Skip to main content
Top
Published in: Molecular Pain 1/2011

Open Access 01-12-2011 | Research

Differential expression of microRNAs in mouse pain models

Authors: Ricardo Kusuda, Flaviane Cadetti, Maria I Ravanelli, Thais A Sousa, Sônia Zanon, Fernando L De Lucca, Guilherme Lucas

Published in: Molecular Pain | Issue 1/2011

Login to get access

Abstract

Background

MicroRNAs (miRNAs) are short non-coding RNAs that inhibit translation of target genes by binding to their mRNAs. The expression of numerous brain-specific miRNAs with a high degree of temporal and spatial specificity suggests that miRNAs play an important role in gene regulation in health and disease. Here we investigate the time course gene expression profile of miR-1, -16, and -206 in mouse dorsal root ganglion (DRG), and spinal cord dorsal horn under inflammatory and neuropathic pain conditions as well as following acute noxious stimulation.

Results

Quantitative real-time polymerase chain reaction analyses showed that the mature form of miR-1, -16 and -206, is expressed in DRG and the dorsal horn of the spinal cord. Moreover, CFA-induced inflammation significantly reduced miRs-1 and -16 expression in DRG whereas miR-206 was downregulated in a time dependent manner. Conversely, in the spinal dorsal horn all three miRNAs monitored were upregulated. After sciatic nerve partial ligation, miR-1 and -206 were downregulated in DRG with no change in the spinal dorsal horn. On the other hand, axotomy increases the relative expression of miR-1, -16, and 206 in a time-dependent fashion while in the dorsal horn there was a significant downregulation of miR-1. Acute noxious stimulation with capsaicin also increased the expression of miR-1 and -16 in DRG cells but, on the other hand, in the spinal dorsal horn only a high dose of capsaicin was able to downregulate miR-206 expression.

Conclusions

Our results indicate that miRNAs may participate in the regulatory mechanisms of genes associated with the pathophysiology of chronic pain as well as the nociceptive processing following acute noxious stimulation. We found substantial evidence that miRNAs are differentially regulated in DRG and the dorsal horn of the spinal cord under different pain states. Therefore, miRNA expression in the nociceptive system shows not only temporal and spatial specificity but is also stimulus-dependent.
Appendix
Available only for authorised users
Literature
1.
go back to reference Bartel DP: MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004, 116: 281–297. 10.1016/S0092-8674(04)00045-5PubMedCrossRef Bartel DP: MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004, 116: 281–297. 10.1016/S0092-8674(04)00045-5PubMedCrossRef
2.
go back to reference He L, Hannon GJ: MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet 2004, 5: 522–531. 10.1038/nrg1379PubMedCrossRef He L, Hannon GJ: MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet 2004, 5: 522–531. 10.1038/nrg1379PubMedCrossRef
3.
go back to reference Kosik KS, Krichevsky AM: The Elegance of the MicroRNAs: A Neuronal Perspective. Neuron 2005, 47: 779–782. 10.1016/j.neuron.2005.08.019PubMedCrossRef Kosik KS, Krichevsky AM: The Elegance of the MicroRNAs: A Neuronal Perspective. Neuron 2005, 47: 779–782. 10.1016/j.neuron.2005.08.019PubMedCrossRef
4.
go back to reference Mattick JS, Makunin IV: Small regulatory RNAs in mammals. Hum Mol Genet 2005,14(1):R121–132. 10.1093/hmg/ddi101PubMedCrossRef Mattick JS, Makunin IV: Small regulatory RNAs in mammals. Hum Mol Genet 2005,14(1):R121–132. 10.1093/hmg/ddi101PubMedCrossRef
6.
go back to reference Lee RC, Feinbaum RL, Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75: 843–854. 10.1016/0092-8674(93)90529-YPubMedCrossRef Lee RC, Feinbaum RL, Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75: 843–854. 10.1016/0092-8674(93)90529-YPubMedCrossRef
7.
go back to reference Ambros V: microRNAs: tiny regulators with great potential. Cell 2001, 107: 823–826. 10.1016/S0092-8674(01)00616-XPubMedCrossRef Ambros V: microRNAs: tiny regulators with great potential. Cell 2001, 107: 823–826. 10.1016/S0092-8674(01)00616-XPubMedCrossRef
8.
