Skip to main content
Top
Published in: Molecular Pain 1/2005

Open Access 01-12-2005 | Research

Controlling neuropathic pain by adeno-associated virus driven production of the anti-inflammatory cytokine, interleukin-10

Authors: Erin D Milligan, Evan M Sloane, Stephen J Langer, Pedro E Cruz, Marucia Chacur, Leah Spataro, Julie Wieseler-Frank, Sayamwong E Hammack, Steven F Maier, Terence R Flotte, John R Forsayeth, Leslie A Leinwand, Raymond Chavez, Linda R Watkins

Published in: Molecular Pain | Issue 1/2005

Login to get access

Abstract

Despite many decades of drug development, effective therapies for neuropathic pain remain elusive. The recent recognition of spinal cord glia and glial pro-inflammatory cytokines as important contributors to neuropathic pain suggests an alternative therapeutic strategy; that is, targeting glial activation or its downstream consequences. While several glial-selective drugs have been successful in controlling neuropathic pain in animal models, none are optimal for human use. Thus the aim of the present studies was to explore a novel approach for controlling neuropathic pain. Here, an adeno-associated viral (serotype II; AAV2) vector was created that encodes the anti-inflammatory cytokine, interleukin-10 (IL-10). This anti-inflammatory cytokine is known to suppress the production of pro-inflammatory cytokines. Upon intrathecal administration, this novel AAV2-IL-10 vector was successful in transiently preventing and reversing neuropathic pain. Intrathecal administration of an AAV2 vector encoding beta-galactosidase revealed that AAV2 preferentially infects meningeal cells surrounding the CSF space. Taken together, these data provide initial support that intrathecal gene therapy to drive the production of IL-10 may prove to be an efficacious treatment for neuropathic pain.
Appendix
Available only for authorised users
Literature
1.
go back to reference Collins SL, Moore A, McQuay HJ, Wiffen P: Antidepressants and anticonvulsants for diabetic neuropathy and postherpetic neuralgia: a quantitative systematic review. J Pain Symptom Manage 2000, 20: 339–457. 10.1016/S0885-3924(00)00218-9CrossRef Collins SL, Moore A, McQuay HJ, Wiffen P: Antidepressants and anticonvulsants for diabetic neuropathy and postherpetic neuralgia: a quantitative systematic review. J Pain Symptom Manage 2000, 20: 339–457. 10.1016/S0885-3924(00)00218-9CrossRef
2.
go back to reference McQuay H, Tramer M, Nye BA, Carroll D, Wiffen P, Moore RA: A systematic review of antidepressants in neuropathic pain. Pain 1996, 68: 217–227. 10.1016/S0304-3959(96)03140-5PubMedCrossRef McQuay H, Tramer M, Nye BA, Carroll D, Wiffen P, Moore RA: A systematic review of antidepressants in neuropathic pain. Pain 1996, 68: 217–227. 10.1016/S0304-3959(96)03140-5PubMedCrossRef
3.
4.
go back to reference DeLeo JA, Tanga FY, Tawfik V: Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. The Neuroscientist 2004, 10: 40–52. 10.1177/1073858403259950PubMedCrossRef DeLeo JA, Tanga FY, Tawfik V: Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. The Neuroscientist 2004, 10: 40–52. 10.1177/1073858403259950PubMedCrossRef
5.
go back to reference Watkins LR, Maier SF: Glia: a novel drug discovery target for clinical pain. Nature Reviews Drug Discovery 2003, 2: 973–985. 10.1038/nrd1251PubMedCrossRef Watkins LR, Maier SF: Glia: a novel drug discovery target for clinical pain. Nature Reviews Drug Discovery 2003, 2: 973–985. 10.1038/nrd1251PubMedCrossRef
6.
go back to reference Watkins LR, Milligan ED, Maier SF: Glial activation: a driving force for pathological pain. Trends Neurosci 2001, 24: 450–455. 10.1016/S0166-2236(00)01854-3PubMedCrossRef Watkins LR, Milligan ED, Maier SF: Glial activation: a driving force for pathological pain. Trends Neurosci 2001, 24: 450–455. 10.1016/S0166-2236(00)01854-3PubMedCrossRef
7.
go back to reference Watkins LR, Maier SF: Targeting glia to control clinical pain: An idea whose time has come. Drug Discovery Today: Therapeutic Strategies 2004. Watkins LR, Maier SF: Targeting glia to control clinical pain: An idea whose time has come. Drug Discovery Today: Therapeutic Strategies 2004.
