeutic target to prevent OA progression.
Collapse
Affiliation(s)
- Munekazu Kanemitsu
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Tomoyuki Nakasa
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan; Medical Center for Translation and Clinical Research, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Yoshiko Shirakawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Masakazu Ishikawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shigeru Miyaki
- Medical Center for Translation and Clinical Research, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
13
|
McKenna M, McDougall JJ. Cannabinoid control of neurogenic inflammation. Br J Pharmacol 2020; 177:4386-4399. [PMID: 33289534 DOI: 10.1111/bph.15208] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/02/2020] [Accepted: 07/13/2020] [Indexed: 01/16/2023] Open
Abstract
A significant number of cannabinoids are known to have analgesic and anti-inflammatory properties in various diseases. Due to their presynaptic/terminal location, cannabinoid receptors can inhibit synaptic transmission and have the potential to regulate neurogenic inflammation. Neurogenic inflammation occurs when a noxious signal is detected in the periphery initiating an antidromic axon reflex in the same sensory neurone leading to depolarization of the afferent terminal. Neuropeptides are subsequently released and contribute to vasodilation, plasma extravasation and modulation of immune cells. Endocannabinoids, synthetic cannabinoids and phytocannabinoids can reduce neuroinflammation by inhibiting afferent firing and inflammatory neuropeptide release. Thus, in addition to a direct effect on vascular smooth muscle and inflammatory cells, cannabinoids can reduce inflammation by silencing small diameter neurones. This review examines the neuropharmacological processes involved in regulating antidromic depolarization of afferent nerve terminals by cannabinoids and the control of neurogenic inflammation in different diseases.
Collapse
Affiliation(s)
- Meagan McKenna
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
14
|
Martínez C, Juarranz Y, Gutiérrez-Cañas I, Carrión M, Pérez-García S, Villanueva-Romero R, Castro D, Lamana A, Mellado M, González-Álvaro I, Gomariz RP. A Clinical Approach for the Use of VIP Axis in Inflammatory and Autoimmune Diseases. Int J Mol Sci 2019; 21:E65. [PMID: 31861827 PMCID: PMC6982157 DOI: 10.3390/ijms21010065] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
The neuroendocrine and immune systems are coordinated to maintain the homeostasis of the organism, generating bidirectional communication through shared mediators and receptors. Vasoactive intestinal peptide (VIP) is the paradigm of an endogenous neuropeptide produced by neurons and endocrine and immune cells, involved in the control of both innate and adaptive immune responses. Exogenous administration of VIP exerts therapeutic effects in models of autoimmune/inflammatory diseases mediated by G-protein-coupled receptors (VPAC1 and VPAC2). Currently, there are no curative therapies for inflammatory and autoimmune diseases, and patients present complex diagnostic, therapeutic, and prognostic problems in daily clinical practice due to their heterogeneous nature. This review focuses on the biology of VIP and VIP receptor signaling, as well as its protective effects as an immunomodulatory factor. Recent progress in improving the stability, selectivity, and effectiveness of VIP/receptors analogues and new routes of administration are highlighted, as well as important advances in their use as biomarkers, contributing to their potential application in precision medicine. On the 50th anniversary of VIP's discovery, this review presents a spectrum of potential clinical benefits applied to inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Carmen Martínez
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Yasmina Juarranz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Irene Gutiérrez-Cañas
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Mar Carrión
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Selene Pérez-García
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Raúl Villanueva-Romero
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - David Castro
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Amalia Lamana
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| | - Mario Mellado
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología (CNB)/CSIC, 28049 Madrid, Spain;
| | - Isidoro González-Álvaro
- Servicio de Reumatología, Instituto de Investigación Médica, Hospital Universitario La Princesa, 28006 Madrid, Spain;
| | - Rosa P. Gomariz
- Departamento de Biología Celular, Facultad de Biología y Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain; (Y.J.); (I.G.-C.); (M.C.); (S.P.-G.); (R.V.-R.); (D.C.); (A.L.); (R.P.G.)
| |
Collapse
|
15
|
McDougall JJ. Osteoarthritis is a neurological disease – an hypothesis. OSTEOARTHRITIS AND CARTILAGE OPEN 2019; 1:100005. [DOI: 10.1016/j.ocarto.2019.100005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/17/2019] [Indexed: 12/20/2022] Open
|
16
|
Contribution of Infrapatellar Fat Pad and Synovial Membrane to Knee Osteoarthritis Pain. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6390182. [PMID: 31049352 PMCID: PMC6462341 DOI: 10.1155/2019/6390182] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is the most common form of joint disease and a major cause of pain and disability in the adult population. Interestingly, there are patients with symptomatic OA displaying pain, while patients with asymptomatic OA that do not experience pain but show radiographic signs of joint damage. Pain is a complex experience integrating sensory, affective, and cognitive processes related to several peripheral and central nociceptive factors besides inflammation. During the last years, the role of infrapatellar fat pad (IFP), other than the synovial membrane, has been investigated as a potential source of pain in OA. Interestingly, new findings suggest that IFP and synovial membrane might act as a functional unit in OA pathogenesis and pain. The present review discuss the role of IFP and synovial membrane in the development of OA, with a particular focus on pain onset and the possible involved mediators that may play a role in OA pathology and pain mechanisms. Inflammation of IFP and synovial membrane may drive peripheral and central sensitization in KOA. Since sensitization is associated with pain severity in knee OA and may potentially contribute to the transition from acute to chronic, persistent pain in knee OA, preventing sensitization would be a potentially effective and novel means of preventing worsening of pain in knee OA.
Collapse
|
17
|
Pujol R, Girard CA, Richard H, Hassanpour I, Binette MP, Beauchamp G, McDougall JJ, Laverty S. Synovial nerve fiber density decreases with naturally-occurring osteoarthritis in horses. Osteoarthritis Cartilage 2018; 26:1379-1388. [PMID: 29958917 DOI: 10.1016/j.joca.2018.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 05/10/2018] [Accepted: 06/07/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To measure the nerve fiber density in synovial membranes from healthy and OA equine joints and to investigate the relationship between synovial innervation and OA severity, synovial vascularity and synovitis. DESIGN Twenty-five equine metacarpophalangeal joints were collected post-mortem. The joints were dissected and the macroscopic lesions of the articular cartilage were scored. Synovial membrane specimens (n = 50) were harvested, fixed, sectioned and scored histologically. Immunohistochemical staining and immunofluorescence with S-100 protein, that identifies nerve fibers, and ⍺-actin, that stains vascular smooth muscle, were also performed on site-matched specimens and the relationships between these tissues was interrogated. RESULTS The nerve fiber density was higher in the superficial layer (≤200 μm) of the synovium when compared to the deeper layer in control equine joints (mean difference (95% C.I.): 0.054% (0.018%, 0.11%)). In osteoarthritic joints, synovial innervation decreased in the superficial layer with increasing macroscopic OA score (β (SEM), 95% C.I.: -0.0061 (0.00021), -0.0011, -0.00017). The blood vessel density was also higher in the superficial layer of the synovium compared to the deep layer in the control (mean difference (95% C.I.): 1.1% (0.36%, 2.3%)) and OA (mean difference (95% C.I.): 0.60% (0.22%, 1.2%)) equine joints. Moreover, considering all synovial specimens, higher nerve fiber density in the deep layer positively correlated with blood vessel density (β (SEM), 95% C.I.: 0.11 (0.036), 0.035, 0.18). CONCLUSION The reduction in nerve fiber density with advanced cartilage degeneration suggests that peripheral neuropathy is associated with equine OA. Whether this link is associated with neuropathic pain, requires further investigation.
