Skip to main content
Top
Published in: Current Osteoporosis Reports 5/2018

01-10-2018 | Bone and Joint Pain (T King and S Amin, Section Editors)

Mechanisms for Joint Pain in Rheumatoid Arthritis (RA): from Cytokines to Central Sensitization

Authors: Angela Zhang, Yvonne C. Lee

Published in: Current Osteoporosis Reports | Issue 5/2018

Login to get access

Abstract

Purpose of Review

Pain in rheumatoid arthritis (RA) may be due to different etiologies, ranging from peripheral inflammation to dysregulation of central nervous system (CNS) processing. This review evaluates relevant literature published on RA pain mechanisms in recent years.

Recent Findings

Despite successes of disease-modifying antirheumatic drugs (DMARDs), pain persists for many RA patients. Studies involving patient-reported outcomes, quantitative sensory testing, and neuroimaging indicate that, in addition to joint inflammation, abnormalities in CNS pain processing may contribute to pain. Some DMARDs (e.g., janus kinus inhibitors) may work via multiple pathways to decrease pain. Adjunctive treatments (e.g., antidepressants, antiepileptics) may also be useful in managing pain in RA patients with well-controlled disease.

Summary

Both peripheral and central mechanisms play key roles in the expression of pain in RA. To effectively manage pain, physicians need accurate assessment tools to identify the pathways involved in each patient so that treatments may be appropriately targeted.
Literature
1.
go back to reference Won S, Cho SK, Kim D, Han M, Lee J, Jang EJ, et al. Update on the prevalence and incidence of rheumatoid arthritis in Korea and an analysis of medical care and drug utilization. Rheumatol Int. 2018;38:649–56.CrossRefPubMed Won S, Cho SK, Kim D, Han M, Lee J, Jang EJ, et al. Update on the prevalence and incidence of rheumatoid arthritis in Korea and an analysis of medical care and drug utilization. Rheumatol Int. 2018;38:649–56.CrossRefPubMed
2.
go back to reference Park J, Mendy A, Viera ER. Various types of arthritis in the United States: prevalence and age-related trends from 1999 to 2014. Am J Public Health. 2018;108(2):256–8.CrossRefPubMed Park J, Mendy A, Viera ER. Various types of arthritis in the United States: prevalence and age-related trends from 1999 to 2014. Am J Public Health. 2018;108(2):256–8.CrossRefPubMed
3.
go back to reference Al Attia HM, Al Abbasi M. Sensing the main health concerns in patients with established rheumatoid arthritis. Clin Rheumatol. 2011;30:1511–4.CrossRefPubMed Al Attia HM, Al Abbasi M. Sensing the main health concerns in patients with established rheumatoid arthritis. Clin Rheumatol. 2011;30:1511–4.CrossRefPubMed
4.
go back to reference Altawil R, Saevarsdottir S, Wedren S, Alfredsson L, Klareskog L, Lampa J. Remaining pain in early rheumatoid arthritis patients treated with methotrexate. Arthritis Care Res (Hoboken). 2016;68:1061–8.CrossRef Altawil R, Saevarsdottir S, Wedren S, Alfredsson L, Klareskog L, Lampa J. Remaining pain in early rheumatoid arthritis patients treated with methotrexate. Arthritis Care Res (Hoboken). 2016;68:1061–8.CrossRef
5.
go back to reference McWilliams DF, Walsh DA. Factors predicting pain and early discontinuation of tumour necrosis factor-α-inhibitors in people with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. BMC Musculoskelet Disord. 2016;17:337.CrossRefPubMedPubMedCentral McWilliams DF, Walsh DA. Factors predicting pain and early discontinuation of tumour necrosis factor-α-inhibitors in people with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. BMC Musculoskelet Disord. 2016;17:337.CrossRefPubMedPubMedCentral
6.
go back to reference Khan NA, Spencer HJ, Abda E, Aggarwal A, Alten R, Ancuta C. Determinants of discordance in patients’ and physicians’ rating of rheumatoid arthritis disease activity. Arthritis Care Res (Hoboken). 2012;64(2):206–14.CrossRefPubMedPubMedCentral Khan NA, Spencer HJ, Abda E, Aggarwal A, Alten R, Ancuta C. Determinants of discordance in patients’ and physicians’ rating of rheumatoid arthritis disease activity. Arthritis Care Res (Hoboken). 2012;64(2):206–14.CrossRefPubMedPubMedCentral