go back to reference Du T, Zamore PD: microPrimer: the biogenesis and function of microRNA. Development 2005, 132: 4645–4652. 10.1242/dev.02070PubMedCrossRef Du T, Zamore PD: microPrimer: the biogenesis and function of microRNA. Development 2005, 132: 4645–4652. 10.1242/dev.02070PubMedCrossRef
9.
go back to reference Kim VN: MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol 2005, 6: 376–385. 10.1038/nrm1644PubMedCrossRef Kim VN: MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol 2005, 6: 376–385. 10.1038/nrm1644PubMedCrossRef
10.
go back to reference Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T: Identification of tissue-specific microRNAs from mouse. Curr Biol 2002, 12: 735–739. 10.1016/S0960-9822(02)00809-6PubMedCrossRef Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T: Identification of tissue-specific microRNAs from mouse. Curr Biol 2002, 12: 735–739. 10.1016/S0960-9822(02)00809-6PubMedCrossRef
11.
go back to reference Schratt GM, Tuebing F, Nigh EA, Kane CG, Sabatini ME, Kiebler M, Greenberg ME: A brain-specific microRNA regulates dendritic spine development. Nature 2006, 439: 283–289. 10.1038/nature04367PubMedCrossRef Schratt GM, Tuebing F, Nigh EA, Kane CG, Sabatini ME, Kiebler M, Greenberg ME: A brain-specific microRNA regulates dendritic spine development. Nature 2006, 439: 283–289. 10.1038/nature04367PubMedCrossRef
12.
go back to reference Chen CZ, Li L, Lodish HF, Bartel DP: MicroRNAs modulate hematopoietic lineage differentiation. Science 2004, 303: 83–86. 10.1126/science.1091903PubMedCrossRef Chen CZ, Li L, Lodish HF, Bartel DP: MicroRNAs modulate hematopoietic lineage differentiation. Science 2004, 303: 83–86. 10.1126/science.1091903PubMedCrossRef
13.
go back to reference Kim HK, Lee YS, Sivaprasad U, Malhotra A, Dutta A: Muscle-specific microRNA miR-206 promotes muscle differentiation. J Cell Biol 2006, 174: 677–687. 10.1083/jcb.200603008PubMedCentralPubMedCrossRef Kim HK, Lee YS, Sivaprasad U, Malhotra A, Dutta A: Muscle-specific microRNA miR-206 promotes muscle differentiation. J Cell Biol 2006, 174: 677–687. 10.1083/jcb.200603008PubMedCentralPubMedCrossRef
14.
go back to reference Lee HJ, Palkovits M, Young WS: miR-7b, a microRNA up-regulated in the hypothalamus after chronic hyperosmolar stimulation, inhibits Fos translation. Proc Natl Acad Sci USA 2006, 103: 15669–15674. 10.1073/pnas.0605781103PubMedCentralPubMedCrossRef Lee HJ, Palkovits M, Young WS: miR-7b, a microRNA up-regulated in the hypothalamus after chronic hyperosmolar stimulation, inhibits Fos translation. Proc Natl Acad Sci USA 2006, 103: 15669–15674. 10.1073/pnas.0605781103PubMedCentralPubMedCrossRef
15.
go back to reference Li Y, Wang F, Lee JA, Gao FB: MicroRNA-9a ensures the precise specification of sensory organ precursors in Drosophila. Genes Dev 2006, 20: 2793–2805. 10.1101/gad.1466306PubMedCentralPubMedCrossRef Li Y, Wang F, Lee JA, Gao FB: MicroRNA-9a ensures the precise specification of sensory organ precursors in Drosophila. Genes Dev 2006, 20: 2793–2805. 10.1101/gad.1466306PubMedCentralPubMedCrossRef
16.
go back to reference Yang B, Lin H, Xiao J, Lu Y, Luo X, Li B, Zhang Y, Xu C, Bai Y, Wang H, Chen G, Wang Z: The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med 2007, 13: 486–491. 10.1038/nm1569PubMedCrossRef Yang B, Lin H, Xiao J, Lu Y, Luo X, Li B, Zhang Y, Xu C, Bai Y, Wang H, Chen G, Wang Z: The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med 2007, 13: 486–491. 10.1038/nm1569PubMedCrossRef
17.
go back to reference Lee YS, Dutta A: MicroRNAs: small but potent oncogenes or tumor suppressors. Curr Opin Investig Drugs 2006, 7: 560–564.PubMed Lee YS, Dutta A: MicroRNAs: small but potent oncogenes or tumor suppressors. Curr Opin Investig Drugs 2006, 7: 560–564.PubMed
18.