Collapse
Affiliation(s)
- R Pujol
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada
| | - C A Girard
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada
| | - H Richard
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada
| | - I Hassanpour
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada
| | - M P Binette
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada
| | - G Beauchamp
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada
| | - J J McDougall
- Department of Pharmacology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia, B3H 4R2, Canada; Department of Anesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, Nova Scotia, B3H 4R2, Canada
| | - S Laverty
- Comparative Orthopedic Research Laboratory, Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 Sicotte, Saint-Hyacinthe, Québec, J2S 7C6, Canada.
| |
Collapse
|
18
|
Attenuation of early phase inflammation by cannabidiol prevents pain and nerve damage in rat osteoarthritis. Pain 2018; 158:2442-2451. [PMID: 28885454 PMCID: PMC5690292 DOI: 10.1097/j.pain.0000000000001052] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Osteoarthritis (OA) is a multifactorial joint disease, which includes joint degeneration, intermittent inflammation, and peripheral neuropathy. Cannabidiol (CBD) is a noneuphoria producing constituent of cannabis that has the potential to relieve pain. The aim of this study was to determine whether CBD is anti-nociceptive in OA, and whether inhibition of inflammation by CBD could prevent the development of OA pain and joint neuropathy. Osteoarthritis was induced in male Wistar rats (150-175 g) by intra-articular injection of sodium monoiodoacetate (MIA; 3 mg). On day 14 (end-stage OA), joint afferent mechanosensitivity was assessed using in vivo electrophysiology, whereas pain behaviour was measured by von Frey hair algesiometry and dynamic incapacitance. To investigate acute joint inflammation, blood flow and leukocyte trafficking were measured on day 1 after MIA. Joint nerve myelination was calculated by G-ratio analysis. The therapeutic and prophylactic effects of peripheral CBD (100-300 μg) were assessed. In end-stage OA, CBD dose-dependently decreased joint afferent firing rate, and increased withdrawal threshold and weight bearing (P < 0.0001; n = 8). Acute, transient joint inflammation was reduced by local CBD treatment (P < 0.0001; n = 6). Prophylactic administration of CBD prevented the development of MIA-induced joint pain at later time points (P < 0.0001; n = 8), and was also found to be neuroprotective (P < 0.05; n = 6-8). The data presented here indicate that local administration of CBD blocked OA pain. Prophylactic CBD treatment prevented the later development of pain and nerve damage in these OA joints. These findings suggest that CBD may be a safe, useful therapeutic for treating OA joint neuropathic pain.
Collapse
|
19
|
Kanaguchi Arita A, Yonemitsu I, Ikeda Y, Miyazaki M, Ono T. Low-intensity pulsed ultrasound stimulation for mandibular condyle osteoarthritis lesions in rats. Oral Dis 2017; 24:600-610. [DOI: 10.1111/odi.12798] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/26/2017] [Accepted: 10/18/2017] [Indexed: 11/30/2022]
Affiliation(s)
- A Kanaguchi Arita
- Department of Orthodontic Science; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - I Yonemitsu
- Department of Orthodontic Science; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Y Ikeda
- Department of Orthodontic Science; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - M Miyazaki
- Department of Orthodontic Science; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - T Ono
- Department of Orthodontic Science; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| |
Collapse
|
20
|
Krustev E, Muley MM, McDougall JJ. Endocannabinoids inhibit neurogenic inflammation in murine joints by a non-canonical cannabinoid receptor mechanism. Neuropeptides 2017; 64:131-135. [PMID: 27567396 DOI: 10.1016/j.npep.2016.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/15/2016] [Accepted: 08/21/2016] [Indexed: 10/21/2022]
Abstract
Neurogenic inflammation is a local inflammatory response that is driven by the peripheral release of neuropeptides from small diameter afferents which occurs in many organs including joints. The knee joint has a rich endocannabinoid system which has been shown to decrease acute synovitis. The aim of this study was to investigate the influence of joint afferents on leukocyte-endothelial interactions within the synovial microcirculation of mice and determine the role of endocannabinoids on this inflammatory response. Electrical, antidromic stimulation of the saphenous nerve decreased leukocyte rolling at the lowest frequency tested (0.5Hz), while increasing leukocyte rolling at higher frequencies (2.0 and 5.0Hz). The leukocyte rolling effect of nerve stimulation was completely abolished by pre-treating the knee with the vasoactive intestinal peptide antagonist VIP6-28; however, neither calcitonin gene related peptide nor substance P antagonism had an effect on this neurogenic inflammatory response. Treating knees with the endocannabinoid breakdown inhibitor URB597 completely blocked leukocyte rolling and this effect could be reversed with the non-canonical cannabinoid antagonist O-1918. These results provide evidence that antidromic stimulation of the mouse saphenous nerve promotes leukocyte rolling within the synovial microcirculation, and that endocannabinoids can attenuate this neurogenic inflammatory response.
Collapse
Affiliation(s)
- Eugene Krustev
- Department of Pharmacology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada; Department of Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada.
| | - Milind M Muley
- Department of Pharmacology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada; Department of Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada.
| | - Jason J McDougall
- Department of Pharmacology, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada; Department of Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
21
|
Liu J, Hao Y, Wang Y, Hu S, Xu K, Lu C. Candidate methylated genes in osteoarthritis explored by bioinformatics analysis. Knee 2016; 23:1035-1043. [PMID: 27810435 DOI: 10.1016/j.knee.2016.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/01/2016] [Accepted: 09/20/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND This study aimed to explore potential novel genes correlated with osteoarthritis (OA). METHODS The gene expression profile of GSE48422 was downloaded from the Gene Expression Omnibus (GEO) database. This dataset included five arthritic cartilage samples and five non-arthritic cartilage samples from five female OA patients. Differentially methylated genes (DMGs) between the two kinds of samples were identified, followed by their functional analysis and protein-protein interaction (PPI) analysis. Furthermore, the Comparative Toxicogenomics Database (CTD) was used to further identify OA-related genes among these DMGs. RESULTS In total, 965 hypermethylated genes and 112 hypomethylated genes were identified in the arthritic cartilage samples. The hypermethylated genes (e.g., ADCY4 and ADCY6) were significantly related to the calcium signaling pathway and gonadotropin-releasing hormone signaling pathway, while the hypomethylated genes were implicated in the mammalian target of rapamycin signaling pathway. In the PPI network, several genes had a higher degree, such as ADCY4, ADCY6 and GPR17, and they interacted with each other. Additionally, 565 DMGs were predicted to be associated with OA, and five of them (e.g., COMP and EDIL3) were previously identified as OA markers. CONCLUSIONS The methylation of genes ADCY4, ADCY6 and GPR17, as well as the gonadotropin-releasing hormone signaling pathway, was newly found to be potentially associated with OA. They may be novel OA markers.
Collapse
Affiliation(s)
- Jie Liu
- Shaanxi University of Chinese Medicine, Shiji Avenue, Xi'an-Xianyang New Economic Zone, Shaanxi 712046, PR China
| | - Yangquan Hao
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China.
| | - Yugui Wang
- Shaanxi University of Chinese Medicine, Shiji Avenue, Xi'an-Xianyang New Economic Zone, Shaanxi 712046, PR China
| | - Shouye Hu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China
| | - Ke Xu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China
| | - Chao Lu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital, Xi'an Jiao Tong University Health Science Center, 555 Youyi East Road, Xi'an, Shaanxi 710068, PR China
| |
Collapse
|
22
|
Kirkpatrick DR, McEntire DM, Smith TA, Dueck NP, Kerfeld MJ, Hambsch ZJ, Nelson TJ, Reisbig MD, Agrawal DK. Transmission pathways and mediators as the basis for clinical pharmacology of pain. Expert Rev Clin Pharmacol 2016; 9:1363-1387. [PMID: 27322358 PMCID: PMC5215101 DOI: 10.1080/17512433.2016.1204231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Mediators in pain transmission are the targets of a multitude of different analgesic pharmaceuticals. This review explores the most significant mediators of pain transmission as well as the pharmaceuticals that act on them. Areas covered: The review explores many of the key mediators of pain transmission. In doing so, this review uncovers important areas for further research. It also highlights agents with potential for producing novel analgesics, probes important interactions between pain transmission pathways that could contribute to synergistic analgesia, and emphasizes transmission factors that participate in transforming acute injury into chronic pain. Expert commentary: This review examines current pain research, particularly in the context of identifying novel analgesics, highlighting interactions between analgesic transmission pathways, and discussing factors that may contribute to the development of chronic pain after an acute injury.