7.
go back to reference Studenic P, Radner H, Smolen JS, Aletaha D. Discrepancies between patients and physicians in their perceptions of rheumatoid arthritis disease activity. Arthritis Rheum. 2012;64(9):2814–23.CrossRefPubMed Studenic P, Radner H, Smolen JS, Aletaha D. Discrepancies between patients and physicians in their perceptions of rheumatoid arthritis disease activity. Arthritis Rheum. 2012;64(9):2814–23.CrossRefPubMed
8.
go back to reference Purabdollah M, Lakdizaji S, Rahmani A. Relationship between sleep, pain and inflammatory markers in patients with rheumatoid arthritis. J Caring Sci. 2017;6(3):249–55.CrossRefPubMedPubMedCentral Purabdollah M, Lakdizaji S, Rahmani A. Relationship between sleep, pain and inflammatory markers in patients with rheumatoid arthritis. J Caring Sci. 2017;6(3):249–55.CrossRefPubMedPubMedCentral
9.
go back to reference Boyden SD, Hossain IN, Wohlfahrt A, Lee YC. Non-inflammatory causes of pain in patients with rheumatoid arthritis. Curr Rheumatol Rep. 2016;18(6):30–7.CrossRefPubMed Boyden SD, Hossain IN, Wohlfahrt A, Lee YC. Non-inflammatory causes of pain in patients with rheumatoid arthritis. Curr Rheumatol Rep. 2016;18(6):30–7.CrossRefPubMed
10.
go back to reference Meeus M, Vervisch S, De Clerck LS, Moorkens G, Hans G, Nijs J. Central sensitization in patients with rheumatoid arthritis: a systematic literature review. Semin Arthritis Rheum. 2012;41(4):556–67.CrossRefPubMed Meeus M, Vervisch S, De Clerck LS, Moorkens G, Hans G, Nijs J. Central sensitization in patients with rheumatoid arthritis: a systematic literature review. Semin Arthritis Rheum. 2012;41(4):556–67.CrossRefPubMed
11.
go back to reference Pinho-Ribeiro FA, Verri WA Jr, Chiu IM. Nociceptor sensory neuron–immune interactions in pain and inflammation. Trends Immunol. 2017;38(1):5–19.CrossRefPubMed Pinho-Ribeiro FA, Verri WA Jr, Chiu IM. Nociceptor sensory neuron–immune interactions in pain and inflammation. Trends Immunol. 2017;38(1):5–19.CrossRefPubMed
13.
go back to reference Boettger MK, Hensellek S, Richter F, Gajda M, Stöckigt R, von Banchet GS, et al. Antinociceptive effects of tumor necrosis factor alpha neutralization in a rat model of antigen-induced arthritis: evidence of a neuronal target. Arthritis Rheum. 2008;58(8):2368–78.CrossRefPubMed Boettger MK, Hensellek S, Richter F, Gajda M, Stöckigt R, von Banchet GS, et al. Antinociceptive effects of tumor necrosis factor alpha neutralization in a rat model of antigen-induced arthritis: evidence of a neuronal target. Arthritis Rheum. 2008;58(8):2368–78.CrossRefPubMed
14.
go back to reference Copray JC, Mantingh I, Brouwer N, Biber K, Küst BM, Liem RS. Expression of interleukin-1 beta in rat dorsal root ganglia. J Neuroimmunol. 2001;118(2):203–11.CrossRefPubMed Copray JC, Mantingh I, Brouwer N, Biber K, Küst BM, Liem RS. Expression of interleukin-1 beta in rat dorsal root ganglia. J Neuroimmunol. 2001;118(2):203–11.CrossRefPubMed
15.
go back to reference Segond von Banchet G, Boettger MK, König C, Iwakura Y, Bräuer R, Schaible HG. Neuronal IL-17 receptor upregulates TRPV4 but not TRPV1 receptors in DRG neurons and mediates mechanical but not thermal hyperalgesia. Mol Sci Neurosci. 2013;52:152–60.CrossRef Segond von Banchet G, Boettger MK, König C, Iwakura Y, Bräuer R, Schaible HG. Neuronal IL-17 receptor upregulates TRPV4 but not TRPV1 receptors in DRG neurons and mediates mechanical but not thermal hyperalgesia. Mol Sci Neurosci. 2013;52:152–60.CrossRef
16.
go back to reference Obreja O, Biasio W, Andratsch M, Lips KS, Rathee PK, Ludwig A, et al. Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons. Brain. 2005;128(Pt 7):1634–41.CrossRefPubMed Obreja O, Biasio W, Andratsch M, Lips KS, Rathee PK, Ludwig A, et al. Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons. Brain. 2005;128(Pt 7):1634–41.CrossRefPubMed