8.
go back to reference Berg-Johnsen J, Paulsen RE, Fonnum F, Langmoen IA: Changes in evoked potentials and amino acid content during fluorocitrate action studied in rat hippocampal cortex. Exp Brain Res 1993, 96: 241–246. 10.1007/BF00227104PubMedCrossRef Berg-Johnsen J, Paulsen RE, Fonnum F, Langmoen IA: Changes in evoked potentials and amino acid content during fluorocitrate action studied in rat hippocampal cortex. Exp Brain Res 1993, 96: 241–246. 10.1007/BF00227104PubMedCrossRef
9.
go back to reference Hassel B, Paulsen RE, Johnson A, Fonnum F: Selective inhibition of glial cell metabolism by fluorocitrate. Brain Res 1992, 249: 120–124. 10.1016/0006-8993(92)90616-HCrossRef Hassel B, Paulsen RE, Johnson A, Fonnum F: Selective inhibition of glial cell metabolism by fluorocitrate. Brain Res 1992, 249: 120–124. 10.1016/0006-8993(92)90616-HCrossRef
10.
go back to reference Milligan ED, Twining C, Chacur M, Biedenkapp J, O'Connor KA, Poole S, Tracey KJ, Martin D, Maier SF, Watkins LR: Spinal glia and proinflammatory cytokines mediate mirror-image neuropathic pain. J Neurosci 2003, 23: 1026–1040.PubMed Milligan ED, Twining C, Chacur M, Biedenkapp J, O'Connor KA, Poole S, Tracey KJ, Martin D, Maier SF, Watkins LR: Spinal glia and proinflammatory cytokines mediate mirror-image neuropathic pain. J Neurosci 2003, 23: 1026–1040.PubMed
11.
go back to reference Willoughby JO, Mackenzie L, Broberg M, Thoren AE, Medvedev A, Sims NR, Nilsson M: Fluorocitrate-mediated astroglial dysfunction causes seizures. J Neurosci Res 2003, 74: 160–166. 10.1002/jnr.10743PubMedCrossRef Willoughby JO, Mackenzie L, Broberg M, Thoren AE, Medvedev A, Sims NR, Nilsson M: Fluorocitrate-mediated astroglial dysfunction causes seizures. J Neurosci Res 2003, 74: 160–166. 10.1002/jnr.10743PubMedCrossRef
12.
go back to reference Tikka T, Fiebich BL, Godsteins G, Keinanen R, Koistinaho J: Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci 2001, 21: 2580–2588.PubMed Tikka T, Fiebich BL, Godsteins G, Keinanen R, Koistinaho J: Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci 2001, 21: 2580–2588.PubMed
13.
go back to reference Raghavendra V, Tanga F, DeLeo JA: Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J Pharmacol Exp Ther 2003, 306: 624–630. 10.1124/jpet.103.052407PubMedCrossRef Raghavendra V, Tanga F, DeLeo JA: Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J Pharmacol Exp Ther 2003, 306: 624–630. 10.1124/jpet.103.052407PubMedCrossRef
14.
go back to reference Ledeboer A, Sloane E, Chacur M, Milligan ED, Maier SF, Watkins LR: Selective inhibition of spinal cord microglial activation attenuates mechanical allodynia in rat models of pathological pain. Proc Soc Neurosci 2003., 29: Ledeboer A, Sloane E, Chacur M, Milligan ED, Maier SF, Watkins LR: Selective inhibition of spinal cord microglial activation attenuates mechanical allodynia in rat models of pathological pain. Proc Soc Neurosci 2003., 29:
15.
go back to reference Sweitzer SM, Martin D, DeLeo JA: Intrathecal interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor exhibits an anti-allodynic action in a rat model of neuropathic pain. Neurosci 2001, 103: 529–539. 10.1016/S0306-4522(00)00574-1CrossRef Sweitzer SM, Martin D, DeLeo JA: Intrathecal interleukin-1 receptor antagonist in combination with soluble tumor necrosis factor receptor exhibits an anti-allodynic action in a rat model of neuropathic pain. Neurosci 2001, 103: 529–539. 10.1016/S0306-4522(00)00574-1CrossRef
16.