Collapse
Affiliation(s)
- Daniel R. Kirkpatrick
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Dan M. McEntire
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Tyler A. Smith
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Nicholas P. Dueck
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Mitchell J. Kerfeld
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Zakary J. Hambsch
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Taylor J. Nelson
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Mark D. Reisbig
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| | - Devendra K. Agrawal
- Departments of Clinical and Translational Science and Anesthesiology, Creighton University School of Medicine, Omaha, NE 68178 USA
| |
Collapse
|
23
|
Jiang W, Wang H, Li YS, Luo W. Role of vasoactive intestinal peptide in osteoarthritis. J Biomed Sci 2016; 23:63. [PMID: 27553659 PMCID: PMC4995623 DOI: 10.1186/s12929-016-0280-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/09/2016] [Indexed: 02/06/2023] Open
Abstract
Vasoactive intestinal peptide (VIP) plays important roles in many biological functions, such as, stimulation of contractility in the heart, vasodilation, promoting neuroendocrine-immune communication, lowering arterial blood pressure, and anti-inflammatory and immune-modulatory activity. Osteoarthritis (OA) is a chronic and degenerative bone disease, which is one of the most common causes of disability and most common in both sexes as people become older. Interestingly VIP can prevent chronic cartilage damage and joint remodeling. This review article provides update information on the association of VIP and OA and its treatment. Evidences suggest that VIP is down-regulated in synovial fluid of OA, and VIP down-regulation leads to increase in the production of pro-inflammatory cytokines that might contribute to the pathogenesis of OA; however contradictory reports also exist suggesting that accumulation of VIP in joints can also contribute OA. A number of studies indicated that up-regulation of VIP can counteract the action of pro-inflammatory stimuli and alleviate the pain in OA. More clinical investigations are necessary to determine the biology of VIP and its therapeutic potential in OA that might represent the future standards of care for OA.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Bone and Joint, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, Guangdong, 518020, China
| | - Hua Wang
- Department of Bone and Joint, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, Guangdong, 518020, China
| | - Yu-Sheng Li
- Department of Orthopaedics, Xiang-ya Hospital, Central South University, Changsha, Hunan, 410078, China.
| | - Wei Luo
- Department of Orthopaedics, Xiang-ya Hospital, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
24
|
Otis C, Gervais J, Guillot M, Gervais JA, Gauvin D, Péthel C, Authier S, Dansereau MA, Sarret P, Martel-Pelletier J, Pelletier JP, Beaudry F, Troncy E. Concurrent validity of different functional and neuroproteomic pain assessment methods in the rat osteoarthritis monosodium iodoacetate (MIA) model. Arthritis Res Ther 2016; 18:150. [PMID: 27338815 PMCID: PMC4918125 DOI: 10.1186/s13075-016-1047-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/09/2016] [Indexed: 11/17/2022] Open
Abstract
Background Lack of validity in osteoarthritis pain models and assessment methods is suspected. Our goal was to 1) assess the repeatability and reproducibility of measurement and the influence of environment, and acclimatization, to different pain assessment outcomes in normal rats, and 2) test the concurrent validity of the most reliable methods in relation to the expression of different spinal neuropeptides in a chemical model of osteoarthritic pain. Methods Repeatability and inter-rater reliability of reflexive nociceptive mechanical thresholds, spontaneous static weight-bearing, treadmill, rotarod, and operant place escape/avoidance paradigm (PEAP) were assessed by the intraclass correlation coefficient (ICC). The most reliable acclimatization protocol was determined by comparing coefficients of variation. In a pilot comparative study, the sensitivity and responsiveness to treatment of the most reliable methods were tested in the monosodium iodoacetate (MIA) model over 21 days. Two MIA (2 mg) groups (including one lidocaine treatment group) and one sham group (0.9 % saline) received an intra-articular (50 μL) injection. Results No effect of environment (observer, inverted circadian cycle, or exercise) was observed; all tested methods except mechanical sensitivity (ICC <0.3), offered good repeatability (ICC ≥0.7). The most reliable acclimatization protocol included five assessments over two weeks. MIA-related osteoarthritic change in pain was demonstrated with static weight-bearing, punctate tactile allodynia evaluation, treadmill exercise and operant PEAP, the latter being the most responsive to analgesic intra-articular lidocaine. Substance P and calcitonin gene-related peptide were higher in MIA groups compared to naive (adjusted P (adj-P) = 0.016) or sham-treated (adj-P = 0.029) rats. Repeated post-MIA lidocaine injection resulted in 34 times lower downregulation for spinal substance P compared to MIA alone (adj-P = 0.029), with a concomitant increase of 17 % in time spent on the PEAP dark side (indicative of increased comfort). Conclusion This study of normal rats and rats with pain established the most reliable and sensitive pain assessment methods and an optimized acclimatization protocol. Operant PEAP testing was more responsive to lidocaine analgesia than other tests used, while neuropeptide spinal concentration is an objective quantification method attractive to support and validate different centralized pain functional assessment methods.
Collapse
Affiliation(s)
- Colombe Otis
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada.,Osteoarthritis Research Unit, Research Center Hospital of Montreal University (CRCHUM), Montreal, Quebec, Canada
| | - Julie Gervais
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada
| | - Martin Guillot
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada.,Osteoarthritis Research Unit, Research Center Hospital of Montreal University (CRCHUM), Montreal, Quebec, Canada
| | - Julie-Anne Gervais
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada
| | - Dominique Gauvin
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada.,Osteoarthritis Research Unit, Research Center Hospital of Montreal University (CRCHUM), Montreal, Quebec, Canada
| | - Catherine Péthel
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada.,Department of Physiology and Biophysics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - Marc-André Dansereau
- Department of Physiology and Biophysics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Philippe Sarret
- Department of Physiology and Biophysics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, Research Center Hospital of Montreal University (CRCHUM), Montreal, Quebec, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, Research Center Hospital of Montreal University (CRCHUM), Montreal, Quebec, Canada
| | - Francis Beaudry
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada.,Osteoarthritis Research Unit, Research Center Hospital of Montreal University (CRCHUM), Montreal, Quebec, Canada
| | - Eric Troncy
- Groupe de Recherche en Pharmacologie Animale du Québec (GREPAQ), Department of Biomedical Sciences, Faculty of veterinary medicine, Université de Montréal, 1500 des Vétérinaires Street, P.O. Box 5000, St-Hyacinthe, Quebec, J2S 7C6, Canada. .,Osteoarthritis Research Unit, Research Center Hospital of Montreal University (CRCHUM), Montreal, Quebec, Canada.
| |
Collapse
|
25
|
Rytel L, Całka J. Neuropeptide profile changes in sensory neurones after partial prepyloric resection in pigs. Ann Anat 2016; 206:48-56. [PMID: 27142347 DOI: 10.1016/j.aanat.2016.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/20/2022]
Abstract
This report details the first identification of the sources of sensory innervation of the porcine stomach prepyloric region. The Fast Blue (FB) retrograde tracing technique detected the sensory prepyloric neurons in the bilateral nodose ganglia (NGs) as well as thoracic dorsal root ganglia (DRGs). Double-labelling immunofluorescence demonstrated expression of substance P (SP), calcitonin gene-related peptide (CGRP), neuronal isoform of nitric oxide synthase (nNOS), vasoactive intestinal polypeptide (VIP) and galanin (GAL) in both NGs and DRGs. Additionally, we found that partial resection of the stomach prepyloric area increased expression of the SP, CGRP, NOS, VIP and GAL in the prepyloric sensory neurons. In the control left NGs, both a higher total number of FB-positive perikarya as well as a higher percentage of the peptides expressing prepyloric neurons were visualized than in the right NGs. However, compared to the control group, prepyloric resection evoked greater increases in peptide expression in the right-side NGs sensory neurons. In the ganglia of this side, the proportion of the SP-IR perikarya increased by approximately 15%, while CGRP-IR increased by 28%, NOS-IR 14%, VIP-IR 43% and GAL-IR 13%. On the opposite left side, the ganglia proportion of the CGRP-IR perikarya increased by approximately 10%, while NOS-IR increased by 3%, VIP-IR 36% and GAL-IR by 2%. The only decrease (by 5%) was observed in the case of SP expression. We also found that 92% of the sensory neurons originated from NGs and 8% from DRGs. Our results indicate that, in the pig, SP, CGRP, NOS, VIP and GAL participate in the vagal sensory transduction from the stomach prepyloric area. Moreover, increased expression of the peptides and neuronal isoform of nitric oxide synthase in the sensory neurons following transection of their peripheral dendrites suggests their possible participation in the neuronal recovery and/or reinnervation process.