go back to reference Bai G, Ambalavanar R, Wei D, Dessem D: Downregulation of selective microRNAs in trigeminal ganglion neurons following inflammatory muscle pain. Mol Pain 2007, 3: 15–18. 10.1186/1744-8069-3-15PubMedCentralPubMedCrossRef Bai G, Ambalavanar R, Wei D, Dessem D: Downregulation of selective microRNAs in trigeminal ganglion neurons following inflammatory muscle pain. Mol Pain 2007, 3: 15–18. 10.1186/1744-8069-3-15PubMedCentralPubMedCrossRef
19.
go back to reference 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: 711–23. 10.1016/j.neuroscience.2009.08.033PubMedCentralPubMedCrossRef 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: 711–23. 10.1016/j.neuroscience.2009.08.033PubMedCentralPubMedCrossRef
20.
go back to reference Zhao J, Lee MC, Momin A, Cendan CM, Shepherd ST, Baker MD, Asante C, Bee L, Bethry A, Perkins JR, Nassar MA, Abrahamsen B, Dickenson A, Cobb BS, Merkenschlager M, Wood JN: Small RNAs control sodium channel expression, nociceptor excitability, and pain thresholds. J Neurosci 2010, 30: 10860–10871. 10.1523/JNEUROSCI.1980-10.2010PubMedCrossRef Zhao J, Lee MC, Momin A, Cendan CM, Shepherd ST, Baker MD, Asante C, Bee L, Bethry A, Perkins JR, Nassar MA, Abrahamsen B, Dickenson A, Cobb BS, Merkenschlager M, Wood JN: Small RNAs control sodium channel expression, nociceptor excitability, and pain thresholds. J Neurosci 2010, 30: 10860–10871. 10.1523/JNEUROSCI.1980-10.2010PubMedCrossRef
21.
go back to reference Hokfelt T, Zhang X, Wiesenfeld-Hallin Z: Messenger plasticity in primary sensory neurons following axotomy and its functional implications. Trends Neurosci 1994, 17: 22–30. 10.1016/0166-2236(94)90031-0PubMedCrossRef Hokfelt T, Zhang X, Wiesenfeld-Hallin Z: Messenger plasticity in primary sensory neurons following axotomy and its functional implications. Trends Neurosci 1994, 17: 22–30. 10.1016/0166-2236(94)90031-0PubMedCrossRef
22.
go back to reference Honore P, Rogers SD, Schwei MJ, Salak-Johnson JL, Luger NM, Sabino MC, Clohisy DR, Mantyh PW: Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neuroscience 2000, 98: 585–598. 10.1016/S0306-4522(00)00110-XPubMedCrossRef Honore P, Rogers SD, Schwei MJ, Salak-Johnson JL, Luger NM, Sabino MC, Clohisy DR, Mantyh PW: Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neuroscience 2000, 98: 585–598. 10.1016/S0306-4522(00)00110-XPubMedCrossRef
23.
go back to reference Honore P, Schwei J, Rogers SD, Salak-Johnson JL, Finke MP, Ramnaraine ML, Clohisy DR, Mantyh PW: Cellular and neurochemical remodeling of the spinal cord in bone cancer pain. Prog Brain Res 2000, 129: 389–397. full_textPubMedCrossRef Honore P, Schwei J, Rogers SD, Salak-Johnson JL, Finke MP, Ramnaraine ML, Clohisy DR, Mantyh PW: Cellular and neurochemical remodeling of the spinal cord in bone cancer pain. Prog Brain Res 2000, 129: 389–397. full_textPubMedCrossRef
24.
go back to reference Kim SK, Nam JW, Rhee JK, Lee WJ, Zhang BT: miTarget: microRNA target gene prediction using a support vector machine. BMC Bioinformatics 2006, 7: 411. 10.1186/1471-2105-7-411PubMedCentralPubMedCrossRef Kim SK, Nam JW, Rhee JK, Lee WJ, Zhang BT: miTarget: microRNA target gene prediction using a support vector machine. BMC Bioinformatics 2006, 7: 411. 10.1186/1471-2105-7-411PubMedCentralPubMedCrossRef
25.
go back to reference Rodriguez Parkitna J, Korostynski M, Kaminska-Chowaniec D, Obara I, Mika J, Przewlocka B, Przewlocki R: Comparison of gene expression profiles in neuropathic and inflammatory pain. J Physiol Pharmacol 2006, 57: 401–414.PubMed Rodriguez Parkitna J, Korostynski M, Kaminska-Chowaniec D, Obara I, Mika J, Przewlocka B, Przewlocki R: Comparison of gene expression profiles in neuropathic and inflammatory pain. J Physiol Pharmacol 2006, 57: 401–414.PubMed
26.
