MicroRNA modulation in complex regional pain syndrome

Irina A Orlova, Guillermo M Alexander, Rehman A Qureshi, Ahmet Sacan, Alessandro Graziano, James E Barrett, Robert J Schwartzman, Seena K Ajit, Irina A Orlova, Guillermo M Alexander, Rehman A Qureshi, Ahmet Sacan, Alessandro Graziano, James E Barrett, Robert J Schwartzman, Seena K Ajit

Abstract

Background: Aberrant expression of small noncoding RNAs called microRNAs (miRNAs) is a common feature of several human diseases. The objective of the study was to identify miRNA modulation in patients with complex regional pain syndrome (CRPS) a chronic pain condition resulting from dysfunction in the central and/or peripheral nervous systems. Due to a multitude of inciting pathologies, symptoms and treatment conditions, the CRPS patient population is very heterogeneous. Our goal was to identify differentially expressed miRNAs in blood and explore their utility in patient stratification.

Methods: We profiled miRNAs in whole blood from 41 patients with CRPS and 20 controls using TaqMan low density array cards. Since neurogenic inflammation is known to play a significant role in CRPS we measured inflammatory markers including chemokines, cytokines, and their soluble receptors in blood from the same individuals. Correlation analyses were performed for miRNAs, inflammatory markers and other parameters including disease symptoms, medication, and comorbid conditions.

Results: Three different groups emerged from miRNA profiling. One group was comprised of 60% of CRPS patients and contained no control subjects. miRNA profiles from the remaining patients were interspersed among control samples in the other two groups. We identified differential expression of 18 miRNAs in CRPS patients. Analysis of inflammatory markers showed that vascular endothelial growth factor (VEGF), interleukin1 receptor antagonist (IL1Ra) and monocyte chemotactic protein-1 (MCP1) were significantly elevated in CRPS patients. VEGF and IL1Ra showed significant correlation with the patients reported pain levels. Analysis of the patients who were clustered according to their miRNA profile revealed correlations that were not significant in the total patient population. Correlation analysis of miRNAs detected in blood with additional parameters identified miRNAs associated with comorbidities such as headache, thyroid disorder and use of narcotics and antiepileptic drugs.

Conclusions: miRNA profiles can be useful in patient stratification and have utility as potential biomarkers for pain. Differentially expressed miRNAs can provide molecular insights into gene regulation and could lead to new therapeutic intervention strategies for CRPS.

Figures

Figure 1
Figure 1
Differential expression of miRNAs in blood from CRPS patients and control samples. A clustergram of the samples and miRNAs identifying sample subpopulations with respect to the significant differentially expressed miRNAs. A total of 60% of the patients (Group 3) showed differential regulation for these miRNAs (mostly downregulation), whereas the rest of the patients were more heterogeneous and clustered together with the control samples. P and C represent patients and controls, respectively. Red, high; black, average; green, low.
Figure 2
Figure 2
Elevation of inflammatory markers in CRPS patients. VEGF, IL1Ra, and MCP1 showed significant differential expression in CRPS patients vs. control samples, with p values 0.0002, 0.0004, and 0.0005, respectively. The error bars show the standard error of the means.
Figure 3
Figure 3
Circos diagram showing the correlation of selected parameters and miRNAs. The Circos diagram showing the correlation of selected medical conditions with other clinical parameters and miRNAs. The nodes along the circle are colored by the total strength of correlation of the corresponding variable, such that variables with many strong correlations are shown in red. The links between variables indicate their correlation. Only correlations with adjusted p value less than 0.01 are shown. Groups of miRNAs showing similar correlations with other variables are labeled as "miRNAs" for brevity. The links are colored by the Spearman's correlation value, with strong negative correlations shown in darker blue (e.g., narcotics vs. hsa-miR-191) and strong positive correlations shown in darker red (e.g., pain level vs. "IL1ra, VEGF, miRNAs"), as implicated by the color-bar. See the Supplementary table 1 for a complete list of correlations.