Collapse
Affiliation(s)
- L Rytel
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland.
| | - J Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
26
|
Sághy É, Payrits M, Helyes Z, Reglődi D, Bánki E, Tóth G, Couvineau A, Szőke É. Stimulatory effect of pituitary adenylate cyclase-activating polypeptide 6-38, M65 and vasoactive intestinal polypeptide 6-28 on trigeminal sensory neurons. Neuroscience 2015; 308:144-156. [PMID: 26321242 DOI: 10.1016/j.neuroscience.2015.08.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/31/2015] [Accepted: 08/20/2015] [Indexed: 02/08/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) acts on G protein-coupled receptors: the specific PAC1 and VPAC1/VPAC2 receptors. PACAP6-38 was described as a potent PAC1/VPAC2 antagonist in several models, but recent studies reported its agonistic behaviors proposing novel receptorial mechanisms. Since PACAP in migraine is an important research tool, we investigated the effect of PACAP and its peptide fragments on trigeminal primary sensory neurons. Effect of the peptides was studied with ratiometric Ca-imaging technique using the fluorescent indicator fura-2 AM on primary cultures of rat and mouse trigeminal ganglia (TRGs) neurons. Specificity testing was performed on PAC1, VPAC1 and VPAC2 receptor-expressing cell lines with both fluorescent and radioactive Ca-uptake methods. Slowly increasing intracellular free calcium concentration [Ca(2+)]i was detected after PACAP1-38, PACAP1-27, vasoactive intestinal polypeptide (VIP) and the selective PAC1 receptor agonist maxadilan administration on TRG neurons, but interestingly, PACAP6-38, VIP6-28 and the PAC1 receptor antagonist M65 also caused similar activation. The VPAC2 receptor agonist BAY 55-9837 induced similar activation, while the VPAC1 receptor agonist Ala(11,22,28)VIP had no significant effect on [Ca(2+)]i. It was proven that the Ca(2+)-influx originated from intracellular stores using radioactive calcium-45 uptake experiment and Ca-free solution. On the specific receptor-expressing cell lines the antagonists inhibited the stimulating actions of the respective agonists, but had no effects by themselves. PACAP6-38, M65 and VIP6-28, which were described as antagonists in numerous studies in several model systems, act as agonists on TRG primary sensory neurons. Currently unknown receptors or splice variants linked to distinct signal transduction pathways might explain these differences.
Collapse
MESH Headings
- Animals
- CHO Cells
- Calcium/metabolism
- Cells, Cultured
- Cricetulus
- Humans
- Insect Proteins/pharmacology
- Mice
- Peptide Fragments/pharmacology
- Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology
- Rats, Wistar
- Receptors, Vasoactive Intestinal Peptide, Type II/antagonists & inhibitors
- Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
- Receptors, Vasoactive Intestinal Polypeptide, Type I/agonists
- Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism
- Sensory Receptor Cells/drug effects
- Sensory Receptor Cells/physiology
- Sensory System Agents/pharmacology
- TRPV Cation Channels/metabolism
- Trigeminal Ganglion/drug effects
- Trigeminal Ganglion/physiology
- Vasoactive Intestinal Peptide/pharmacology
- Voltage-Sensitive Dye Imaging
Collapse
Affiliation(s)
- É Sághy
- Department of Pharmacology and Pharmacotherapy, MTA-PTE Chronic Pain Research Group, Szentágothai Research Center, University of Pécs, Pécs-7624, Szigeti Street 12, Hungary.
| | - M Payrits
- Department of Pharmacology and Pharmacotherapy, MTA-PTE Chronic Pain Research Group, Szentágothai Research Center, University of Pécs, Pécs-7624, Szigeti Street 12, Hungary.
| | - Zs Helyes
- Department of Pharmacology and Pharmacotherapy, MTA-PTE Chronic Pain Research Group, Szentágothai Research Center, University of Pécs, Pécs-7624, Szigeti Street 12, Hungary.
| | - D Reglődi
- Department of Anatomy, MTA-PTE "Lendület" PACAP Research Team, University of Pécs, Pécs-7624, Szigeti Street 12, Hungary.
| | - E Bánki
- Department of Anatomy, MTA-PTE "Lendület" PACAP Research Team, University of Pécs, Pécs-7624, Szigeti Street 12, Hungary.
| | - G Tóth
- Department of Medical Chemistry, University of Szeged, Szeged-6720, Dugonics Street 13, Hungary.
| | - A Couvineau
- UMR 1149 INSERM/Centre de Recherche sur l'Inflammation, Université Paris Diderot, Faculte de Medecine Paris 7 - Site Bichat, 16 Rue Henri Huchard, 75890 Paris Cedex 18, France.
| | - É Szőke
- Department of Pharmacology and Pharmacotherapy, MTA-PTE Chronic Pain Research Group, Szentágothai Research Center, University of Pécs, Pécs-7624, Szigeti Street 12, Hungary.
| |
Collapse
|
27
|
Thysen S, Luyten FP, Lories RJU. Targets, models and challenges in osteoarthritis research. Dis Model Mech 2015; 8:17-30. [PMID: 25561745 PMCID: PMC4283647 DOI: 10.1242/dmm.016881] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis is a chronic degenerative disorder of the joint and represents one of the most common diseases worldwide. Its prevalence and severity are increasing owing to aging of the population, but treatment options remain largely limited to painkillers and anti-inflammatory drugs, which only provide symptomatic relief. In the late stages of the disease, surgical interventions are often necessary to partially restore joint function. Although the focus of osteoarthritis research has been originally on the articular cartilage, novel findings are now pointing to osteoarthritis as a disease of the whole joint, in which failure of different joint components can occur. In this Review, we summarize recent progress in the field, including data from novel ‘omics’ technologies and from a number of preclinical and clinical trials. We describe different in vitro and in vivo systems that can be used to study molecules, pathways and cells that are involved in osteoarthritis. We illustrate that a comprehensive and multisystem approach is necessary to understand the complexity and heterogeneity of the disease and to better guide the development of novel therapeutic strategies for osteoarthritis.
Collapse
Affiliation(s)
- Sarah Thysen
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Frank P Luyten
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000 Leuven, Belgium. Division of Rheumatology, University Hospitals Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Rik J U Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, 3000 Leuven, Belgium. Division of Rheumatology, University Hospitals Leuven, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
28
|
Rahman W, Patel R, Dickenson AH. Electrophysiological evidence for voltage-gated calcium channel 2 (Cav2) modulation of mechano- and thermosensitive spinal neuronal responses in a rat model of osteoarthritis. Neuroscience 2015; 305:76-85. [PMID: 26247695 PMCID: PMC4564012 DOI: 10.1016/j.neuroscience.2015.07.073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/06/2015] [Accepted: 07/28/2015] [Indexed: 12/31/2022]
Abstract
MIA-dependent antinociceptive effect of TROX-1 on neuronal activity. Alterations in Cav2.2 channel function contribute to osteoarthritic (OA) pain. Blocking Cav2.2 channels has therapeutic potential for treating OA pain.
Osteoarthritis (OA) remains one of the greatest healthcare burdens in western society, with chronic debilitating pain-dominating clinical presentation yet therapeutic strategies are inadequate in many patients. Development of better analgesics is contingent on improved understanding of the molecular mechanisms mediating OA pain. Voltage-gated calcium channels 2.2 (Cav2.2) play a critical role in spinal nociceptive transmission, therefore blocking Cav2.2 activity represents an attractive opportunity for OA pain treatment, but the only available licensed Cav2.2 antagonist ziconitide (PrilatTM) is of limited use. TROX-1 is an orally available, use dependent and state-selective Cav2 antagonist, exerting its analgesic effect primarily via Cav2.2 blockade, with an improved therapeutic window compared with ziconitide. Using a rat model of monosodium iodoacetate (MIA), 2 mg, induced OA we used in vivo electrophysiology to assess the effects of spinal or systemic administration of TROX-1 on the evoked activity of wide dynamic range spinal dorsal horn neurons in response to electrical, natural mechanical (dynamic brush and von Frey 2, 8, 26 and 6 g) and thermal (40, 45 and 45 °C) stimuli applied to the peripheral receptive field. MIA injection into the knee joint resulted in mechanical hypersensitivity of the ipsilateral hind paw and weight-bearing asymmetry. Spinal administration of TROX-1 (0.1 and 1 μg/50 μl) produced a significant dose-related inhibition of dynamic brush, mechanical (von Frey filament (vF) 8, 26 and 60 g) and noxious thermal-(45 and 48 °C) evoked neuronal responses in MIA rats only. Systemic administration of TROX-1 produced a significant inhibition of the mechanical-(vF 8, 26 and 60 g) evoked neuronal responses in MIA rats. TROX-1 did not produce any significant effect on any neuronal measure in Sham controls. Our in vivo electrophysiological results demonstrate a pathological state-dependent effect of TROX-1, which suggests an increased functional role of Cav2, likely Cav2.2, channels in mediating OA pain.