27.
go back to reference Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M, Rajewsky N: Combinatorial microRNA target predictions. Nat Genet 2005, 37: 495–500. 10.1038/ng1536PubMedCrossRef Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M, Rajewsky N: Combinatorial microRNA target predictions. Nat Genet 2005, 37: 495–500. 10.1038/ng1536PubMedCrossRef
28.
go back to reference Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB: Prediction of mammalian microRNA targets. Cell 2003, 115: 787–798. 10.1016/S0092-8674(03)01018-3PubMedCrossRef Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB: Prediction of mammalian microRNA targets. Cell 2003, 115: 787–798. 10.1016/S0092-8674(03)01018-3PubMedCrossRef
29.
go back to reference Zimmermann M: Ethical guidelines for investigations of experimental pain in conscious animals. Pain 1983, 16: 109–110. 10.1016/0304-3959(83)90201-4PubMedCrossRef Zimmermann M: Ethical guidelines for investigations of experimental pain in conscious animals. Pain 1983, 16: 109–110. 10.1016/0304-3959(83)90201-4PubMedCrossRef
30.
go back to reference Seltzer Z, Dubner R, Shir Y: A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990, 43: 205–218. 10.1016/0304-3959(90)91074-SPubMedCrossRef Seltzer Z, Dubner R, Shir Y: A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990, 43: 205–218. 10.1016/0304-3959(90)91074-SPubMedCrossRef
31.
go back to reference Dixon WJ: Efficient analysis of experimental observations. Annu Rev Pharmacol Toxicol 1980, 20: 441–462. 10.1146/annurev.pa.20.040180.002301PubMedCrossRef Dixon WJ: Efficient analysis of experimental observations. Annu Rev Pharmacol Toxicol 1980, 20: 441–462. 10.1146/annurev.pa.20.040180.002301PubMedCrossRef
32.
go back to reference Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL: Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 1994, 53: 55–63. 10.1016/0165-0270(94)90144-9PubMedCrossRef Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL: Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods 1994, 53: 55–63. 10.1016/0165-0270(94)90144-9PubMedCrossRef
33.
go back to reference Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25: 402–408. 10.1006/meth.2001.1262PubMedCrossRef Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25: 402–408. 10.1006/meth.2001.1262PubMedCrossRef
34.
go back to reference Alvares D, Fitzgerald M: Building blocks of pain: the regulation of key molecules in spinal sensory neurones during development and following peripheral axotomy. Pain 1999,6(Suppl):S71–85. 10.1016/S0304-3959(99)00140-2PubMedCrossRef Alvares D, Fitzgerald M: Building blocks of pain: the regulation of key molecules in spinal sensory neurones during development and following peripheral axotomy. Pain 1999,6(Suppl):S71–85. 10.1016/S0304-3959(99)00140-2PubMedCrossRef
35.
go back to reference Woolf CJ: Phenotypic modification of primary sensory neurons: the role of nerve growth factor in the production of persistent pain. Philos Trans R Soc Lond B Biol Sci 1996, 351: 441–448. 10.1098/rstb.1996.0040PubMedCrossRef Woolf CJ: Phenotypic modification of primary sensory neurons: the role of nerve growth factor in the production of persistent pain. Philos Trans R Soc Lond B Biol Sci 1996, 351: 441–448. 10.1098/rstb.1996.0040PubMedCrossRef
36.
go back to reference Mannion RJ, Costigan M, Decosterd I, Amaya F, Ma QP, Holstege JC, Ji RR, Acheson A, Lindsay RM, Wilkinson GA, Woolf CJ: Neurotrophins: peripherally and centrally acting modulators of tactile stimulus-induced inflammatory pain hypersensitivity. Proc Natl Acad Sci USA 1999, 96: 9385–9390. 10.1073/pnas.96.16.9385PubMedCentralPubMedCrossRef Mannion RJ, Costigan M, Decosterd I, Amaya F, Ma QP, Holstege JC, Ji RR, Acheson A, Lindsay RM, Wilkinson GA, Woolf CJ: Neurotrophins: peripherally and centrally acting modulators of tactile stimulus-induced inflammatory pain hypersensitivity. Proc Natl Acad Sci USA 1999, 96: 9385–9390. 10.1073/pnas.96.16.9385PubMedCentralPubMedCrossRef
37.
