Show simple item record

The Biology of Pain: Through the Rheumatology Lens

dc.contributor.authorSunzini, Flavia
dc.contributor.authorSchrepf, Andrew
dc.contributor.authorClauw, Daniel J.
dc.contributor.authorBasu, Neil
dc.date.accessioned2023-05-01T19:10:37Z
dc.date.available2024-06-01 15:10:36en
dc.date.available2023-05-01T19:10:37Z
dc.date.issued2023-05
dc.identifier.citationSunzini, Flavia; Schrepf, Andrew; Clauw, Daniel J.; Basu, Neil (2023). "The Biology of Pain: Through the Rheumatology Lens." Arthritis & Rheumatology 75(5): 650-660.
dc.identifier.issn2326-5191
dc.identifier.issn2326-5205
dc.identifier.urihttps://hdl.handle.net/2027.42/176267
dc.publisherWiley Periodicals, Inc.
dc.titleThe Biology of Pain: Through the Rheumatology Lens
dc.typeArticle
dc.rights.robotsIndexNoFollow
dc.subject.hlbsecondlevelRheumatology
dc.subject.hlbtoplevelHealth Sciences
dc.description.peerreviewedPeer Reviewed
dc.description.bitstreamurlhttp://deepblue.lib.umich.edu/bitstream/2027.42/176267/1/art42429.pdf
dc.description.bitstreamurlhttp://deepblue.lib.umich.edu/bitstream/2027.42/176267/2/art42429_am.pdf
dc.description.bitstreamurlhttp://deepblue.lib.umich.edu/bitstream/2027.42/176267/3/art42429-sup-0001-Disclosureform.pdf
dc.identifier.doi10.1002/art.42429
dc.identifier.sourceArthritis & Rheumatology
dc.identifier.citedreferenceStrand V, Tundia N, Bergman M, et al. Upadacitinib improves patient-reported outcomes vs placebo or adalimumab in patients with rheumatoid arthritis: results from SELECT-COMPARE. Rheumatology (Oxford) 2021; 60: 5583 – 94.
dc.identifier.citedreferenceWalsh DA, Mapp PI, Kelly S. Calcitonin gene-related peptide in the joint: contributions to pain and inflammation. Br J Clin Pharmacol 2015; 80: 965 – 78.
dc.identifier.citedreferencePezet S, McMahon SB. Neurotrophins: mediators and modulators of pain [review]. Annu Rev Neurosci 2006; 29: 507 – 38.
dc.identifier.citedreferenceJi RR, Berta T, Nedergaard M. Glia and pain: is chronic pain a gliopathy? Pain 2013; 154 Suppl: S10 – 28.
dc.identifier.citedreferenceGuo W, Wang H, Watanabe M, et al. Glial-cytokine-neuronal interactions underlying the mechanisms of persistent pain. J Neurosci Off J Soc Neurosci 2007; 27: 6006 – 18.
dc.identifier.citedreferenceWigerblad G, Bas DB, Fernades-Cerqueira C, et al. Autoantibodies to citrullinated proteins induce joint pain independent of inflammation via a chemokine-dependent mechanism. Ann Rheum Dis 2016; 75: 730 – 8.
dc.identifier.citedreferenceVargas-Caraveo A, Sayd A, Robledo-Montaña J, et al. Toll-like receptor 4 agonist and antagonist lipopolysaccharides modify innate immune response in rat brain circumventricular organs. J Neuroinflammation 2020; 17: 6.
dc.identifier.citedreferenceVanderwall AG, Milligan ED. Cytokines in pain: harnessing endogenous anti-inflammatory signaling for improved pain management. Front Immunol 2019; 10: 3009.
dc.identifier.citedreferenceKapás L, Hansen MK, Chang HY, et al. Vagotomy attenuates but does not prevent the somnogenic and febrile effects of lipopolysaccharide in rats. Am J Physiol-Regul Integr Comp Physiol 1998; 274: R406 – 11.
dc.identifier.citedreferenceDantzer R, O’Connor JC, Freund GG, et al. From inflammation to sickness and depression: when the immune system subjugates the brain [review]. Nat Rev Neurosci 2008; 9: 46 – 56.
dc.identifier.citedreferenceIshida M, Kuroiwa Y, Yoshida E, et al. Residual symptoms and disease burden among patients with rheumatoid arthritis in remission or low disease activity: a systematic literature review. Mod Rheumatol 2018; 28: 789 – 99.