Collapse
Affiliation(s)
- W Rahman
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| | - R Patel
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - A H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
29
|
Abstract
There are over 100 different types of arthritis and each can differ greatly in their aetiology and pathophysiology; however, one characteristic that is common to all arthritic conditions is joint pain. Musculoskeletal pain is the leading cause of disability in the world, and the number one reason arthritis patients visit their primary care physician. Despite the prevalence and burden of arthritis pain, current analgesics lack sufficient efficacy and are plagued by multiple adverse side effects. In this review, we outline the current landscape of research concerning joint pain, drawing from both preclinical and clinical studies. Specifically, this review is a discussion of the different neurophysiological processes that occur during joint disease and how inflammatory and neuropathic aspects contribute to the development of arthritis pain.
Collapse
Affiliation(s)
- Eugene Krustev
- Department of Pharmacology, Dalhousie University, 5850 College Street, Halifax, NS, B3H 4R2, Canada,
| | | | | |
Collapse
|
30
|
Emerging targets and therapeutic approaches for the treatment of osteoarthritis pain. Curr Opin Support Palliat Care 2015; 9:124-30. [DOI: 10.1097/spc.0000000000000125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Rahman W, Dickenson AH. Osteoarthritis-dependent changes in antinociceptive action of Nav1.7 and Nav1.8 sodium channel blockers: An in vivo electrophysiological study in the rat. Neuroscience 2015; 295:103-16. [PMID: 25818052 PMCID: PMC4414363 DOI: 10.1016/j.neuroscience.2015.03.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/16/2015] [Accepted: 03/19/2015] [Indexed: 02/06/2023]
Abstract
MIA-dependent antinociceptive effect of ProTxII and A-803467 on neuronal activity. Changes in Nav1.7 and 1.8 channel function contribute to osteoarthritic pain. Blocking Nav1.7 and Nav1.8 channels has therapeutic potential for the treatment of osteoarthritic pain.
Voltage-gated sodium channel blockers are not traditionally recommended for osteoarthritis (OA) pain therapy, but given the large peripheral drive that follows OA development there is a rationale for their use. Using a rat model of monosodium iodoacetate (MIA)-induced OA we used in vivo electrophysiology to assess the effects of the Nav1.7- and Nav1.8-selective antagonists, ProTxII and A-803467 respectively, on the evoked activity of spinal dorsal horn neurons in response to electrical, mechanical and thermal stimuli applied to the peripheral receptive field. These studies allow examination of the roles of these channels in suprathreshold stimuli, not amenable to behavioral threshold measures. Spinal administration of ProTxII significantly reduced neuronal responses evoked by mechanical punctate (von Frey (vF) 8–60 g) and noxious thermal (45 and 48 °C) stimuli in MIA rats only. A-803467 significantly inhibited neuronal responses evoked by vF 8–60 g and 48 °C heat after spinal administration; significantly inhibited responses evoked by brush, vFs 26–60 g and 40–48 °C stimuli after systemic administration; significantly inhibited the electrically evoked Aδ-, C-fiber, post-discharge, Input and wind-up responses and the brush, vFs 8–60 g and 45–48 °C evoked neuronal responses after intra plantar injection in the MIA group. In comparison A-803467 effects in the sham group were minimal and included a reduction of the neuronal response evoked by vF 60 g and 45 °C heat stimulation after spinal administration, no effect after systemic administration and an inhibition of the evoked response to 45 °C heat after intra plantar injection only. The observed selective inhibitory effect of ProTxII and A-803467 for the MIA-treated group suggests an increased role of Nav1.7 and 1.8 within nociceptive pathways in the arthritic condition, located at peripheral and central sites. These findings demonstrate the importance of, and add to, the mechanistic understanding of these channels in osteoarthritic pain.
Collapse
Affiliation(s)
- W Rahman
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| | - A H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
32
|
Kelly S, Chapman RJ, Woodhams S, Sagar DR, Turner J, Burston JJ, Bullock C, Paton K, Huang J, Wong A, McWilliams DF, Okine BN, Barrett DA, Hathway GJ, Walsh DA, Chapman V. Increased function of pronociceptive TRPV1 at the level of the joint in a rat model of osteoarthritis pain. Ann Rheum Dis 2015; 74:252-9. [PMID: 24152419 PMCID: PMC4283626 DOI: 10.1136/annrheumdis-2013-203413] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 08/13/2013] [Accepted: 09/20/2013] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Blockade of transient receptor potential vanilloid 1 (TRPV1) with systemic antagonists attenuates osteoarthritis (OA) pain behaviour in rat models, but on-target-mediated hyperthermia has halted clinical trials. The present study investigated the potential for targeting TRPV1 receptors within the OA joint in order to produce analgesia. METHODS The presence of TRPV1 receptors in human synovium was detected using western blotting and immunohistochemistry. In a rat model of OA, joint levels of an endogenous ligand for TRPV1, 12-hydroxy-eicosatetraenoic acid (12-HETE), were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Effects of peripheral administration of the TRPV1 receptor antagonist JNJ-17203212 on afferent fibre activity, pain behaviour and core body temperature were investigated. Effects of a spinal administration of JNJ-17203212 on dorsal horn neuronal responses were studied. RESULTS We demonstrate increased TRPV1 immunoreactivity in human OA synovium, confirming the diseased joint as a potential therapeutic target for TRPV1-mediated analgesia. In a model of OA pain, we report increased joint levels of 12-HETE, and the sensitisation of joint afferent neurones to mechanical stimulation of the knee. Local administration of JNJ-17203212 reversed this sensitisation of joint afferents and inhibited pain behaviour (weight-bearing asymmetry), to a comparable extent as systemic JNJ-17203212, in this model of OA pain, but did not alter core body temperature. There was no evidence for increased TRPV1 function in the spinal cord in this model of OA pain. CONCLUSIONS Our data provide a clinical and mechanistic rationale for the future investigation of the therapeutic benefits of intra-articular administration of TRPV1 antagonists for the treatment of OA pain.
Collapse
Affiliation(s)
- S Kelly
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire, UK
| | - R J Chapman
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire, UK
| | - S Woodhams
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - D R Sagar
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - J Turner
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - J J Burston
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - C Bullock
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire, UK
| | - K Paton
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire, UK
| | - J Huang
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - A Wong
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - D F McWilliams
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK Division of Academic Rheumatology, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - B N Okine
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - D A Barrett
- Centre for Analytical Bioscience, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - G J Hathway
- School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| | - D A Walsh
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK Division of Academic Rheumatology, University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - V Chapman
- Arthritis Research UK Pain Centre, University of Nottingham, Nottingham, UK School of Biomedical Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
33
|
Rahman W, Dickenson AH. Antinociceptive effects of lacosamide on spinal neuronal and behavioural measures of pain in a rat model of osteoarthritis. Arthritis Res Ther 2014; 16:509. [PMID: 25533381 PMCID: PMC4308925 DOI: 10.1186/s13075-014-0509-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 12/09/2014] [Indexed: 01/27/2023] Open
Abstract
Introduction Alterations in voltage-gated sodium channel (VGSC) function have been linked to chronic pain and are good targets for analgesics. Lacosamide (LCM) is a novel anticonvulsant that enhances the slow inactivation state of VGSCs. This conformational state can be induced by repeated neuronal firing and/or under conditions of sustained membrane depolarisation, as is expected for hyperexcitable neurones in pathological conditions such as epilepsy and neuropathy, and probably osteoarthritis (OA). In this study, therefore, we examined the antinociceptive effect of LCM on spinal neuronal and behavioural measures of pain, in vivo, in a rat OA model. Methods OA was induced in Sprague Dawley rats by intraarticular injection of 2 mg of monosodium iodoacetate (MIA). Sham rats received saline injections. Behavioural responses to mechanical and cooling stimulation of the ipsilateral hind paw and hindlimb weight-bearing were recorded. In vivo electrophysiology experiments were performed in anaesthetised MIA or sham rats, and we recorded the effects of spinal or systemic administration of LCM on the evoked responses of dorsal horn neurones to electrical, mechanical (brush, von Frey, 2 to 60 g) and heat (40°C to 50°C) stimulation of the peripheral receptive field. The effect of systemic LCM on nociceptive behaviours was assessed. Results Behavioural hypersensitivity ipsilateral to knee injury was seen as a reduced paw withdrawal threshold to mechanical stimulation, an increase in paw withdrawal frequency to cooling stimulation and hind limb weight-bearing asymmetry in MIA-treated rats only. Spinal and systemic administration of LCM produced significant reductions of the electrical Aβ- and C-fibre evoked neuronal responses and the mechanical and thermal evoked neuronal responses in the MIA group only. Systemic administration of LCM significantly reversed the behavioural hypersensitive responses to mechanical and cooling stimulation of the ipsilateral hind paw, but hind limb weight-bearing asymmetry was not corrected. Conclusions Our in vivo electrophysiological results show that the inhibitory effects of LCM were MIA-dependent. This suggests that, if used in OA patients, LCM may allow physiological transmission but suppress secondary hyperalgesia and allodynia. The inhibitory effect on spinal neuronal firing aligned with analgesic efficacy on nociceptive behaviours and suggests that LCM may still prove worthwhile for OA pain treatment and merits further clinical investigation.