go back to reference DeLeo JA, Colburn RW, Nichols M, Malhotra A: Interleukin (IL)-6 mediated hyperalgesia/allodynia and increased spinal IL-6 in a rat mononeuropathy model. J Interferon Cytokine Res 1996, 16: 695–700.PubMedCrossRef DeLeo JA, Colburn RW, Nichols M, Malhotra A: Interleukin (IL)-6 mediated hyperalgesia/allodynia and increased spinal IL-6 in a rat mononeuropathy model. J Interferon Cytokine Res 1996, 16: 695–700.PubMedCrossRef
17.
go back to reference Sweitzer SM, Schubert P, DeLeo JA: Propentofylline, a glial modulating agent, exhibits anti-allodynic properties in a rat model of neuropathic pain. J Pharmacol Exp Ther 2001, 297: 1210–1217.PubMed Sweitzer SM, Schubert P, DeLeo JA: Propentofylline, a glial modulating agent, exhibits anti-allodynic properties in a rat model of neuropathic pain. J Pharmacol Exp Ther 2001, 297: 1210–1217.PubMed
18.
go back to reference Hashizume H, Rutkowski MD, Weinstein JN, DeLeo JA: Central administration of methotrexate reduces mechanical allodynia in an animal model of radiculopathy/sciatica. Pain 2000, 87: 159–169. 10.1016/S0304-3959(00)00281-5PubMedCrossRef Hashizume H, Rutkowski MD, Weinstein JN, DeLeo JA: Central administration of methotrexate reduces mechanical allodynia in an animal model of radiculopathy/sciatica. Pain 2000, 87: 159–169. 10.1016/S0304-3959(00)00281-5PubMedCrossRef
19.
go back to reference Moore KW, Ho ASY, Xu-Amano J: Molecular biology of interleukin-10 and its receptor. In Interleukin-10. Edited by: DeVries JE and de Waal Malefyt R. , R.G. Landes Company; 1995:1–9.CrossRef Moore KW, Ho ASY, Xu-Amano J: Molecular biology of interleukin-10 and its receptor. In Interleukin-10. Edited by: DeVries JE and de Waal Malefyt R. , R.G. Landes Company; 1995:1–9.CrossRef
20.
go back to reference Chacur M, Milligan ED, Gazda LS, Armstrong C, Wang H, Tracey KJ, Maier SF, Watkins LR: A new model of sciatic inflammatory neuritis (SIN): induction of unilateral and bilateral mechanical allodynia following acute unilateral peri-sciatic immune activation in rats. Pain 2001, 94: 231–244. 10.1016/S0304-3959(01)00354-2PubMedCrossRef Chacur M, Milligan ED, Gazda LS, Armstrong C, Wang H, Tracey KJ, Maier SF, Watkins LR: A new model of sciatic inflammatory neuritis (SIN): induction of unilateral and bilateral mechanical allodynia following acute unilateral peri-sciatic immune activation in rats. Pain 2001, 94: 231–244. 10.1016/S0304-3959(01)00354-2PubMedCrossRef
21.
go back to reference Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988, 33: 87–107. 10.1016/0304-3959(88)90209-6PubMedCrossRef Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988, 33: 87–107. 10.1016/0304-3959(88)90209-6PubMedCrossRef
22.
go back to reference Spataro LE, Sloane EM, Milligan ED, Wieseler-Frank J, Schoeniger D, Jekich BM, Barrientos RM, Maier SF, Watkins LR: Spinal gap junctions: Potential involvement in pain facilitation. J Pain 2004, 5: 392–405. 10.1016/j.jpain.2004.06.006PubMedCrossRef Spataro LE, Sloane EM, Milligan ED, Wieseler-Frank J, Schoeniger D, Jekich BM, Barrientos RM, Maier SF, Watkins LR: Spinal gap junctions: Potential involvement in pain facilitation. J Pain 2004, 5: 392–405. 10.1016/j.jpain.2004.06.006PubMedCrossRef
23.
go back to reference Twining CM, Sloane EM, Schoeniger DK, Milligan ED, Martin D, Marsh H, Maier SF, Watkins LR: Activation of the spinal cord complement cascade may contribute to mechanical allodynia induced by three animal models. Journal of Pain 2004, in press. Twining CM, Sloane EM, Schoeniger DK, Milligan ED, Martin D, Marsh H, Maier SF, Watkins LR: Activation of the spinal cord complement cascade may contribute to mechanical allodynia induced by three animal models. Journal of Pain 2004, in press.