17.
go back to reference Richter F, Natura G, Löser S, Schmidt K, Viisanen H, Schaible HG. Tumor necrosis factor causes persistent sensitization of joint nociceptors to mechanical stimuli in rats. Arthritis Rheum. 2010;62(12):3806–14.CrossRefPubMed Richter F, Natura G, Löser S, Schmidt K, Viisanen H, Schaible HG. Tumor necrosis factor causes persistent sensitization of joint nociceptors to mechanical stimuli in rats. Arthritis Rheum. 2010;62(12):3806–14.CrossRefPubMed
18.
go back to reference Brenn D, Richter F, Schaible HG. Sensitization of unmyelinated sensory fibers of the joint nerve to mechanical stimuli by interleukin-6 in the rat: an inflammatory mechanism of joint pain. Arthritis Rheum. 2007;56(1):351–9.CrossRefPubMed Brenn D, Richter F, Schaible HG. Sensitization of unmyelinated sensory fibers of the joint nerve to mechanical stimuli by interleukin-6 in the rat: an inflammatory mechanism of joint pain. Arthritis Rheum. 2007;56(1):351–9.CrossRefPubMed
19.
go back to reference Richter F, Natura G, Ebbinghaus M, von Banchet GS, Hensellek S, König C, et al. Interleukin-17 sensitizes joint nociceptors to mechanical stimuli and contributes to arthritis pain through neuronal interleukin-17 receptors in rodents. Arthritis Rheum. 2012;64(12):4125–34.CrossRefPubMed Richter F, Natura G, Ebbinghaus M, von Banchet GS, Hensellek S, König C, et al. Interleukin-17 sensitizes joint nociceptors to mechanical stimuli and contributes to arthritis pain through neuronal interleukin-17 receptors in rodents. Arthritis Rheum. 2012;64(12):4125–34.CrossRefPubMed
20.
go back to reference Ebbinghaus M, Uhlig B, Richter F, von Banchet GS, Gajda M, Bräuer R, et al. The role of interleukin-1B in arthritic pain: main involvement in thermal, but not mechanical hyperalgesia in rat antigen-induced arthritis. Arthritis Rheum. 2012;64(12):3897–907.CrossRefPubMed Ebbinghaus M, Uhlig B, Richter F, von Banchet GS, Gajda M, Bräuer R, et al. The role of interleukin-1B in arthritic pain: main involvement in thermal, but not mechanical hyperalgesia in rat antigen-induced arthritis. Arthritis Rheum. 2012;64(12):3897–907.CrossRefPubMed
21.
go back to reference Kwon M, Altin M, Duenas H, Alev L. The role of descending inhibitory pathways on chronic pain modulation and clinical implications. Pain Pract. 2014;14(7):656–67.CrossRefPubMed Kwon M, Altin M, Duenas H, Alev L. The role of descending inhibitory pathways on chronic pain modulation and clinical implications. Pain Pract. 2014;14(7):656–67.CrossRefPubMed
22.
go back to reference Zhou M. Descending facilitation. Mol Pain. 2017;13:1744806917699212. Zhou M. Descending facilitation. Mol Pain. 2017;13:1744806917699212.
23.
go back to reference Vanegas H, Schaible HG. Descending control of persistent pain: inhibitory or facilitatory? Brain Res Brain Res Rev. 2004;46:295–309.CrossRefPubMed Vanegas H, Schaible HG. Descending control of persistent pain: inhibitory or facilitatory? Brain Res Brain Res Rev. 2004;46:295–309.CrossRefPubMed
24.
25.
go back to reference Petersen KK, Simonsen O, Laursen MB, Arendt-Nielsen L. The role of preoperative radiological severity, sensory testing, and temporal summation on chronic postoperative pain following total knee arthroplasty. Clin J Pain. 2018;34(3):193–7.PubMed Petersen KK, Simonsen O, Laursen MB, Arendt-Nielsen L. The role of preoperative radiological severity, sensory testing, and temporal summation on chronic postoperative pain following total knee arthroplasty. Clin J Pain. 2018;34(3):193–7.PubMed
26.
go back to reference Huang J, Burston JJ, Li L, Ashraf S, Mapp PI, Bennett AJ, et al. Targeting the D series resolvin receptor system for the treatment of osteoarthritis pain. Arthritis Rheumatol. 2017;69(5):996–1008.CrossRefPubMedPubMedCentral Huang J, Burston JJ, Li L, Ashraf S, Mapp PI, Bennett AJ, et al. Targeting the D series resolvin receptor system for the treatment of osteoarthritis pain. Arthritis Rheumatol. 2017;69(5):996–1008.CrossRefPubMedPubMedCentral