dc.identifier.citedreferenceWolfe F. Fibromyalgianess [editorial]. Arthritis Rheum 2009; 61: 715 – 6.
dc.identifier.citedreferenceBasu N, Kaplan CM, Ichesco E, et al. Neurobiologic features of fibromyalgia are also present among rheumatoid arthritis patients. Arthritis Rheumatol 2018; 70: 1000 – 7.
dc.identifier.citedreferenceLee YC, Lu B, Edwards RR, et al. The role of sleep problems in central pain processing in rheumatoid arthritis. Arthritis Rheum 2013; 65: 59 – 68.
dc.identifier.citedreferenceLee YC, Bingham CO III, Edwards RR, et al. Association between pain sensitization and disease activity in patients with rheumatoid arthritis: a cross-sectional study. Arthritis Care Res (Hoboken) 2018; 70: 197 – 204.
dc.identifier.citedreferenceHeisler AC, Song J, Muhammad LN, et al. Association of dysregulated central pain processing and response to disease-modifying antirheumatic drug therapy in rheumatoid arthritis. Arthritis Rheumatol (Hoboken) 2020; 72: 2017 – 24.
dc.identifier.citedreferenceKaplan CM, Schrepf A, Ichesco E, et al. Association of inflammation with pronociceptive brain connections in rheumatoid arthritis patients with concomitant fibromyalgia. Arthritis Rheumatol (Hoboken) 2020; 72: 41 – 6.
dc.identifier.citedreferenceErickson MA, Liang WS, Fernandez EG, et al. Genetics and sex influence peripheral and central innate immune responses and blood-brain barrier integrity. PLoS ONE 2018; 13: e0205769.
dc.identifier.citedreferenceMills SE, Nicolson KP, Smith BH. Chronic pain: a review of its epidemiology and associated factors in population-based studies. Br J Anaesth 2019; 123: e273 – 83.
dc.identifier.citedreferenceBreivik H, Collett B, Ventafridda V, et al. Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain Lond Engl 2006; 10: 287 – 333.
dc.identifier.citedreferenceTsang A, Von Korff M, Lee S, et al. Common chronic pain conditions in developed and developing countries: gender and age differences and comorbidity with depression-anxiety disorders. J Pain 2008; 9: 883 – 91.
dc.identifier.citedreferenceSmolen JS, Aletaha D, McInnes IB. Rheumatoid arthritis. Lancet Lond Engl 2016; 388: 2023 – 38.
dc.identifier.citedreferenceTaylor P, Manger B, Alvaro-Gracia J, et al. Patient perceptions concerning pain management in the treatment of rheumatoid arthritis. J Int Med Res 2010; 38: 1213 – 24.
dc.identifier.citedreferenceLee YC. Effect and treatment of chronic pain in inflammatory arthritis. Curr Rheumatol Rep 2013; 15: 300.
dc.identifier.citedreferenceInternational Association for the Study of Pain. Terminology; 2021. URL: https://www.iasp-pain.org/Education/Content.aspx?ItemNumber=1698#Pain.
dc.identifier.citedreferenceZhang S, Malik Sharif S, et al. Clinical features for diagnosis and management of patients with PRDM12 congenital insensitivity to pain. J Med Genet 2016; 53: 533 – 5.
dc.identifier.citedreferenceFitzcharles MA, Cohen SP, Clauw DJ, et al. Nociplastic pain: towards an understanding of prevalent pain conditions. Lancet Lond Engl 2021; 397: 2098 – 110.
dc.identifier.citedreferenceKosek E, Clauw D, Nijs J, et al. Chronic nociplastic pain affecting the musculoskeletal system: clinical criteria and grading system. PAIN 2021; 162: 2629 – 34.
dc.identifier.citedreferenceSmallwood RF, Laird AR, Ramage AE, et al. Structural brain anomalies and chronic pain: a quantitative meta-analysis of gray matter volume. J Pain 2013; 14: 663 – 75.
dc.identifier.citedreferenceWolfe F, Michaud K, Busch RE, et al. Polysymptomatic distress in patients with rheumatoid arthritis: understanding disproportionate response and its spectrum. Arthritis Care Res (Hoboken) 2014; 66: 1465 – 71.
dc.identifier.citedreferenceWoolf CJ, Ma Q. Nociceptors—noxious stimulus detectors. Neuron 2007; 55: 353 – 64.