References

    1. Schwartzman RJ, Alexander GM, Grothusen J. Pathophysiology of complex regional pain syndrome. Expert Rev Neurother. 2006;6:669–681. doi: 10.1586/14737175.6.5.669.
    1. Schwartzman RJ, Erwin KL, Alexander GM. The natural history of complex regional pain syndrome. Clin J Pain. 2009;25:273–280. doi: 10.1097/AJP.0b013e31818ecea5.
    1. Costigan M, Scholz J, Woolf CJ. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci. 2009;32:1–32. doi: 10.1146/annurev.neuro.051508.135531.
    1. Maihofner C, Seifert F, Markovic K. Complex regional pain syndromes: new pathophysiological concepts and therapies. Eur J Neurol. 2010;17:649–660. doi: 10.1111/j.1468-1331.2010.02947.x.
    1. Harden RN, Bruehl S, Stanton-Hicks M, Wilson PR. Proposed new diagnostic criteria for complex regional pain syndrome. Pain Med. 2007;8:326–331. doi: 10.1111/j.1526-4637.2006.00169.x.
    1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002.
    1. Ceman S, Saugstad J. MicroRNAs: Meta-controllers of gene expression in synaptic activity emerge as genetic and diagnostic markers of human disease. Pharmacol Ther. 2011;130:26–37. doi: 10.1016/j.pharmthera.2011.01.004.
    1. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X. et al.Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18:997–1006. doi: 10.1038/cr.2008.282.
    1. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A. et al.Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105:10513–10518. doi: 10.1073/pnas.0804549105.
    1. Cortez MA, Calin GA. MicroRNA identification in plasma and serum: a new tool to diagnose and monitor diseases. Expert Opin Biol Ther. 2009;9:703–711. doi: 10.1517/14712590902932889.
    1. Park NJ, Zhou H, Elashoff D, Henson BS, Kastratovic DA, Abemayor E, Wong DT. Salivary microRNA: discovery, characterization, and clinical utility for oral cancer detection. Clin Cancer Res. 2009;15:5473–5477. doi: 10.1158/1078-0432.CCR-09-0736.
    1. Gilad S, Meiri E, Yogev Y, Benjamin S, Lebanony D, Yerushalmi N, Benjamin H, Kushnir M, Cholakh H, Melamed N. et al.Serum microRNAs are promising novel biomarkers. PLoS One. 2008;3:e3148. doi: 10.1371/journal.pone.0003148.
    1. Hanke M, Hoefig K, Merz H, Feller AC, Kausch I, Jocham D, Warnecke JM, Sczakiel G. A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer. Urol Oncol. 2009.
    1. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, Galas DJ, Wang K. The microRNA spectrum in 12 body fluids. Clin Chem. 2010;56:1733–1741. doi: 10.1373/clinchem.2010.147405.
    1. Etheridge A, Lee I, Hood L, Galas D, Wang K. Extracellular microRNA: A new source of biomarkers. Mutat Res. 2011.
    1. Scholer N, Langer C, Dohner H, Buske C, Kuchenbauer F. Serum microRNAs as a novel class of biomarkers: a comprehensive review of the literature. Exp Hematol. 2010;38:1126–1130. doi: 10.1016/j.exphem.2010.10.004.
    1. Wehling M. Translational medicine: can it really facilitate the transition of research "from bench to bedside"? Eur J Clin Pharmacol. 2006;62:91–95.
    1. Frank R, Hargreaves R. Clinical biomarkers in drug discovery and development. Nat Rev Drug Discov. 2003;2:566–580. doi: 10.1038/nrd1130.
    1. Bai G, Ambalavanar R, Wei D, Dessem D. Downregulation of selective microRNAs in trigeminal ganglion neurons following inflammatory muscle pain. Mol Pain. 2007;3:15. doi: 10.1186/1744-8069-3-15.
    1. Kusuda R, Cadetti F, Ravanelli MI, Sousa TA, Zanon S, De Lucca FL, Lucas G. Differential expression of microRNAs in mouse pain models. Mol Pain. 2011;7:17. doi: 10.1186/1744-8069-7-17.
    1. Poh KW, Yeo JF, Ong WY. MicroRNA changes in the mouse prefrontal cortex after inflammatory pain. Eur J Pain. 2011;15:801. e801-812.
    1. Aldrich BT, Frakes EP, Kasuya J, Hammond DL, Kitamoto T. Changes in expression of sensory organ-specific microRNAs in rat dorsal root ganglia in association with mechanical hypersensitivity induced by spinal nerve ligation. Neuroscience. 2009;164:711–723. doi: 10.1016/j.neuroscience.2009.08.033.
    1. von Schack D, Agostino MJ, Murray BS, Li Y, Reddy PS, Chen J, Choe SE, Strassle BW, Li C, Bates B. et al.Dynamic changes in the microRNA expression profile reveal multiple regulatory mechanisms in the spinal nerve ligation model of neuropathic pain. PLoS One. 2011;6:e17670. doi: 10.1371/journal.pone.0017670.