27.
go back to reference Felson DT, Niu J, Quinn EK, Neogi T, Lewis CL, Frey Law L, et al. Multiple nonspecific sites of joint pain outside the knees develop in persons with knee pain. Arthritis Rheumatol. 2017;69(5):335–42.CrossRefPubMedPubMedCentral Felson DT, Niu J, Quinn EK, Neogi T, Lewis CL, Frey Law L, et al. Multiple nonspecific sites of joint pain outside the knees develop in persons with knee pain. Arthritis Rheumatol. 2017;69(5):335–42.CrossRefPubMedPubMedCentral
28.
go back to reference Fischer BD, Adeyemo A, O’Leary ME, Bottaro A. Animal models of rheumatoid pain: experimental systems and insights. Arthritis Res Ther. 2017;19(1):146.CrossRefPubMedPubMedCentral Fischer BD, Adeyemo A, O’Leary ME, Bottaro A. Animal models of rheumatoid pain: experimental systems and insights. Arthritis Res Ther. 2017;19(1):146.CrossRefPubMedPubMedCentral
29.
go back to reference Hu Y, Cheng W, Cai W, Yue Y, Li J, Zhang P. Advances in research on animal models of rheumatoid arthritis. Clin Rheumatol. 2013;32(2):161–5.CrossRefPubMed Hu Y, Cheng W, Cai W, Yue Y, Li J, Zhang P. Advances in research on animal models of rheumatoid arthritis. Clin Rheumatol. 2013;32(2):161–5.CrossRefPubMed
30.
go back to reference • Wigerblad G, Bas DB, Fernades-Cergueira C, Krishnamurthy A, Nandakumar KS, Rogoz K, et al. Autoantibodies to citrullinated proteins induce joint pain independent of inflammation via a chemokine-dependent mechanism. Ann Rheum Dis. 2016;75(4):730–8. This study identifies the CXCL1/IL-8 pathway as potential novel nociceptive pathway, which may act independently of peripheral joint inflammation, to cause pain in inflammatory arthritis. CrossRefPubMed • Wigerblad G, Bas DB, Fernades-Cergueira C, Krishnamurthy A, Nandakumar KS, Rogoz K, et al. Autoantibodies to citrullinated proteins induce joint pain independent of inflammation via a chemokine-dependent mechanism. Ann Rheum Dis. 2016;75(4):730–8. This study identifies the CXCL1/IL-8 pathway as potential novel nociceptive pathway, which may act independently of peripheral joint inflammation, to cause pain in inflammatory arthritis. CrossRefPubMed
31.
go back to reference Su J, Gao T, Shi T, Xiang Q, Xu X, Wiesenfeld-Hallin Z, et al. Phenotypic changes in dorsal root ganglion and spinal cord in the collagen antibody-induced arthritis mouse model. J Comp Neurol. 2015;523(10):1505–28.CrossRefPubMed Su J, Gao T, Shi T, Xiang Q, Xu X, Wiesenfeld-Hallin Z, et al. Phenotypic changes in dorsal root ganglion and spinal cord in the collagen antibody-induced arthritis mouse model. J Comp Neurol. 2015;523(10):1505–28.CrossRefPubMed
32.
go back to reference Lang R, Gundlach AL, Holmes FE, Hobson SA, Wynick D, Hokfelt T, et al. Physiology, signaling and pharmacology of galanin peptides and receptors: three decades of emerging diversity. Pharmacol Rev. 2015;67(1):118–75.CrossRefPubMed Lang R, Gundlach AL, Holmes FE, Hobson SA, Wynick D, Hokfelt T, et al. Physiology, signaling and pharmacology of galanin peptides and receptors: three decades of emerging diversity. Pharmacol Rev. 2015;67(1):118–75.CrossRefPubMed
33.
go back to reference Li CY, Zhang XL, Matthews EA, Li KW, Kurwa A, Boroujerdi A, et al. Calcium channel alpha2delta1 subunit mediates spinal hyperexcitability in pain modulation. Pain. 2016;125(1–2):20–34. Li CY, Zhang XL, Matthews EA, Li KW, Kurwa A, Boroujerdi A, et al. Calcium channel alpha2delta1 subunit mediates spinal hyperexcitability in pain modulation. Pain. 2016;125(1–2):20–34.
34.
go back to reference Scheytt S, Riediger N, Braunsdorf S, Sommer C, Uceyler N. Increased gene expression of growth associated protein-43 in skin of patients with early-stage peripheral neuropathies. J Neurol Sci. 2015;355(1–2):131–7.CrossRefPubMed Scheytt S, Riediger N, Braunsdorf S, Sommer C, Uceyler N. Increased gene expression of growth associated protein-43 in skin of patients with early-stage peripheral neuropathies. J Neurol Sci. 2015;355(1–2):131–7.CrossRefPubMed