24.
go back to reference Milligan ED, Zapata V, Chacur M, Schoeniger D, Biedenkapp J, O’Connor K, Verge GM, Chapman G, Green P, Foster AC, Naeve GS, Maier SF, Watkins LR: Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation. Eur J Neurosci 2004, 20: 2294–2302. 10.1111/j.1460-9568.2004.03709.xPubMedCrossRef Milligan ED, Zapata V, Chacur M, Schoeniger D, Biedenkapp J, O’Connor K, Verge GM, Chapman G, Green P, Foster AC, Naeve GS, Maier SF, Watkins LR: Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation. Eur J Neurosci 2004, 20: 2294–2302. 10.1111/j.1460-9568.2004.03709.xPubMedCrossRef
25.
go back to reference Mannes AJ, Caudle RM, O'Connell BC, Iadarola MJ: Adenoviral gene transfer to spinal cord neurons: intrathecal vs. intraparenchymal administration. Brain Res 1998, 793: 1–6. 10.1016/S0006-8993(97)01422-4PubMedCrossRef Mannes AJ, Caudle RM, O'Connell BC, Iadarola MJ: Adenoviral gene transfer to spinal cord neurons: intrathecal vs. intraparenchymal administration. Brain Res 1998, 793: 1–6. 10.1016/S0006-8993(97)01422-4PubMedCrossRef
26.
go back to reference Milligan ED, Maier SF, Watkins LR: Sciatic Inflammatory Neuropathy (SIN) in the rat: Surgical procedures, induction of inflammation and behavioral testing. In Pain Research: Methods and Protocols. Volume 99. Edited by: Luo ZD. Totowa, Humana Press; 2004:67–89.CrossRef Milligan ED, Maier SF, Watkins LR: Sciatic Inflammatory Neuropathy (SIN) in the rat: Surgical procedures, induction of inflammation and behavioral testing. In Pain Research: Methods and Protocols. Volume 99. Edited by: Luo ZD. Totowa, Humana Press; 2004:67–89.CrossRef
27.
go back to reference Gazda LS, Milligan ED, Hansen MK, Twining CM, Paulos N, Chacur M, O’Connor KA, Armstrong C, Maier SF: Sciatic inflammatory neuritis (SIN): behavioral allodynia is paralleled by peri-sciatic proinflammatory cytokine and superoxide production. J Peripheral Nerv Sys 2001, 6: 111–129. 10.1046/j.1529-8027.2001.006001111.xCrossRef Gazda LS, Milligan ED, Hansen MK, Twining CM, Paulos N, Chacur M, O’Connor KA, Armstrong C, Maier SF: Sciatic inflammatory neuritis (SIN): behavioral allodynia is paralleled by peri-sciatic proinflammatory cytokine and superoxide production. J Peripheral Nerv Sys 2001, 6: 111–129. 10.1046/j.1529-8027.2001.006001111.xCrossRef
28.
go back to reference Yoshimura A, Mori H, Ohishi M, Aki D, Hanada T: Negative regulation of cytokine signaling influences inflammation. Curr Opion Immunol 2003, 15: 704–708. 10.1016/j.coi.2003.09.004CrossRef Yoshimura A, Mori H, Ohishi M, Aki D, Hanada T: Negative regulation of cytokine signaling influences inflammation. Curr Opion Immunol 2003, 15: 704–708. 10.1016/j.coi.2003.09.004CrossRef
29.
go back to reference Opal SM, DePalo VA: Anti-inflammatory cytokines. Chest 2000, 117: 1162–1172. 10.1378/chest.117.4.1162PubMedCrossRef Opal SM, DePalo VA: Anti-inflammatory cytokines. Chest 2000, 117: 1162–1172. 10.1378/chest.117.4.1162PubMedCrossRef
30.
go back to reference Knight D: Leukaemia inhibitory factor (LIF): a cytokine of emerging importance in chronic airway inflammation. Pulm Pharmacol Ther 2001, 14: 169–176. 10.1006/pupt.2001.0282PubMedCrossRef Knight D: Leukaemia inhibitory factor (LIF): a cytokine of emerging importance in chronic airway inflammation. Pulm Pharmacol Ther 2001, 14: 169–176. 10.1006/pupt.2001.0282PubMedCrossRef
31.