Collapse
|
34
|
Krustev E, Reid A, McDougall JJ. Tapping into the endocannabinoid system to ameliorate acute inflammatory flares and associated pain in mouse knee joints. Arthritis Res Ther 2014; 16:437. [PMID: 25260980 PMCID: PMC4201700 DOI: 10.1186/s13075-014-0437-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 08/26/2014] [Indexed: 01/01/2023] Open
Abstract
Introduction During the progression of rheumatoid arthritis (RA), there are frequent but intermittent flares in which the joint becomes acutely inflamed and painful. Although a number of drug therapies are currently used to treat RA, their effectiveness is variable and side effects are common. Endocannabinoids have the potential to ameliorate joint pain and inflammation, but these beneficial effects are limited by their rapid degradation. One enzyme responsible for endocannabinoid breakdown is fatty acid amide hydrolase (FAAH). The present study examined whether URB597, a potent and selective FAAH inhibitor, could alter inflammation and pain in a mouse model of acute synovitis. Methods Acute joint inflammation was induced in male C57BL/6 mice by intra-articular injection of 2% kaolin/2% carrageenan. After 24 hr, articular leukocyte kinetics and blood flow were used as measures of inflammation, while hindlimb weight bearing and von Frey hair algesiometry were used as measures of joint pain. The effects of local URB597 administration were then determined in the presence or absence of either the cannabinoid (CB)1 receptor antagonist AM251, or the CB2 receptor antagonist AM630. Results URB597 decreased leukocyte rolling and adhesion, as well as inflammation-induced hyperaemia. However, these effects were only apparent at low doses and the effects of URB597 were absent at higher doses. In addition to the anti-inflammatory effects of URB597, fatty acid amide hydrolase (FAAH) inhibition improved both hindlimb weight bearing and von Frey hair withdrawal thresholds. The anti-inflammatory effects of URB597 on leukocyte rolling and vascular perfusion were blocked by both CB1 and CB2 antagonism, while the effect on leukocyte adherence was independent of cannabinoid receptor activation. The analgesic effects of URB597 were CB1 mediated. Conclusions These results suggest that the endocannabinoid system of the joint can be harnessed to decrease acute inflammatory reactions and the concomitant pain associated with these episodes.
Collapse
|
35
|
Rytel L, Palus K, Całka J. Co-expression of PACAP with VIP, SP and CGRP in the porcine nodose ganglion sensory neurons. Anat Histol Embryol 2014; 44:86-91. [PMID: 24713103 DOI: 10.1111/ahe.12111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/16/2014] [Indexed: 11/28/2022]
Abstract
Our previous study revealed the expression of substance P (SP) and calcitonin gene-related peptide (CGRP) in sensory distal ganglion of the vagus (nodose ganglion) neurons in the pig. As these neuropeptides may be involved in nociception, the goal of these investigations was to determine possible expression of vasoactive intestinal polypeptide (VIP), SP and CGRP in the pituitary adenylate cyclase-activating polypeptide-immunoreactive (PACAP-IR) porcine nodose perikarya. Co-expression of these substances was examined using a double-labelling immunofluorescence technique. To reveal the ganglionic cell bodies, the pan-neuronal marker protein gene product 9.5 (PGP 9.5) was used. Quantitative analysis of the neurons revealed that 67.25% of the PGP 9.5+ somata in the right-side ganglion and 66.5% in the left side, respectively, co-expressed PACAP-IR. Moreover, 60.6% of the PACAP-IR cells in the right-side ganglion and 62.1% in the left, respectively, co-expressed VIP. SP-IR was observed in 52.2 and 39.9% of the right and left ganglia, respectively. CGRP was found in 27.7 and 34.1% of the right and left distal ganglion of the vagus, respectively. High level of co-expression of PACAP with VIP, SP and CGRP in the distal ganglia of the vagus sensory perikarya directly implicates studied peptides in their functional interaction during nociceptive vagal transduction.
Collapse
Affiliation(s)
- L Rytel
- Division of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | | | |
Collapse
|
36
|
Al-Jefout M, Tokushige N, Hey-Cunningham AJ, Manconi F, Ng C, Schulke L, Berbic M, Markham R, Fraser IS. Microanatomy and function of the eutopic endometrium in women with endometriosis. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17474108.4.1.61] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
37
|
Zhang RX, Ren K, Dubner R. Osteoarthritis pain mechanisms: basic studies in animal models. Osteoarthritis Cartilage 2013; 21:1308-15. [PMID: 23973145 PMCID: PMC3771690 DOI: 10.1016/j.joca.2013.06.013] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a complex and painful disease of the whole joint. At present there are no satisfying agents for treating OA. To promote OA research and improved treatment, this review summarizes current preclinical evidence on the development of OA. METHODS Preclinical OA research was searched and key findings are summarized and commented. RESULTS Mechanisms of OA-associated pain have been studied in rodent knee OA models produced by intra-knee injection of the chondrocyte glycolytic inhibitor mono-iodoacetate (MIA), surgery, or spontaneous development in some species. These models are clinically relevant in terms of histological damage and functional changes, and are used to study mechanisms underlying mechanical, thermal, ambulatory, body weight supporting-evoked, and ongoing OA pain. Recent peripheral, spinal, and supraspinal biochemical and electrophysiological studies in these models suggest that peripheral pro-inflammatory mediators and neuropeptides sensitize knee nociceptors. Spinal cytokines and neuropeptides promote OA pain, and peripheral and spinal cannabinoids inhibit OA pain respectively through cannabinoid-1 (CB1) and CB1/CB2 receptors. TRPV1 and metalloproteinases contribute and supraspinal descending facilitation of 5-hydroxytryptamine (5-HT)/5-HT 3 receptors may also contribute to OA pain. Conditioned place preference tests demonstrate that OA pain induces aversive behaviors, suggesting the involvement of brain. During OA, brain functional connectivity is enhanced, but at present it is unclear how this change is related to OA pain. CONCLUSION Animal studies demonstrate that peripheral and central sensitization contributes to OA pain, involving inflammatory cytokines, neuropeptides, and a variety of chemical mediators. Interestingly, brainstem descending facilitation of 5-HT/5-HT3 receptors plays a role OA pain.
Collapse
Affiliation(s)
- Rui-Xin Zhang
- Center for Integrative Medicine, School of Medicine, University of Maryland, Baltimore, MD 21201 USA
| | - Ke Ren
- Department of Neural and Pain Sciences, Dental School, University of Maryland, Baltimore, MD 21201 USA
| | - Ronald Dubner
- Department of Neural and Pain Sciences, Dental School, University of Maryland, Baltimore, MD 21201 USA
| |
Collapse
|
38
|
Malfait AM, Little CB, McDougall JJ. A commentary on modelling osteoarthritis pain in small animals. Osteoarthritis Cartilage 2013; 21:1316-26. [PMID: 23973146 PMCID: PMC3903124 DOI: 10.1016/j.joca.2013.06.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 05/23/2013] [Accepted: 06/05/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To describe the currently used animal models for the study of osteoarthritis (OA) pain, with an emphasis on small animals (predominantly mice and rats). OUTLINE Narrative review summarizing the opportunities and limitations of the most commonly used small animal models for the study of pain and pain pathways associated with OA, and discussing currently used methods for pain assessment. Involvement of neural degeneration in OA is briefly discussed. A list of considerations when studying pain-related behaviours and pathways in animal models of OA is proposed. CONCLUSIONS Animal models offer great potential to unravel the complex pathophysiology of OA pain, its molecular and temporal regulation. They constitute a critical pathway for developing and testing disease-specific symptom-modifying therapeutic interventions. However, a number of issues remain to be resolved in order to standardize pre-clinical OA pain research and to optimize translation to clinical trials and patient therapies.