go back to reference Liu J, Valencia-Sanchez MA, Hannon GJ, Parker R: MicroRNA-dependent localization of targeted mRNAs to mammalian P-bodies. Nat Cell Biol 2005, 7: 719–723. 10.1038/ncb1274PubMedCentralPubMedCrossRef Liu J, Valencia-Sanchez MA, Hannon GJ, Parker R: MicroRNA-dependent localization of targeted mRNAs to mammalian P-bodies. Nat Cell Biol 2005, 7: 719–723. 10.1038/ncb1274PubMedCentralPubMedCrossRef
38.
go back to reference Fabian MR, Sonenberg N, Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem 2010, 79: 351–379. 10.1146/annurev-biochem-060308-103103PubMedCrossRef Fabian MR, Sonenberg N, Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem 2010, 79: 351–379. 10.1146/annurev-biochem-060308-103103PubMedCrossRef
39.
go back to reference Herranz H, Cohen SM: MicroRNAs and gene regulatory networks: managing the impact of noise in biological systems. Genes Dev 2010, 24: 1339–1344. 10.1101/gad.1937010PubMedCentralPubMedCrossRef Herranz H, Cohen SM: MicroRNAs and gene regulatory networks: managing the impact of noise in biological systems. Genes Dev 2010, 24: 1339–1344. 10.1101/gad.1937010PubMedCentralPubMedCrossRef
41.
go back to reference Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D: The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 1997, 389: 816–824. 10.1038/39807PubMedCrossRef Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D: The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 1997, 389: 816–824. 10.1038/39807PubMedCrossRef
42.
go back to reference Barth AL: Visualizing circuits and systems using transgenic reporters of neural activity. Curr Opin Neurobiol 2007, 17: 567–571. 10.1016/j.conb.2007.10.003PubMedCentralPubMedCrossRef Barth AL: Visualizing circuits and systems using transgenic reporters of neural activity. Curr Opin Neurobiol 2007, 17: 567–571. 10.1016/j.conb.2007.10.003PubMedCentralPubMedCrossRef
43.
go back to reference Kovacs KJ: Measurement of immediate-early gene activation-c-fos and beyond. J Neuroendocrinol 2008, 20: 665–672. 10.1111/j.1365-2826.2008.01734.xPubMedCrossRef Kovacs KJ: Measurement of immediate-early gene activation-c-fos and beyond. J Neuroendocrinol 2008, 20: 665–672. 10.1111/j.1365-2826.2008.01734.xPubMedCrossRef
44.
go back to reference Sng JC, Taniura H, Yoneda Y: A tale of early response genes. Biol Pharm Bull 2004, 27: 606–612. 10.1248/bpb.27.606PubMedCrossRef Sng JC, Taniura H, Yoneda Y: A tale of early response genes. Biol Pharm Bull 2004, 27: 606–612. 10.1248/bpb.27.606PubMedCrossRef
45.
go back to reference Coggeshall RE: Fos, nociception and the dorsal horn. Prog Neurobiol 2005, 77: 299–352.PubMed Coggeshall RE: Fos, nociception and the dorsal horn. Prog Neurobiol 2005, 77: 299–352.PubMed
46.
go back to reference Lima D, Almeida A: The medullary dorsal reticular nucleus as a pronociceptive centre of the pain control system. Prog Neurobiol 2002, 66: 81–108. 10.1016/S0301-0082(01)00025-9PubMedCrossRef Lima D, Almeida A: The medullary dorsal reticular nucleus as a pronociceptive centre of the pain control system. Prog Neurobiol 2002, 66: 81–108. 10.1016/S0301-0082(01)00025-9PubMedCrossRef
47.
go back to reference Zhao ZQ: Neural mechanism underlying acupuncture analgesia. Prog Neurobiol 2008, 85: 355–375. 10.1016/j.pneurobio.2008.05.004PubMedCrossRef Zhao ZQ: Neural mechanism underlying acupuncture analgesia. Prog Neurobiol 2008, 85: 355–375. 10.1016/j.pneurobio.2008.05.004PubMedCrossRef
48.
go back to reference Herdegen T, Zimmermann M: Immediate early genes (IEGs) encoding for inducible transcription factors (ITFs) and neuropeptides in the nervous system: functional network for long-term plasticity and pain. Prog Brain Res 1995, 104: 299–321. full_textPubMedCrossRef Herdegen T, Zimmermann M: Immediate early genes (IEGs) encoding for inducible transcription factors (ITFs) and neuropeptides in the nervous system: functional network for long-term plasticity and pain. Prog Brain Res 1995, 104: 299–321. full_textPubMedCrossRef
49.