go back to reference Jones SA, Horiuchi S, Topley N, Yamamoto N, Fuller GM: The soluble interleukin 6 receptor: mechanisms of production and implications in disease. FASEB J 2001, 15: 43–58. 10.1096/fj.99-1003revPubMedCrossRef Jones SA, Horiuchi S, Topley N, Yamamoto N, Fuller GM: The soluble interleukin 6 receptor: mechanisms of production and implications in disease. FASEB J 2001, 15: 43–58. 10.1096/fj.99-1003revPubMedCrossRef
32.
go back to reference Tilg H, Peschel C: Interferon-alpha and its effects on the cytokine cascade: a pro- and anti-inflammatory cytokine. Leuk Lymphoma 1996, 23: 55–60.PubMedCrossRef Tilg H, Peschel C: Interferon-alpha and its effects on the cytokine cascade: a pro- and anti-inflammatory cytokine. Leuk Lymphoma 1996, 23: 55–60.PubMedCrossRef
33.
go back to reference Strle K, Zhou JH, Broussard SR, Johnson RW, Freund GG, Dantzer R, Kelley KW: Interleukin-10 in the brain. Crit Rev Immunology 2001, 21: 427–449.CrossRef Strle K, Zhou JH, Broussard SR, Johnson RW, Freund GG, Dantzer R, Kelley KW: Interleukin-10 in the brain. Crit Rev Immunology 2001, 21: 427–449.CrossRef
34.
go back to reference Bluthe RM, Laye S, Michaud B, Combe C, Dantzer R, Parnet P: Role of interleukin-1beta and tumour necrosis factor-alpha in lipolysaccharide-induced sickness behaviour: a study with interleukin-1 type I receptor-deficient mice. Eur J Neurosci 2000, 12: 4447–4456. 10.1046/j.1460-9568.2000.01348.xPubMed Bluthe RM, Laye S, Michaud B, Combe C, Dantzer R, Parnet P: Role of interleukin-1beta and tumour necrosis factor-alpha in lipolysaccharide-induced sickness behaviour: a study with interleukin-1 type I receptor-deficient mice. Eur J Neurosci 2000, 12: 4447–4456. 10.1046/j.1460-9568.2000.01348.xPubMed
35.
go back to reference Raghavendra V, Tanga FY, DeLeo JA: Complete Freunds adjuvant-induced peripheral inflammation evokes glial activation and proinflammatory cytokine expression in the CNS. Eur J Neurosci 2004, 20: 467–473. 10.1111/j.1460-9568.2004.03514.xPubMedCrossRef Raghavendra V, Tanga FY, DeLeo JA: Complete Freunds adjuvant-induced peripheral inflammation evokes glial activation and proinflammatory cytokine expression in the CNS. Eur J Neurosci 2004, 20: 467–473. 10.1111/j.1460-9568.2004.03514.xPubMedCrossRef
36.
go back to reference Laughlin TM, Bethea JR, Yezierski RP, Wilcox GL: Cytokine involvement in dynorphin-induced allodynia. Pain 2000, 84: 159–167. 10.1016/S0304-3959(99)00195-5PubMedCrossRef Laughlin TM, Bethea JR, Yezierski RP, Wilcox GL: Cytokine involvement in dynorphin-induced allodynia. Pain 2000, 84: 159–167. 10.1016/S0304-3959(99)00195-5PubMedCrossRef
37.
go back to reference Chacur M, Milligan ED, Sloane EM, Wieseler-Frank J, Barrientos RM, Martin D, Poole S, Lomonte B, Gutierrez JM, Maier SF, Cury Y, Watkins LR: Snake venom phospholipase A2s (Asp49 and Lys49) induce mechanical allodynia upon peri-sciatic administration: involvement of spinal cord glia, proinflammatory cytokines and nitric oxide. Pain 2004, 108: 180–191. 10.1016/j.pain.2003.12.023PubMedCrossRef Chacur M, Milligan ED, Sloane EM, Wieseler-Frank J, Barrientos RM, Martin D, Poole S, Lomonte B, Gutierrez JM, Maier SF, Cury Y, Watkins LR: Snake venom phospholipase A2s (Asp49 and Lys49) induce mechanical allodynia upon peri-sciatic administration: involvement of spinal cord glia, proinflammatory cytokines and nitric oxide. Pain 2004, 108: 180–191. 10.1016/j.pain.2003.12.023PubMedCrossRef
38.