Collapse
Affiliation(s)
- Anne-Marie Malfait
- Department of Medicine, Section of Rheumatology, and Department of Biochemistry, Rush University Medical Center, Chicago IL, To whom correspondence should be addressed
| | - Christopher B. Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Institute of Bone and Joint Research, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Jason J. McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
39
|
Kelly S, Dunham JP, Murray F, Read S, Donaldson LF, Lawson SN. Spontaneous firing in C-fibers and increased mechanical sensitivity in A-fibers of knee joint-associated mechanoreceptive primary afferent neurones during MIA-induced osteoarthritis in the rat. Osteoarthritis Cartilage 2012; 20:305-13. [PMID: 22285737 DOI: 10.1016/j.joca.2012.01.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 12/15/2011] [Accepted: 01/01/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) pain mechanisms are poorly understood. We used the monosodium iodoacetate (MIA) model of knee OA to characterize changes in excitability during the course of OA in different classes of mechanosensitive afferents projecting to joint-associated tissues, and examine whether these afferent responses and pain behavior are correlated. METHODS Rats were injected intra-articularly with MIA (1mg in 50 μl). Hind-limb weight bearing was studied 3 (MIA3) and 14 (MIA14) days after MIA, followed by deep anesthesia and teased-nerve-fiber recordings. Spontaneous activity (SA) and mechanically evoked responses of A- and C-mechanosensitive fibers (AM and CM respectively, probably nociceptive) innervating tissues associated with the ipsilateral knee joint were examined. RESULTS MIA3 and MIA14 rats exhibited reduced ipsilateral weight bearing. SA (>0.02 impulses/s) occurred in ∼50% of CMs from MIA rats vs 0% in normals. SA firing rates in CMs were significantly higher than normal; decreased weight bearing was correlated with increased CM SA rates. Neither percentages of AMs with SA (20%) nor their firing rates (0-0.01 impulses/s) significantly increased after MIA. In contrast, in MIA rats AMs, but not CMs, exhibited decreased mechanical thresholds and increased firing rates in response to suprathreshold mechanical stimulation. CONCLUSIONS These findings of increased SA firing rate in CMs but not AMs and increased mechanical sensitivity of AMs, but not CMs, have not previously been reported. These are two distinct important physiological mechanisms that may underpin spontaneous pain (CMs) and stimulus-evoked pain (AMs) in OA. Our data contribute to a mechanism-based understanding of OA pain.
Collapse
Affiliation(s)
- S Kelly
- School of Physiology and Pharmacology, Medical Sciences Building, University of Bristol, BS8 1TD, UK
| | | | | | | | | | | |
Collapse
|
40
|
Okun A, Liu P, Davis P, Ren J, Remeniuk B, Brion T, Ossipov MH, Xie J, Dussor GO, King T, Porreca F. Afferent drive elicits ongoing pain in a model of advanced osteoarthritis. Pain 2012; 153:924-933. [PMID: 22387095 DOI: 10.1016/j.pain.2012.01.022] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/20/2012] [Accepted: 01/21/2012] [Indexed: 11/18/2022]
Abstract
Osteoarthritis (OA) is a chronic condition characterized by pain during joint movement. Additionally, patients with advanced disease experience pain at rest (ie, ongoing pain) that is generally resistant to nonsteroidal antiinflammatory drugs. Injection of monosodium iodoacetate (MIA) into the intraarticular space of the rodent knee is a well-established model of OA that elicits weight-bearing asymmetry and referred tactile and thermal hypersensitivity. Whether ongoing pain is present in this model is unknown. Additionally, the possible relationship of ongoing pain to MIA dose is not known. MIA produced weight asymmetry, joint osteolysis, and cartilage erosion across a range of doses (1, 3, and 4.8 mg). However, only rats treated with the highest dose of MIA showed conditioned place preference to a context paired with intraarticular lidocaine, indicating relief from ongoing pain. Diclofenac blocked the MIA-induced weight asymmetry but failed to block MIA-induced ongoing pain. Systemic AMG9810, a transient receptor potential V1 channel (TRPV1) antagonist, effectively blocked thermal hypersensitivity, but failed to block high-dose MIA-induced weight asymmetry or ongoing pain. Additionally, systemic or intraarticular HC030031, a TRPA1 antagonist, failed to block high-dose MIA-induced weight asymmetry or ongoing pain. Our studies suggest that a high dose of intraarticular MIA induces ongoing pain originating from the site of injury that is dependent on afferent fiber activity but apparently independent of TRPV1 or TRPA1 activation. Identification of mechanisms driving ongoing pain may enable development of improved treatments for patients with severe OA pain and diminish the need for joint replacement surgery.
Collapse
Affiliation(s)
- Alec Okun
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan 250012, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Schuelert N, McDougall JJ. Involvement of Nav 1.8 sodium ion channels in the transduction of mechanical pain in a rodent model of osteoarthritis. Arthritis Res Ther 2012; 14:R5. [PMID: 22225591 PMCID: PMC3392791 DOI: 10.1186/ar3553] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 12/16/2011] [Accepted: 01/07/2012] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION A subgroup of voltage gated sodium channels including Nav1.8 are exclusively expressed on small diameter primary afferent neurons and are therefore believed to be integral to the neurotransmission of nociceptive pain. The present study examined whether local application of A-803467, a selective blocker of the Nav 1.8 sodium channel, can reduce nociceptive transmission from the joint in a rodent model of osteoarthritis (OA). METHODS OA-like changes were induced in male Wistar rats by an intra-articular injection of 3 mg sodium monoiodoacetate (MIA). Joint nociception was measured at day 14 by recording electrophysiologically from knee joint primary afferents in response to non-noxious and noxious rotation of the joint both before and following close intra-arterial injection of A-803467. The effect of Nav1.8 blockade on joint pain perception and secondary allodynia were determined in MIA treated animals by hindlimb incapacitance and von Frey hair algesiometry respectively. RESULTS A-803467 significantly reduced the firing rate of joint afferents during noxious rotation of the joint but had no effect during non-noxious rotation. In the pain studies, peripheral injection of A-803467 into OA knees attenuated hindlimb incapacitance and secondary allodynia. CONCLUSIONS These studies show for the first time that the Nav1.8 sodium channel is part of the molecular machinery involved in mechanotransduction of joint pain. Targeting the Nav1.8 sodium channel on joint nociceptors could therefore be useful for the treatment of OA pain, avoiding the unwanted side effects of non-selective nerve blocks.
Collapse
Affiliation(s)
- Niklas Schuelert
- Department of Physiology & Pharmacology, University of Calgary, 3330, Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | | |
Collapse
|
42
|
Local application of the endocannabinoid hydrolysis inhibitor URB597 reduces nociception in spontaneous and chemically induced models of osteoarthritis. Pain 2011; 152:975-981. [DOI: 10.1016/j.pain.2010.11.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2010] [Revised: 11/10/2010] [Accepted: 11/22/2010] [Indexed: 11/22/2022]
|
43
|
Liu P, Okun A, Ren J, Guo RC, Ossipov MH, Xie J, King T, Porreca F. Ongoing pain in the MIA model of osteoarthritis. Neurosci Lett 2011; 493:72-5. [PMID: 21241772 DOI: 10.1016/j.neulet.2011.01.027] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/06/2011] [Accepted: 01/09/2011] [Indexed: 11/17/2022]
Abstract
Osteoarthritis (OA) is a chronic pain condition characterized by pain during joint use as well as pain at rest (i.e., ongoing pain). Although injection of monosodium iodoacetate (MIA) into the intra-articular space of the rodent knee is a well established model of OA pain that is characterized by changes in weight bearing and hypersensitivity to tactile and thermal stimuli, it is not known if this procedure elicits ongoing pain. Further, the time-course and possible underlying mechanisms of these components of pain remain poorly understood. In these studies, we demonstrated the presence of ongoing pain in addition to changes in weight bearing and evoked hypersensitivity. Twenty-eight days following MIA injection, spinal clonidine blocked changes in weight bearing and thermal hypersensitivity and produced place preference indicating that MIA induces ongoing and evoked pain. These findings demonstrate the presence of ongoing pain in this model that is present at a late-time point after MIA allowing for mechanistic investigation.