50.
go back to reference Harris JA: Using c-fos as a neural marker of pain. Brain Res Bull 1998, 45: 1–8. 10.1016/S0361-9230(97)00277-3PubMedCrossRef Harris JA: Using c-fos as a neural marker of pain. Brain Res Bull 1998, 45: 1–8. 10.1016/S0361-9230(97)00277-3PubMedCrossRef
51.
go back to reference Zimmermann M: Immediate-early genes in the nervous system-are they involved in mechanisms of chronic pain? Patol Fiziol Eksp Ter 1992, 4: 47–51.PubMed Zimmermann M: Immediate-early genes in the nervous system-are they involved in mechanisms of chronic pain? Patol Fiziol Eksp Ter 1992, 4: 47–51.PubMed
52.
go back to reference Kerr BJ, Bradbury EJ, Bennett DL, Trivedi PM, Dassan P, French J, Shelton DB, McMahon SB, Thompson SW: Brain-derived neurotrophic factor modulates nociceptive sensory inputs and NMDA-evoked responses in the rat spinal cord. J Neurosci 1999, 19: 5138–5148.PubMed Kerr BJ, Bradbury EJ, Bennett DL, Trivedi PM, Dassan P, French J, Shelton DB, McMahon SB, Thompson SW: Brain-derived neurotrophic factor modulates nociceptive sensory inputs and NMDA-evoked responses in the rat spinal cord. J Neurosci 1999, 19: 5138–5148.PubMed
53.
go back to reference Merighi A, Salio C, Ghirri A, Lossi L, Ferrini F, Betelli C, Bardoni R: BDNF as a pain modulator. Prog Neurobiol 2008, 85: 297–317. 10.1016/j.pneurobio.2008.04.004PubMedCrossRef Merighi A, Salio C, Ghirri A, Lossi L, Ferrini F, Betelli C, Bardoni R: BDNF as a pain modulator. Prog Neurobiol 2008, 85: 297–317. 10.1016/j.pneurobio.2008.04.004PubMedCrossRef
54.
go back to reference Thompson SW, Bennett DL, Kerr BJ, Bradbury EJ, McMahon SB: Brain-derived neurotrophic factor is an endogenous modulator of nociceptive responses in the spinal cord. Proc Natl Acad Sci USA 1999, 96: 7714–7718. 10.1073/pnas.96.14.7714PubMedCentralPubMedCrossRef Thompson SW, Bennett DL, Kerr BJ, Bradbury EJ, McMahon SB: Brain-derived neurotrophic factor is an endogenous modulator of nociceptive responses in the spinal cord. Proc Natl Acad Sci USA 1999, 96: 7714–7718. 10.1073/pnas.96.14.7714PubMedCentralPubMedCrossRef
55.
go back to reference Galan A, Lopez-Garcia JA, Cervero F, Laird JM: Activation of spinal extracellular signaling-regulated kinase-1 and -2 by intraplantar carrageenan in rodents. Neurosci Lett 2002, 322: 37–40. 10.1016/S0304-3940(02)00078-2PubMedCrossRef Galan A, Lopez-Garcia JA, Cervero F, Laird JM: Activation of spinal extracellular signaling-regulated kinase-1 and -2 by intraplantar carrageenan in rodents. Neurosci Lett 2002, 322: 37–40. 10.1016/S0304-3940(02)00078-2PubMedCrossRef
56.
go back to reference Wei F, Qiu CS, Kim SJ, Muglia L, Maas JW, Pineda VV, Xu HM, Chen ZF, Storm DR, Muglia LJ, Zhuo M: Genetic elimination of behavioral sensitization in mice lacking calmodulin-stimulated adenylyl cyclases. Neuron 2002, 36: 713–726. 10.1016/S0896-6273(02)01019-XPubMedCrossRef Wei F, Qiu CS, Kim SJ, Muglia L, Maas JW, Pineda VV, Xu HM, Chen ZF, Storm DR, Muglia LJ, Zhuo M: Genetic elimination of behavioral sensitization in mice lacking calmodulin-stimulated adenylyl cyclases. Neuron 2002, 36: 713–726. 10.1016/S0896-6273(02)01019-XPubMedCrossRef
57.