go back to reference Plunkett JA, Yu CG, Easton J, Bethea JR, Yezierski RP: Effects of interleukin-10 (IL-10) on pain behavior and gene expression following excitotoxic spinal cord injury in the rat. Exper Neurol 2001, 168: 144–154. 10.1006/exnr.2000.7604CrossRef Plunkett JA, Yu CG, Easton J, Bethea JR, Yezierski RP: Effects of interleukin-10 (IL-10) on pain behavior and gene expression following excitotoxic spinal cord injury in the rat. Exper Neurol 2001, 168: 144–154. 10.1006/exnr.2000.7604CrossRef
39.
go back to reference Yu CG, Fairbanks CA, Wilcox GL, Yezierski RP: Effects of agmatine, interleukin-10 and cyclosporin on spontaneous pain behavior following excitotoxic spinal cord injury in rats. J Pain 2003, 4: 129–140. 10.1054/jpai.2003.11PubMedCrossRef Yu CG, Fairbanks CA, Wilcox GL, Yezierski RP: Effects of agmatine, interleukin-10 and cyclosporin on spontaneous pain behavior following excitotoxic spinal cord injury in rats. J Pain 2003, 4: 129–140. 10.1054/jpai.2003.11PubMedCrossRef
40.
go back to reference Abraham KE, McMillen D, Brewer KL: The effects of endogenous interleukin-10 on gray matter damage and pain behaviors following excitotoxic spinal cord injury in the mouse. Neurosci 2004, 124: 945–922. 10.1016/j.neuroscience.2004.01.004CrossRef Abraham KE, McMillen D, Brewer KL: The effects of endogenous interleukin-10 on gray matter damage and pain behaviors following excitotoxic spinal cord injury in the mouse. Neurosci 2004, 124: 945–922. 10.1016/j.neuroscience.2004.01.004CrossRef
41.
go back to reference Ledeboer A, Wierinckx A, Bol JGJM, Floris S, Renardel de Lavaletter C, DeVries HE, van den Berg T, Dijkstra CD, Tilders FJH, Van Dam AM: Regional and temporal expression patterns of interleukin-10, interleukin-10 receptor and adhesion molecules in the rat spinal cord during chronic relapsing EAE. J Neuroimmunol 2003, 136: 94–103. 10.1016/S0165-5728(03)00031-6PubMedCrossRef Ledeboer A, Wierinckx A, Bol JGJM, Floris S, Renardel de Lavaletter C, DeVries HE, van den Berg T, Dijkstra CD, Tilders FJH, Van Dam AM: Regional and temporal expression patterns of interleukin-10, interleukin-10 receptor and adhesion molecules in the rat spinal cord during chronic relapsing EAE. J Neuroimmunol 2003, 136: 94–103. 10.1016/S0165-5728(03)00031-6PubMedCrossRef
42.
go back to reference Koeberle PD, Gauldie J, Ball AK: Effects of adenoviral-mediated gene transfer of interleukin-10, interleukin-4, and transforming growth factor-beta on the survival of axotomized retinal ganglion cells. Neurosci 2004, 125: 903–920. 10.1016/S0306-4522(03)00398-1CrossRef Koeberle PD, Gauldie J, Ball AK: Effects of adenoviral-mediated gene transfer of interleukin-10, interleukin-4, and transforming growth factor-beta on the survival of axotomized retinal ganglion cells. Neurosci 2004, 125: 903–920. 10.1016/S0306-4522(03)00398-1CrossRef
43.
go back to reference Lynch AM, Walsh C, Delaney A, Nolan Y, Campbell VA, Lynch MA: Lipopolysaccharide-induced increase in signalling in hippocampus is abrogated by IL-10 -- a role for IL-1 beta? J Neurosci 2004, 88: 635–646. Lynch AM, Walsh C, Delaney A, Nolan Y, Campbell VA, Lynch MA: Lipopolysaccharide-induced increase in signalling in hippocampus is abrogated by IL-10 -- a role for IL-1 beta? J Neurosci 2004, 88: 635–646.