    1. Zhao J, Lee MC, Momin A, Cendan CM, Shepherd ST, Baker MD, Asante C, Bee L, Bethry A, Perkins JR. et al.Small RNAs control sodium channel expression, nociceptor excitability, and pain thresholds. J Neurosci. 2010;30:10860–10871. doi: 10.1523/JNEUROSCI.1980-10.2010.
    1. Favereaux A, Thoumine O, Bouali-Benazzouz R, Roques V, Papon MA, Salam SA, Drutel G, Leger C, Calas A, Nagy F, Landry M. Bidirectional integrative regulation of Cav1.2 calcium channel by microRNA miR-103: role in pain. EMBO J. 2011;30:3830–3841. doi: 10.1038/emboj.2011.249.
    1. Li X, Gibson G, Kim JS, Kroin J, Xu S, van Wijnen AJ, Im HJ. MicroRNA-146a is linked to pain-related pathophysiology of osteoarthritis. Gene. 2011;480:34–41. doi: 10.1016/j.gene.2011.03.003.
    1. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3:1101–1108. doi: 10.1038/nprot.2008.73.
    1. Hochberg YBaY. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society. 1995;57:289–300.
    1. Krzywinski M, Schein J, Birol I, Connors J, Gascoyne R, Horsman D, Jones SJ, Marra MA. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19:1639–1645. doi: 10.1101/gr.092759.109.
    1. Sethupathy P, Megraw M, Hatzigeorgiou AG. A guide through present computational approaches for the identification of mammalian microRNA targets. Nat Methods. 2006;3:881–886. doi: 10.1038/nmeth954.
    1. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20. doi: 10.1016/j.cell.2004.12.035.
    1. Gee HE, Buffa FM, Camps C, Ramachandran A, Leek R, Taylor M, Patil M, Sheldon H, Betts G, Homer J. et al.The small-nucleolar RNAs commonly used for microRNA normalisation correlate with tumour pathology and prognosis. Br J Cancer. 2011;104:1168–1177. doi: 10.1038/sj.bjc.6606076.
    1. Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT. GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 2009;28:195–208. doi: 10.1038/onc.2008.373.
    1. Williams GT, Mourtada-Maarabouni M, Farzaneh F. A critical role for non-coding RNA GAS5 in growth arrest and rapamycin inhibition in human T-lymphocytes. Biochem Soc Trans. 2011;39:482–486. doi: 10.1042/BST0390482.
    1. Schinkel C, Gaertner A, Zaspel J, Zedler S, Faist E, Schuermann M. Inflammatory mediators are altered in the acute phase of posttraumatic complex regional pain syndrome. Clin J Pain. 2006;22:235–239. doi: 10.1097/01.ajp.0000169669.70523.f0.
    1. Huygen FJ, De Bruijn AG, De Bruin MT, Groeneweg JG, Klein J, Zijlstra FJ. Evidence for local inflammation in complex regional pain syndrome type 1. Mediators Inflamm. 2002;11:47–51. doi: 10.1080/09629350210307.
    1. Marchand F, Perretti M, McMahon SB. Role of the immune system in chronic pain. Nat Rev Neurosci. 2005;6:521–532.
    1. Abbadie C. Chemokines, chemokine receptors and pain. Trends Immunol. 2005;26:529–534. doi: 10.1016/j.it.2005.08.001.
    1. Watkins LR, Maier SF. Glia: a novel drug discovery target for clinical pain. Nat Rev Drug Discov. 2003;2:973–985. doi: 10.1038/nrd1251.
    1. DeLeo JA, Yezierski RP. The role of neuroinflammation and neuroimmune activation in persistent pain. Pain. 2001;90:1–6. doi: 10.1016/S0304-3959(00)00490-5.
    1. Austin PJ, Moalem-Taylor G. The neuro-immune balance in neuropathic pain: involvement of inflammatory immune cells, immune-like glial cells and cytokines. J Neuroimmunol. 2010;229:26–50. doi: 10.1016/j.jneuroim.2010.08.013.
    1. Uceyler N, Eberle T, Rolke R, Birklein F, Sommer C. Differential expression patterns of cytokines in complex regional pain syndrome. Pain. 2007;132:195–205. doi: 10.1016/j.pain.2007.07.031.
    1. Uceyler N, Rogausch JP, Toyka KV, Sommer C. Differential expression of cytokines in painful and painless neuropathies. Neurology. 2007;69:42–49. doi: 10.1212/01.wnl.0000265062.92340.a5.
    1. Ritz BW, Alexander GM, Nogusa S, Perreault MJ, Peterlin BL, Grothusen JR, Schwartzman RJ. Elevated blood levels of inflammatory monocytes (CD14+ CD16+) in patients with complex regional pain syndrome. Clin Exp Immunol. 2011;164:108–117. doi: 10.1111/j.1365-2249.2010.04308.x.
    1. Alexander GM, Peterlin LB, Perreault MJ, Grothusen JR, Schwartzman RJ. Changes in plasma cytokines and their soluble receptors in complex regional pain syndrome. J Pain. 2011. in press .
    1. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol. 2011;13:423–433. doi: 10.1038/ncb2210.

Source: PubMed

3
Tilaa