35.
go back to reference Ghilardi JR, Freeman KT, Jimenez-Andrade JM, Coughlin K, Kaczmarska MJ, Castaneda-Corral G, et al. Neuroplasticity of sensory and sympathetic nerve fibers in the painful arthritic joint. Arhritis Rheum. 2012;64(7):2223–32.CrossRef Ghilardi JR, Freeman KT, Jimenez-Andrade JM, Coughlin K, Kaczmarska MJ, Castaneda-Corral G, et al. Neuroplasticity of sensory and sympathetic nerve fibers in the painful arthritic joint. Arhritis Rheum. 2012;64(7):2223–32.CrossRef
36.
go back to reference McWilliams DF, Zhang W, Mansell JS, Kiely PD, Young A, Walsh DA. Predictors of change in bodily pain in early rheumatoid arthritis: an inception cohort study. Arthritis Care Res (Hoboken). 2012;64(10):1505–13.CrossRef McWilliams DF, Zhang W, Mansell JS, Kiely PD, Young A, Walsh DA. Predictors of change in bodily pain in early rheumatoid arthritis: an inception cohort study. Arthritis Care Res (Hoboken). 2012;64(10):1505–13.CrossRef
37.
go back to reference Ahmed S, Magan T, Vargas M, Harrison A, Sofat N. Use of the painDETECT tool in rheumatoid arthritis suggests neuropathic and sensitization components in pain reporting. J Pain Res. 2014;7:579–88.PubMedPubMedCentral Ahmed S, Magan T, Vargas M, Harrison A, Sofat N. Use of the painDETECT tool in rheumatoid arthritis suggests neuropathic and sensitization components in pain reporting. J Pain Res. 2014;7:579–88.PubMedPubMedCentral
38.
go back to reference Freynhagen R, Baron R, Gockel U, Tolle TR. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin. 2006;22(10):1911–20.CrossRefPubMed Freynhagen R, Baron R, Gockel U, Tolle TR. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin. 2006;22(10):1911–20.CrossRefPubMed
39.
go back to reference Freynhagen R, Tölle TR, Gockel U, Baron R. The painDETECT project – far more than a screening tool on neuropathic pain. Curr Med Res Opin. 2016;32(6):1033–57.CrossRefPubMed Freynhagen R, Tölle TR, Gockel U, Baron R. The painDETECT project – far more than a screening tool on neuropathic pain. Curr Med Res Opin. 2016;32(6):1033–57.CrossRefPubMed
40.
go back to reference Rifbjerg-Madsen S, Waehrens EE, Danneskiold-Samsoe B, Amris K. Psychometric properties of the painDETECT questionnaire in rheumatoid arthritis, psoriatic arthritis and spondyloarthritis: Rasch analysis and test-retest reliability. Health Qual Life Outcomes. 2017;15(1):110.CrossRefPubMedPubMedCentral Rifbjerg-Madsen S, Waehrens EE, Danneskiold-Samsoe B, Amris K. Psychometric properties of the painDETECT questionnaire in rheumatoid arthritis, psoriatic arthritis and spondyloarthritis: Rasch analysis and test-retest reliability. Health Qual Life Outcomes. 2017;15(1):110.CrossRefPubMedPubMedCentral
41.
go back to reference Hochman JR, Davis AM, Eikayam J, Gagliese L, Hawker GA. Neuropathic pain symptoms on the modified painDETECT correlated with signs of central sensitization in knee osteoarthritis. Osteoarthr Cartil. 2013;21(9):1236–42.CrossRefPubMed Hochman JR, Davis AM, Eikayam J, Gagliese L, Hawker GA. Neuropathic pain symptoms on the modified painDETECT correlated with signs of central sensitization in knee osteoarthritis. Osteoarthr Cartil. 2013;21(9):1236–42.CrossRefPubMed
42.
go back to reference Gwilym SE, Keltner JR, Warnaby CE, Carr AJ, Chizh B, Chessell I, et al. Psychosocial and functional imaging evidence supporint the presence of central sensitization in a cohort of osteoarthritis patients. Arthritis Rheum. 2009;61(9):1226–34.CrossRefPubMed Gwilym SE, Keltner JR, Warnaby CE, Carr AJ, Chizh B, Chessell I, et al. Psychosocial and functional imaging evidence supporint the presence of central sensitization in a cohort of osteoarthritis patients. Arthritis Rheum. 2009;61(9):1226–34.CrossRefPubMed
43.