go back to reference Zeitz KP, Giese KP, Silva AJ, Basbaum AI: The contribution of autophosphorylated alpha-calcium-calmodulin kinase II to injury-induced persistent pain. Neuroscience 2004, 128: 889–898. 10.1016/j.neuroscience.2004.07.029PubMedCrossRef Zeitz KP, Giese KP, Silva AJ, Basbaum AI: The contribution of autophosphorylated alpha-calcium-calmodulin kinase II to injury-induced persistent pain. Neuroscience 2004, 128: 889–898. 10.1016/j.neuroscience.2004.07.029PubMedCrossRef
58.
go back to reference Davar G, Shalish C, Blumenfeld A, Breakfield XO: Exclusion of p75NGFR and other candidate genes in a family with hereditary sensory neuropathy type II. Pain 1996, 67: 135–139. 10.1016/0304-3959(96)03113-2PubMedCrossRef Davar G, Shalish C, Blumenfeld A, Breakfield XO: Exclusion of p75NGFR and other candidate genes in a family with hereditary sensory neuropathy type II. Pain 1996, 67: 135–139. 10.1016/0304-3959(96)03113-2PubMedCrossRef
59.
go back to reference Fukui Y, Ohtori S, Yamashita M, Yamauchi K, Inoue G, Suzuki M, Orita S, Eguchi Y, Ochiai N, Kishida S, Takaso M, Wakai K, Hayashi Y, Aoki Y, Takahashi K: Low affinity NGF receptor (p75 neurotrophin receptor) inhibitory antibody reduces pain behavior and CGRP expression in DRG in the mouse sciatic nerve crush model. J Orthop Res 2009, 28: 279–283. Fukui Y, Ohtori S, Yamashita M, Yamauchi K, Inoue G, Suzuki M, Orita S, Eguchi Y, Ochiai N, Kishida S, Takaso M, Wakai K, Hayashi Y, Aoki Y, Takahashi K: Low affinity NGF receptor (p75 neurotrophin receptor) inhibitory antibody reduces pain behavior and CGRP expression in DRG in the mouse sciatic nerve crush model. J Orthop Res 2009, 28: 279–283.
60.
go back to reference Watanabe T, Ito T, Inoue G, Ohtori S, Kitajo K, Doya H, Takahashi K, Yamashita T: The p75 receptor is associated with inflammatory thermal hypersensitivity. J Neurosci Res 2008, 86: 3566–3574. 10.1002/jnr.21808PubMedCrossRef Watanabe T, Ito T, Inoue G, Ohtori S, Kitajo K, Doya H, Takahashi K, Yamashita T: The p75 receptor is associated with inflammatory thermal hypersensitivity. J Neurosci Res 2008, 86: 3566–3574. 10.1002/jnr.21808PubMedCrossRef
61.
go back to reference Hasegawa S, Kohro Y, Shiratori M, Ishii S, Shimizu T, Tsuda M, Inoue K: Role of PAF receptor in proinflammatory cytokine expression in the dorsal root ganglion and tactile allodynia in a rodent model of neuropathic pain. PLoS One 2010, 5: e10467. 10.1371/journal.pone.0010467PubMedCentralPubMedCrossRef Hasegawa S, Kohro Y, Shiratori M, Ishii S, Shimizu T, Tsuda M, Inoue K: Role of PAF receptor in proinflammatory cytokine expression in the dorsal root ganglion and tactile allodynia in a rodent model of neuropathic pain. PLoS One 2010, 5: e10467. 10.1371/journal.pone.0010467PubMedCentralPubMedCrossRef
62.
go back to reference Inoue M, Ma L, Aoki J, Ueda H: Simultaneous stimulation of spinal NK1 and NMDA receptors produces LPC which undergoes ATX-mediated conversion to LPA, an initiator of neuropathic pain. J Neurochem 2008, 107: 1556–1565. 10.1111/j.1471-4159.2008.05725.xPubMedCrossRef Inoue M, Ma L, Aoki J, Ueda H: Simultaneous stimulation of spinal NK1 and NMDA receptors produces LPC which undergoes ATX-mediated conversion to LPA, an initiator of neuropathic pain. J Neurochem 2008, 107: 1556–1565. 10.1111/j.1471-4159.2008.05725.xPubMedCrossRef
63.