dc.identifier.citedreferenceKras JV, Weisshaar CL, Pall PS, et al. Pain from intra-articular NGF or joint injury in the rat requires contributions from peptidergic joint afferents. Neurosci Lett 2015; 604: 193 – 8.
dc.identifier.citedreferenceVan den Broeke EN, van Rijn CM, Biurrun Manresa JA, et al. Neurophysiological correlates of nociceptive heterosynaptic long-term potentiation in humans. J Neurophysiol 2010; 103: 2107 – 13.
dc.identifier.citedreferenceLatremoliere A, Woolf CJ. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain 2009; 10: 895 – 926.
dc.identifier.citedreferenceMartínez-Lavín M. Dorsal root ganglia: fibromyalgia pain factory? Clin Rheumatol 2021; 1 – 5.
dc.identifier.citedreferenceCruz-Almeida Y, Fillingim RB. Can quantitative sensory testing move us closer to mechanism-based pain management? Pain Med Malden Mass 2014; 15: 61 – 72.
dc.identifier.citedreferenceGeisser ME, Donnell CS, Petzke F, et al. Comorbid somatic symptoms and functional status in patients with fibromyalgia and chronic fatigue syndrome: sensory amplification as a common mechanism. Psychosomatics 2008; 49: 235 – 42.
dc.identifier.citedreferenceLópez-Solà M, Woo CW, Pujol J, et al. Towards a neurophysiological signature for fibromyalgia. Pain 2017; 158: 34 – 47.
dc.identifier.citedreferenceLarkin TE, Kaplan CM, Schrepf A, et al. Altered network architecture of functional brain communities in chronic nociplastic pain. NeuroImage. 2021; 226: 117504.
dc.identifier.citedreferenceTodd AJ. Neuronal circuitry for pain processing in the dorsal horn [review]. Nat Rev Neurosci 2010; 11: 823 – 36.
dc.identifier.citedreferenceAl-Khater KM, Todd AJ. Collateral projections of neurons in laminae I, III, and IV of rat spinal cord to thalamus, periaqueductal gray matter, and lateral parabrachial area. J Comp Neurol 2009; 515: 629 – 46.
dc.identifier.citedreferenceDum RP, Levinthal DJ, Strick PL. The spinothalamic system targets motor and sensory areas in the cerebral cortex of monkeys. J Neurosci Off J Soc Neurosci 2009; 29: 14223 – 35.
dc.identifier.citedreferenceBernard JF, Alden M, Besson JM. The organization of the efferent projections from the pontine parabrachial area to the amygdaloid complex: a Phaseolus vulgaris leucoagglutinin (PHA-L) study in the rat. J Comp Neurol 1993; 329: 201 – 29.
dc.identifier.citedreferenceJensen KB, Regenbogen C, Ohse MC, et al. Brain activations during pain: a neuroimaging meta-analysis of patients with pain and healthy controls. Pain 2016; 157: 1279 – 86.
dc.identifier.citedreferenceApkarian VA, Hashmi JA, Baliki MN. Pain and the brain: specificity and plasticity of the brain in clinical chronic pain. Pain 2011; 152 Suppl: S49 – 64.
dc.identifier.citedreferenceBorsook D, Becerra LR. Breaking down the barriers: fMRI applications in pain, analgesia and analgesics. Mol Pain 2006; 2: 30.
dc.identifier.citedreferenceDehghan M, Schmidt-Wilcke T, Pfleiderer B, et al. Coordinate-based (ALE) meta-analysis of brain activation in patients with fibromyalgia. Hum Brain Mapp 2016; 37: 1749 – 58.
dc.identifier.citedreferenceLieberman MD, Eisenberger NI. The dorsal anterior cingulate cortex is selective for pain: results from large-scale reverse inference. Proc Natl Acad Sci U S A 2015; 112: 15250 – 5.
dc.identifier.citedreferenceMetzger CD, Eckert U, Steiner J, et al. High field FMRI reveals thalamocortical integration of segregated cognitive and emotional processing in mediodorsal and intralaminar thalamic nuclei. Front Neuroanat 2010; 4: 138.
dc.identifier.citedreferenceWager TD, Atlas LY, Lindquist MA, et al. An fMRI-based neurologic signature of physical pain. N Engl J Med 2013; 368: 1388 – 97.