go back to reference Cruz-Almeida Y, Fillingim RB. Can quantitative sensory testing move us closer to mechanism-based pain management? Pain Med. 2014;15(1):61–72.CrossRefPubMed Cruz-Almeida Y, Fillingim RB. Can quantitative sensory testing move us closer to mechanism-based pain management? Pain Med. 2014;15(1):61–72.CrossRefPubMed
44.
45.
go back to reference Joharatnam N, McWilliams DF, Wilson D, Wheeler M, Pande I, Walsh DA. A cross-sectional study of pain sensitivity, disease-activity assessment, mental health, and fibromyalgia status in rheumatoid arthritis. Arthritis Res Ther. 2015;17:11.CrossRefPubMedPubMedCentral Joharatnam N, McWilliams DF, Wilson D, Wheeler M, Pande I, Walsh DA. A cross-sectional study of pain sensitivity, disease-activity assessment, mental health, and fibromyalgia status in rheumatoid arthritis. Arthritis Res Ther. 2015;17:11.CrossRefPubMedPubMedCentral
46.
go back to reference Graven-Nielsen T, Arendt-Nielsen L. Assessment of mechanisms in localized and widespread musculoskeletal pain. Nat Rev Rheumatol. 2010;6(10):599–606.CrossRefPubMed Graven-Nielsen T, Arendt-Nielsen L. Assessment of mechanisms in localized and widespread musculoskeletal pain. Nat Rev Rheumatol. 2010;6(10):599–606.CrossRefPubMed
47.
go back to reference Vladimirova N, Jespersen A, Barteis EM, Christensen AW, Bliddal H, Danneskioid-Samsoe B. Pain sensitization in women with active rheumatoid arthritis: a comparative cross-sectional study. Arthritis. 2015;2015:434109.CrossRefPubMedPubMedCentral Vladimirova N, Jespersen A, Barteis EM, Christensen AW, Bliddal H, Danneskioid-Samsoe B. Pain sensitization in women with active rheumatoid arthritis: a comparative cross-sectional study. Arthritis. 2015;2015:434109.CrossRefPubMedPubMedCentral
48.
go back to reference Meeus M, Ickmans K, Struyf F, Hermans L, Van Noesel K, Oderkerk J, et al. Does acetaminophen activate endogenous pain inhibition in chronic fatigue syndrome/ fibromyalgia and rheumatoid arthritis? A double-blind randomized controlled cross-over trial. Pain Physician. 2013;16(2):E61–70.PubMed Meeus M, Ickmans K, Struyf F, Hermans L, Van Noesel K, Oderkerk J, et al. Does acetaminophen activate endogenous pain inhibition in chronic fatigue syndrome/ fibromyalgia and rheumatoid arthritis? A double-blind randomized controlled cross-over trial. Pain Physician. 2013;16(2):E61–70.PubMed
49.
go back to reference •• Christensen AW, Rifbjerg-Madsen S, Christensen R, Dreyer L, Boesen M, Ellegaard K, et al. Ultrasound Doppler but not temporal summation of pain predicts DAS28 response in rheumatoid arthritis: a prospective cohort study. Rheumatology (Oxford). 2016;55(6):1091–8. This study indicates that changes in disease activity are better predicted by baseline measures of subclinical inflammation (assessed by ultrasound Doppler) than central sensitization (assessed by temporal summation). CrossRef •• Christensen AW, Rifbjerg-Madsen S, Christensen R, Dreyer L, Boesen M, Ellegaard K, et al. Ultrasound Doppler but not temporal summation of pain predicts DAS28 response in rheumatoid arthritis: a prospective cohort study. Rheumatology (Oxford). 2016;55(6):1091–8. This study indicates that changes in disease activity are better predicted by baseline measures of subclinical inflammation (assessed by ultrasound Doppler) than central sensitization (assessed by temporal summation). CrossRef
50.
go back to reference Lee YC, Lu B, Edwards RR, Wasan AD, Nassikas NJ, Clauw DJ, et al. The role of sleep problems in central pain processing in rheumatoid arthritis. Arthritis Rheum. 2013;65(1):59–6.CrossRefPubMedPubMedCentral Lee YC, Lu B, Edwards RR, Wasan AD, Nassikas NJ, Clauw DJ, et al. The role of sleep problems in central pain processing in rheumatoid arthritis. Arthritis Rheum. 2013;65(1):59–6.CrossRefPubMedPubMedCentral
51.