Collapse
Affiliation(s)
- Ping Liu
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Im HJ, Kim JS, Li X, Kotwal N, Sumner DR, van Wijnen AJ, Davis FJ, Yan D, Levine B, Henry JL, Desevré J, Kroin JS. Alteration of sensory neurons and spinal response to an experimental osteoarthritis pain model. ACTA ACUST UNITED AC 2010; 62:2995-3005. [PMID: 20556813 DOI: 10.1002/art.27608] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To verify the biologic links between progressive cellular and structural alterations within knee joint components and development of symptomatic chronic pain that are characteristic of osteoarthritis (OA), and to investigate the molecular basis of alterations in nociceptive pathways caused by OA-induced pain. METHODS An animal model of knee joint OA pain was generated by intraarticular injection of mono-iodoacetate (MIA) in Sprague-Dawley rats, and symptomatic pain behavior tests were performed. Relationships between development of OA with accompanying pain responses and gradual alterations in cellular and structural knee joint components (i.e., cartilage, synovium, meniscus, subchondral bone) were examined by histologic and immunohistologic analysis, microscopic examination, and microfocal computed tomography. Progressive changes in the dynamic interrelationships between peripheral knee joint tissue and central components of nociceptive pathways caused by OA-induced pain were examined by investigating cytokine production and expression in sensory neurons of the dorsal root ganglion and spinal cord. RESULTS We observed that structural changes in components of the peripheral knee joint correlate with alterations in the central compartments (dorsal root ganglia and the spinal cord) and symptomatic pain assessed by behavioral hyperalgesia. Our comparative gene expression studies revealed that the pain pathways in MIA-induced knee OA may overlap, at least in part, with neuropathic pain mechanisms. Similar results were also observed upon destabilization of the knee joint in the anterior cruciate ligament transection and destabilization of the medial meniscus models of OA. CONCLUSION Our results indicate that MIA-induced joint degeneration in rats generates an animal model that is suitable for mechanistic and pharmacologic studies on nociceptive pain pathways caused by OA, and provide key in vivo evidence that OA pain is caused by central sensitization through communication between peripheral OA nociceptors and the central sensory system. Furthermore, our data suggest a mechanistic overlap between OA-induced pain and neuropathic pain.
Collapse
Affiliation(s)
- Hee-Jeong Im
- Rush University Medical Center and University of Illinois at Chicago, IL, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wu Q, Henry JL. Changes in Abeta non-nociceptive primary sensory neurons in a rat model of osteoarthritis pain. Mol Pain 2010; 6:37. [PMID: 20594346 PMCID: PMC2908067 DOI: 10.1186/1744-8069-6-37] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 07/01/2010] [Indexed: 11/18/2022] Open
Abstract
Background Pain is a major debilitating factor in osteoarthritis (OA), yet few mechanism-based therapies are available. To address the need to understand underlying mechanisms the aim of the present study was to determine changes in sensory neurons in an animal model of OA pain. Results The model displayed typical osteoarthritis pathology characterized by cartilage degeneration in the knee joint and also manifested knee pathophysiology (edema and increased vasculature permeability of the joint) and altered nociception of the affected limb (hind paw tenderness and knee articulation-evoked reduction in the tail flick latency). Neurons included in this report innervated regions throughout the entire hind limb. Aβ-fiber low threshold mechanoreceptors exhibited a slowing of the dynamics of action potential (AP) genesis, including wider AP duration and slower maximum rising rate, and muscle spindle neurons were the most affected subgroup. Only minor AP configuration changes were observed in either C- or Aδ-fiber nociceptors. Conclusion Thus, at one month after induction of the OA model Aβ-fiber low threshold mechanoreceptors but not C- or Aδ-fiber nociceptors had undergone changes in electrophysiological properties. If these changes reflect a change in functional role of these neurons in primary afferent sensory processing, then Aβ-fiber non-nociceptive primary sensory neurons may be involved in the pathogenesis of OA pain. Further, it is important to point out that the patterns of the changes we observed are consistent with observations in models of peripheral neuropathy but not models of peripheral inflammation.
Collapse
Affiliation(s)
- Qi Wu
- Psychiatry and Behavioral Neurosciences, McMaster University, HSC 4N35, Hamilton, Ontario, Canada
| | | |
Collapse
|
47
|
Goldring MB. The link between structural damage and pain in a genetic model of osteoarthritis and intervertebral disc degeneration: a joint misadventure. ARTHRITIS AND RHEUMATISM 2009; 60:2550-2. [PMID: 19714635 PMCID: PMC2748171 DOI: 10.1002/art.24771] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Mary B. Goldring
- Mary B. Goldring, PhD: Research Division, Hospital for Special Surgery at Weill Cornell Medical College, Caspary Research Building, 535 E. 70th Street, New York, NY 10021, USA
| |
Collapse
|
48
|
Grading of monosodium iodoacetate-induced osteoarthritis reveals a concentration-dependent sensitization of nociceptors in the knee joint of the rat. Neurosci Lett 2009; 465:184-8. [PMID: 19716399 DOI: 10.1016/j.neulet.2009.08.063] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 08/14/2009] [Accepted: 08/24/2009] [Indexed: 11/21/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by joint pain for which there is currently no effective treatment. Previous studies have found that intra-articular injection of monosodium iodoacetate (MIA) caused a dose-dependent destruction of rat knees with concomitant increased pain. In this study, varying degrees of OA were induced by intra-articular injection of 0.1 mg, 0.3 mg and 3 mg MIA. Electrophysiological recordings were made from knee joint primary afferents in response to rotation of the joint and firing frequencies were determined and compared to saline-injected control joints. The analgesic effect of local application of the classic non-steroidal anti-inflammatory drug (NSAID) diclofenac (0.1 mg/0.1 ml bolus) was also determined in each group. Joint afferent firing frequency was significantly enhanced in OA knees compared to saline injected control joints and the magnitude of this sensitization showed a direct relationship with increasing dose of MIA. Diclofenac reduced nociception significantly in the 3 mg MIA treated joint, but had no effect on nerve mechanosensitivity in rats with milder OA. This study shows for the first time that MIA produces a graded sensitization of joint nociceptors making this a useful model for the study of pain mechanisms in joints with progressive OA severity. The anti-nociceptive effect of diclofenac further indicates that the MIA model offers an attractive means of objectively testing potential therapeutic agents.
Collapse
|
49
|
Bove SE, Flatters SJ, Inglis JJ, Mantyh PW. New advances in musculoskeletal pain. BRAIN RESEARCH REVIEWS 2009; 60:187-201. [PMID: 19166876 PMCID: PMC4460838 DOI: 10.1016/j.brainresrev.2008.12.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 12/29/2008] [Indexed: 11/28/2022]
Abstract
Non-malignant musculoskeletal pain is the most common clinical symptom that causes patients to seek medical attention and is a major cause of disability in the world. Musculoskeletal pain can arise from a variety of common conditions including osteoarthritis, rheumatoid arthritis, osteoporosis, surgery, low back pain and bone fracture. A major problem in designing new therapies to treat musculoskeletal pain is that the underlying mechanisms driving musculoskeletal pain are not well understood. This lack of knowledge is largely due to the scarcity of animal models that closely mirror the human condition which would allow the development of a mechanistic understanding and novel therapies to treat this pain. To begin to develop a mechanism-based understanding of the factors involved in generating musculoskeletal pain, in this review we present recent advances in preclinical models of osteoarthritis, post-surgical pain and bone fracture pain. The models discussed appear to offer an attractive platform for understanding the factors that drive this pain and the preclinical screening of novel therapies to treat musculoskeletal pain. Developing both an understanding of the mechanisms that drive persistent musculoskeletal pain and novel mechanism-based therapies to treat these unique pain states would address a major unmet clinical need and have significant clinical, economic and societal benefits.
Collapse
Affiliation(s)
- Susan E. Bove
- Department of Neurosciences Biology, Pfizer Global Research and Development, Groton Laboratories, Groton, CT, USA
| | - Sarah J.L. Flatters
- Wolfson Center for Age-Related Diseases, King’s College London, London SE1 1UL, UK
| | - Julia J. Inglis
- Kennedy Institute of Rheumatology, Imperial College London, London W6 8LH, UK
| | - Patrick W. Mantyh
- Department of Pharmacology and the Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
- VA Medical Center, Minneapolis, MN, USA
| |
Collapse
|
50
|
Abstract
This article delineates the characteristic symptoms and signs associated with OA and how they can be used to make the clinical diagnosis. The predominant symptom in most patients is pain. The remainder of the article focuses on what we know causes pain in OA and contributes to its severity. Much has been learned over recent years; however, for the budding researcher much of this puzzle remains unexplored or inadequately understood.
Collapse
Affiliation(s)
- David J Hunter
- Division of Research, New England Baptist Hospital, 125 Parker Hill Avenue, Boston MA 02120, USA.
| | | | | |
Collapse
|