dc.identifier.citedreferenceGiesecke T, Gracely RH, Williams DA, et al. The relationship between depression, clinical pain, and experimental pain in a chronic pain cohort. Arthritis Rheum 2005; 52: 1577 – 84.
dc.identifier.citedreferenceSegerdahl AR, Mezue M, Okell TW, et al. The dorsal posterior insula subserves a fundamental role in human pain. Nat Neurosci 2015; 18: 499 – 500.
dc.identifier.citedreferenceJiang Y, Oathes D, Hush J, et al. Perturbed connectivity of the amygdala and its subregions with the central executive and default mode networks in chronic pain. Pain 2016; 157: 1970 – 8.
dc.identifier.citedreferenceGracely RH, Geisser ME, Giesecke T, et al. Pain catastrophizing and neural responses to pain among persons with fibromyalgia. Brain J Neurol 2004; 127: 835 – 43.
dc.identifier.citedreferenceVachon-Presseau E, Tétreault P, Petre B, et al. Corticolimbic anatomical characteristics predetermine risk for chronic pain. Brain J Neurol 2016; 139: 1958 – 70.
dc.identifier.citedreferenceBolwerk A, Seifert F, Maihöfner C. Altered resting-state functional connectivity in complex regional pain syndrome. J Pain 2013; 14: 1107 – 15.
dc.identifier.citedreferenceLetzen JE, Robinson ME. Negative mood influences default mode network functional connectivity in patients with chronic low back pain: implications for functional neuroimaging biomarkers. Pain 2017; 158: 48 – 57.
dc.identifier.citedreferenceLoggia ML, Kim J, Gollub RL, et al. Default mode network connectivity encodes clinical pain: An arterial spin labeling study. PAIN 2013; 154: 24 – 33.
dc.identifier.citedreferenceBaliki MN, Apkarian AV. Nociception, pain, negative moods, and behavior selection. Neuron 2015; 87: 474 – 91.
dc.identifier.citedreferenceJoharatnam N, McWilliams DF, Wilson D, et al. A cross-sectional study of pain sensitivity, disease-activity assessment, mental health, and fibromyalgia status in rheumatoid arthritis. Arthritis Res Ther 2015; 17: 11.
dc.identifier.citedreferenceHeisler AC, Song J, Dunlop DD, et al. Association of pain centralization and patient-reported pain in active rheumatoid arthritis. Arthritis Care Res (Hoboken) 2020; 72: 1122 – 9.
dc.identifier.citedreferenceSchrepf A, Kaplan CM, Ichesco E, et al. A multi-modal MRI study of the central response to inflammation in rheumatoid arthritis. Nat Commun 2018; 9: 2243.
dc.identifier.citedreferenceFoerster BR, Petrou M, Edden RA, et al. Reduced insular γ-aminobutyric acid in fibromyalgia. Arthritis Rheum 2012; 64: 579 – 83.
dc.identifier.citedreferencePyke TL, Osmotherly PG, Baines S. Measuring glutamate levels in the brains of fibromyalgia patients and a potential role for glutamate in the pathophysiology of fibromyalgia symptoms: a systematic review. Clin J Pain 2017; 33: 944 – 54.
dc.identifier.citedreferenceEmmer BJ, van der Bijl AE, Huizinga TW, et al. Brain involvement in rheumatoid arthritis: a magnetic resonance spectroscopy study. Arthritis Rheum 2009; 60: 3190 – 5.
dc.identifier.citedreferenceJensen KB, Loitoile R, Kosek E, et al. Patients with fibromyalgia display less functional connectivity in the brain’s pain inhibitory network. Mol Pain 2012; 8: 32.
dc.identifier.citedreferenceOssipov MH, Morimura K, Porreca F. Descending pain modulation and chronification of pain. Curr Opin Support Palliat Care 2014; 8: 143 – 51.
dc.identifier.citedreferenceYarnitsky D. Conditioned pain modulation (the diffuse noxious inhibitory control-like effect): its relevance for acute and chronic pain states. Curr Opin Anesthesiol 2010; 23: 611 – 5.
dc.identifier.citedreferenceLewis GN, Rice DA, McNair PJ. Conditioned pain modulation in populations with chronic pain: a systematic review and meta-analysis. J Pain 2012; 13: 936 – 44.
dc.identifier.citedreferenceJensen KB, Kosek E, Petzke F, et al. Evidence of dysfunctional pain inhibition in fibromyalgia reflected in rACC during provoked pain. Pain 2009; 144: 95 – 100.