go back to reference Lee YC, Bingham CO 3rd, Edwards RR, Marder W, Kristine P, Bolster MB, et al. Pain sensitization is associated with disease activity in rheumatoid arthritis patients: a cross-sectional study. Arthritis Care Res (Hoboken). 2018;70(2):197–204.CrossRef Lee YC, Bingham CO 3rd, Edwards RR, Marder W, Kristine P, Bolster MB, et al. Pain sensitization is associated with disease activity in rheumatoid arthritis patients: a cross-sectional study. Arthritis Care Res (Hoboken). 2018;70(2):197–204.CrossRef
52.
go back to reference Jones AKP, Huneke NTM, Lloyd D, Brown CA, Watson A. Role of functional brain imaging in understanding rheumatic pain. Curr Rheumatol Rep. 2012;14(6):557–67.CrossRefPubMed Jones AKP, Huneke NTM, Lloyd D, Brown CA, Watson A. Role of functional brain imaging in understanding rheumatic pain. Curr Rheumatol Rep. 2012;14(6):557–67.CrossRefPubMed
53.
go back to reference Harvey AK, Taylor AM, Wise RG. Imaging pain in arthritis: advances in structural and functional neuroimaging. Curr Pain Hadache Rep. 2012;16(6):492–501.CrossRef Harvey AK, Taylor AM, Wise RG. Imaging pain in arthritis: advances in structural and functional neuroimaging. Curr Pain Hadache Rep. 2012;16(6):492–501.CrossRef
54.
go back to reference Wartolowska K, Hough MG, Jenkinson M, Andersson J, Wordsworth BP, Tracey I. Structural changes of the brain in rheumatoid arthritis. Arthritis Rheum. 2012;64(2):371–9.CrossRefPubMed Wartolowska K, Hough MG, Jenkinson M, Andersson J, Wordsworth BP, Tracey I. Structural changes of the brain in rheumatoid arthritis. Arthritis Rheum. 2012;64(2):371–9.CrossRefPubMed
55.
go back to reference Chudler EH, Dong WK. The role of the basal ganglia in nociception and pain. Pain. 1995;60(1):3–38.CrossRefPubMed Chudler EH, Dong WK. The role of the basal ganglia in nociception and pain. Pain. 1995;60(1):3–38.CrossRefPubMed
56.
go back to reference Schweinhardt P, Kalk N, Wartolowska K, Chessell I, Wordsworth P, Tracey I. Investigation into the neural correlates of emotional augmentation of clinical pain. NeuroImage. 2008;40:759–66.CrossRefPubMed Schweinhardt P, Kalk N, Wartolowska K, Chessell I, Wordsworth P, Tracey I. Investigation into the neural correlates of emotional augmentation of clinical pain. NeuroImage. 2008;40:759–66.CrossRefPubMed
57.
go back to reference Phan KL, Wager T, Taylor SF, Liberzon I. Functional neuroanatomy of emotion: a meta-analysis of emotion activation studies in PET and fMRI. NeuroImage. 2002;16(2):331–48.CrossRefPubMed Phan KL, Wager T, Taylor SF, Liberzon I. Functional neuroanatomy of emotion: a meta-analysis of emotion activation studies in PET and fMRI. NeuroImage. 2002;16(2):331–48.CrossRefPubMed
58.
go back to reference Ridderinkhof KR, Ullsperger M, Crone EA, Nieuwenhuis S. The role of the medial frontal cortex in cognitive control. Science. 2004;306(5695):443–7.CrossRef Ridderinkhof KR, Ullsperger M, Crone EA, Nieuwenhuis S. The role of the medial frontal cortex in cognitive control. Science. 2004;306(5695):443–7.CrossRef
59.
go back to reference Hess A, Axmann R, Rech J, Finzel S, Heindl C, Kreitz S, et al. Blockade of TNF-alpha rapidly inhibits pain responses in the central nervous system. Proc Natl Acad Sci U S A. 2011;108(9):3731–6.CrossRefPubMedPubMedCentral Hess A, Axmann R, Rech J, Finzel S, Heindl C, Kreitz S, et al. Blockade of TNF-alpha rapidly inhibits pain responses in the central nervous system. Proc Natl Acad Sci U S A. 2011;108(9):3731–6.CrossRefPubMedPubMedCentral
60.
go back to reference Taylor PC, Keystone EC, vander Heijde D, Weinblatt ME, Del Carmen Morales L, Reyes Gonzaga J, et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl Med. 2017;376(7):652–62.CrossRef Taylor PC, Keystone EC, vander Heijde D, Weinblatt ME, Del Carmen Morales L, Reyes Gonzaga J, et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl Med. 2017;376(7):652–62.CrossRef
61.