go back to reference Yeo JF, Ong WY, Ling SF, Farooqui AA: Intracerebroventricular injection of phospholipases A2 inhibitors modulates allodynia after facial carrageenan injection in mice. Pain 2004, 112: 148–155. 10.1016/j.pain.2004.08.009PubMedCrossRef Yeo JF, Ong WY, Ling SF, Farooqui AA: Intracerebroventricular injection of phospholipases A2 inhibitors modulates allodynia after facial carrageenan injection in mice. Pain 2004, 112: 148–155. 10.1016/j.pain.2004.08.009PubMedCrossRef
64.
go back to reference Contreras PC, Vaught JL, Gruner JA, Brosnan C, Steffler C, Arezzo JC, Lewis ME, Kessler JA, Apfel SC: Insulin-like growth factor-I prevents development of a vincristine neuropathy in mice. Brain Res 1997, 774: 20–26. 10.1016/S0006-8993(97)81682-4PubMedCrossRef Contreras PC, Vaught JL, Gruner JA, Brosnan C, Steffler C, Arezzo JC, Lewis ME, Kessler JA, Apfel SC: Insulin-like growth factor-I prevents development of a vincristine neuropathy in mice. Brain Res 1997, 774: 20–26. 10.1016/S0006-8993(97)81682-4PubMedCrossRef
65.
go back to reference de Pablo F, Banner LR, Patterson PH: IGF-I expression is decreased in LIF-deficient mice after peripheral nerve injury. Neuroreport 2000, 11: 1365–1368. 10.1097/00001756-200004270-00043PubMedCrossRef de Pablo F, Banner LR, Patterson PH: IGF-I expression is decreased in LIF-deficient mice after peripheral nerve injury. Neuroreport 2000, 11: 1365–1368. 10.1097/00001756-200004270-00043PubMedCrossRef
66.
go back to reference Staaf S, Oerther S, Lucas G, Mattsson JP, Ernfors P: Differential regulation of TRP channels in a rat model of neuropathic pain. Pain 2009, 144: 187–199. 10.1016/j.pain.2009.04.013PubMedCrossRef Staaf S, Oerther S, Lucas G, Mattsson JP, Ernfors P: Differential regulation of TRP channels in a rat model of neuropathic pain. Pain 2009, 144: 187–199. 10.1016/j.pain.2009.04.013PubMedCrossRef
67.
go back to reference Gaveriaux-Ruff C, Karchewski LA, Hever X, Matifas A, Kieffer BL: Inflammatory pain is enhanced in delta opioid receptor-knockout mice. Eur J Neurosci 2008, 27: 2558–2567. 10.1111/j.1460-9568.2008.06223.xPubMedCentralPubMedCrossRef Gaveriaux-Ruff C, Karchewski LA, Hever X, Matifas A, Kieffer BL: Inflammatory pain is enhanced in delta opioid receptor-knockout mice. Eur J Neurosci 2008, 27: 2558–2567. 10.1111/j.1460-9568.2008.06223.xPubMedCentralPubMedCrossRef
68.
go back to reference Nadal X, Banos JE, Kieffer BL, Maldonado R: Neuropathic pain is enhanced in delta-opioid receptor knockout mice. Eur J Neurosci 2006, 23: 830–834. 10.1111/j.1460-9568.2006.04569.xPubMedCrossRef Nadal X, Banos JE, Kieffer BL, Maldonado R: Neuropathic pain is enhanced in delta-opioid receptor knockout mice. Eur J Neurosci 2006, 23: 830–834. 10.1111/j.1460-9568.2006.04569.xPubMedCrossRef
Metadata
Title
Differential expression of microRNAs in mouse pain models
Authors
Ricardo Kusuda
Flaviane Cadetti
Maria I Ravanelli
Thais A Sousa
Sônia Zanon
Fernando L De Lucca
Guilherme Lucas
Publication date
01-12-2011
Publisher
BioMed Central
Published in
Molecular Pain / Issue 1/2011
Electronic ISSN: 1744-8069
DOI
https://doi.org/10.1186/1744-8069-7-17

Other articles of this Issue 1/2011

Molecular Pain 1/2011 Go to the issue