dc.identifier.citedreferenceCui M, Feng Y, McAdoo DJ, et al. Periaqueductal gray stimulation-induced inhibition of nociceptive dorsal horn neurons in rats is associated with the release of norepinephrine, serotonin, and amino acids. J Pharmacol Exp Ther 1999; 289: 868 – 76.
dc.identifier.citedreferenceHarper DE, Ichesco E, Schrepf A, et al. Resting functional connectivity of the periaqueductal gray is associated with normal inhibition and pathological facilitation in conditioned pain modulation. J Pain 2018; 19: 635. e1 – 15.
dc.identifier.citedreferenceKato G, Yasaka T, Katafuchi T, et al. Direct GABAergic and glycinergic inhibition of the substantia gelatinosa from the rostral ventromedial medulla revealed by in vivo patch-clamp analysis in rats. J Neurosci Off J Soc Neurosci 2006; 26: 1787 – 94.
dc.identifier.citedreferenceWang YY, Wu SX, Liu XY, et al. Effects of c-fos antisense oligodeoxynucleotide on 5-HT-induced upregulation of preprodynorphin, preproenkephalin, and glutamic acid decarboxylase mRNA expression in cultured rat spinal dorsal horn neurons. Biochem Biophys Res Commun 2003; 309: 631 – 6.
dc.identifier.citedreferenceHosobuchi Y, Adams JE, Linchitz R. Pain relief by electrical stimulation of the central gray matter in humans and its reversal by naloxone. Science 1977; 197: 183 – 6.
dc.identifier.citedreferenceHarris RE, Clauw DJ, Scott DJ, et al. Decreased central μ-opioid receptor availability in fibromyalgia. J Neurosci 2007; 27: 10000 – 6.
dc.identifier.citedreferenceSchrepf A, Harper DE, Harte SE, et al. Endogenous opioidergic dysregulation of pain in fibromyalgia: a PET and fMRI study. Pain 2016; 157: 2217 – 25.
dc.identifier.citedreferenceBajic D, Proudfit HK. Projections of neurons in the periaqueductal gray to pontine and medullary catecholamine cell groups involved in the modulation of nociception. J Comp Neurol 1999; 405: 359 – 79.
dc.identifier.citedreferenceMcWilliams DF, Walsh DA. Pain mechanisms in rheumatoid arthritis. Clin Exp Rheumatol 2017; 35 Suppl: 94 – 101.
dc.identifier.citedreferenceGlinatsi D, Brahe CH, Hetland ML, et al. Association between MRI findings and patient-reported outcomes in patients with rheumatoid arthritis in clinical remission and at relapse. Int J Rheum Dis 2020; 23: 488 – 98.
dc.identifier.citedreferenceJansen JP, Buckley F, Dejonckheere F, et al. Comparative efficacy of biologics as monotherapy and in combination with methotrexate on patient reported outcomes (PROs) in rheumatoid arthritis patients with an inadequate response to conventional DMARDs—a systematic review and network meta-analysis. Health Qual Life Outcomes 2014; 12: 102.
dc.identifier.citedreferenceTaylor PC, Keystone EC, van der Heijde D, et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med 2017; 376: 652 – 62.
dc.identifier.citedreferenceKeystone EC, Taylor PC, Tanaka Y, 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: 1853 – 61.
dc.identifier.citedreferenceBergman M, Tundia N, Martin N, et al. Patient-reported outcomes of upadacitinib versus abatacept in patients with rheumatoid arthritis and an inadequate response to biologic disease-modifying antirheumatic drugs: 12- and 24-week results of a phase 3 trial. Arthritis Res Ther 2022; 24: 155.
dc.identifier.citedreferenceSchaible HG. Nociceptive neurons detect cytokines in arthritis. Arthritis Res Ther 2014; 16: 470.
dc.working.doiNOen
dc.owningcollnameInterdisciplinary and Peer-Reviewed


Files in this item

Show simple item record

Remediation of Harmful Language

The University of Michigan Library aims to describe library materials in a way that respects the people and communities who create, use, and are represented in our collections. Report harmful or offensive language in catalog records, finding aids, or elsewhere in our collections anonymously through our metadata feedback form. More information at Remediation of Harmful Language.

Accessibility

If you are unable to use this file in its current format, please select the Contact Us link and we can modify it to make it more accessible to you.