go back to reference • Keystone EC, Taylor PC, Tanaka Y, Gaich C, De Lozier AM, Dudek A, et al. Patient reported outcomes from a phase 3 study of baricitinib versus placebo or adalimumab in rheumatoid arthritis: secondary analyses from the RA-BEAM study. Ann Rheum Dis. 2017;76(11):1853–61. This study reveals differences in the magnitude and rapidity of pain improvement among RA patients randomized to receive a janus kinase inhibitor vs. a TNF-α inhibitor. CrossRefPubMedPubMedCentral • Keystone EC, Taylor PC, Tanaka Y, Gaich C, De Lozier AM, Dudek A, et al. Patient reported outcomes from a phase 3 study of baricitinib versus placebo or adalimumab in rheumatoid arthritis: secondary analyses from the RA-BEAM study. Ann Rheum Dis. 2017;76(11):1853–61. This study reveals differences in the magnitude and rapidity of pain improvement among RA patients randomized to receive a janus kinase inhibitor vs. a TNF-α inhibitor. CrossRefPubMedPubMedCentral
63.
go back to reference Lee YC, Massarotti E, Edwards RR, Lu B, Liu C, Lo Y, et al. Effect of milnacipran on pain in patients with rheumatoid arthritis with widespread pain: a randomized blinded crossover trial. J Rheumatol. 2016;43(1):38–45.CrossRefPubMed Lee YC, Massarotti E, Edwards RR, Lu B, Liu C, Lo Y, et al. Effect of milnacipran on pain in patients with rheumatoid arthritis with widespread pain: a randomized blinded crossover trial. J Rheumatol. 2016;43(1):38–45.CrossRefPubMed
64.
go back to reference Gendreau RM, Thorn MD, Gendreau JF, Kranzler JD, Ribeiro S, Gracely RH, et al. Efficacy of milnacipran in patients with fibromyalgia. J Rheumatol. 2005;32(10):1975–85.PubMed Gendreau RM, Thorn MD, Gendreau JF, Kranzler JD, Ribeiro S, Gracely RH, et al. Efficacy of milnacipran in patients with fibromyalgia. J Rheumatol. 2005;32(10):1975–85.PubMed
65.
go back to reference Clauw DJ, Mease P, Palmer RH, Gendreau RM, Wang Y. Milnacipran for the treatment of fibromyalgia in adults: a 15-week, multicenter, randomized, double-blind, placebo-controlled, multiple dose clinical trial. Clin Ther. 2008;30(11):1988–2004.CrossRefPubMed Clauw DJ, Mease P, Palmer RH, Gendreau RM, Wang Y. Milnacipran for the treatment of fibromyalgia in adults: a 15-week, multicenter, randomized, double-blind, placebo-controlled, multiple dose clinical trial. Clin Ther. 2008;30(11):1988–2004.CrossRefPubMed
66.
go back to reference Mease PJ, Clauw DJ, Gendreau RM, Rao SG, Kranzler J, Chen W, et al. The efficacy and safety of milnacipran for treatment of fibromyalgia: a randomized, double-blind, placebo-controlled trial. J Rheumatol. 2009;36(2):398–409.CrossRefPubMed Mease PJ, Clauw DJ, Gendreau RM, Rao SG, Kranzler J, Chen W, et al. The efficacy and safety of milnacipran for treatment of fibromyalgia: a randomized, double-blind, placebo-controlled trial. J Rheumatol. 2009;36(2):398–409.CrossRefPubMed
Metadata
Title
Mechanisms for Joint Pain in Rheumatoid Arthritis (RA): from Cytokines to Central Sensitization
Authors
Angela Zhang
Yvonne C. Lee
Publication date
01-10-2018
Publisher
Springer US
Published in
Current Osteoporosis Reports / Issue 5/2018
Print ISSN: 1544-1873
Electronic ISSN: 1544-2241
DOI
https://doi.org/10.1007/s11914-018-0473-5

Other articles of this Issue 5/2018

Current Osteoporosis Reports 5/2018 Go to the issue

Cancer-induced Musculoskeletal Diseases (J Sterling and E Keller, Section Editors)

Interactions Between Disseminated Tumor Cells and Bone Marrow Stromal Cells Regulate Tumor Dormancy

Epidemiology and Pathophysiology (F Cosman and D Shoback, Section Editors)

Current Understanding of the Pathophysiology of Osteonecrosis of the Jaw

Epidemiology and Pathophysiology (F Cosman and D Shoback, Section Editors)

Vertebral Fracture Identification as Part of a Comprehensive Risk Assessment in Patients with Osteoporosis

Epidemiology and Pathophysiology (F Cosman and D Shoback, Section Editors)

The Influence of Cortical Porosity on the Strength of Bone During Growth and Advancing Age

Epidemiology and Pathophysiology (F Cosman and D Shoback, Section Editors)

The Effects of Homocysteine on the Skeleton