44.
go back to reference Grilli M, Barbieri I, Basudev H, Brusa R, Casati C, Lozza G, Ongini E: Interleukin-10 modulates neuronal threshold of vulnerability to ischaemic damage. Eur J Neurosci 2000, 12: 2265–2272. 10.1046/j.1460-9568.2000.00090.xPubMedCrossRef Grilli M, Barbieri I, Basudev H, Brusa R, Casati C, Lozza G, Ongini E: Interleukin-10 modulates neuronal threshold of vulnerability to ischaemic damage. Eur J Neurosci 2000, 12: 2265–2272. 10.1046/j.1460-9568.2000.00090.xPubMedCrossRef
45.
go back to reference Brewer KL, Bethea JR, Yezierski RP: Neuroprotective effects of interleukin-10 folowing spinal cord injury. Exp Neurol 1999, 159: 484–493. 10.1006/exnr.1999.7173PubMedCrossRef Brewer KL, Bethea JR, Yezierski RP: Neuroprotective effects of interleukin-10 folowing spinal cord injury. Exp Neurol 1999, 159: 484–493. 10.1006/exnr.1999.7173PubMedCrossRef
46.
go back to reference Buchschacher GLJ, Wong-Staal F: Development of lentiviral vectors for gene therapy for human diseases. Blood 2000, 95: 2499–2504.PubMed Buchschacher GLJ, Wong-Staal F: Development of lentiviral vectors for gene therapy for human diseases. Blood 2000, 95: 2499–2504.PubMed
47.
go back to reference Gudmundsson G, Bosch A, Davidson BL, Hunninghake GW: Interleukin-10 modulates the severity of hypersensitivity pneumonitis. Amer J Resp Cell & Molec Biol 1998, 19: 812–818.CrossRef Gudmundsson G, Bosch A, Davidson BL, Hunninghake GW: Interleukin-10 modulates the severity of hypersensitivity pneumonitis. Amer J Resp Cell & Molec Biol 1998, 19: 812–818.CrossRef
48.
go back to reference Daly TM: Overview of adeno-associated viral vectors. Methods Mol Biol 2004, 246: 157–165.PubMed Daly TM: Overview of adeno-associated viral vectors. Methods Mol Biol 2004, 246: 157–165.PubMed
49.
go back to reference Blits B, Carlstedt TP, Ruitenberg MJ, DeWinter F, Hermens WT, Dijkhuizen PA, Claasens JW, Eggers R, Van der Sluis R, Tenenbaum L, Boer GJ, Verhaagen J: Rescue and sprouting of motoneurons following ventral root avulsion and reimplantation combined with intraspinal adeno-associated viral vector-mediated expression of glial cell line-derived neurotrophic factor or brain-derived neurotrophic factor. Exp Neurol 2004, 189: 303–316. 10.1016/j.expneurol.2004.05.014PubMedCrossRef Blits B, Carlstedt TP, Ruitenberg MJ, DeWinter F, Hermens WT, Dijkhuizen PA, Claasens JW, Eggers R, Van der Sluis R, Tenenbaum L, Boer GJ, Verhaagen J: Rescue and sprouting of motoneurons following ventral root avulsion and reimplantation combined with intraspinal adeno-associated viral vector-mediated expression of glial cell line-derived neurotrophic factor or brain-derived neurotrophic factor. Exp Neurol 2004, 189: 303–316. 10.1016/j.expneurol.2004.05.014PubMedCrossRef
50.
go back to reference Xu Y, Gu Y, Wu PC, Li GW, Huang LYM: Efficiencies of transgene expression in nociceptive neurons through different routes of delivery of adeno-associated viral vectors. Human Gene Ther 2003, 14: 897–906. 10.1089/104303403765701187CrossRef Xu Y, Gu Y, Wu PC, Li GW, Huang LYM: Efficiencies of transgene expression in nociceptive neurons through different routes of delivery of adeno-associated viral vectors. Human Gene Ther 2003, 14: 897–906. 10.1089/104303403765701187CrossRef
51.
go back to reference Bartlett JS, Samulski RJ, McCown TJ: Selective and rapid uptake of adeno-associated virus type 2 in brain. Hum Gene Ther 1998, 9: 1181–1186.PubMedCrossRef Bartlett JS, Samulski RJ, McCown TJ: Selective and rapid uptake of adeno-associated virus type 2 in brain. Hum Gene Ther 1998, 9: 1181–1186.PubMedCrossRef
52.
go back to reference Kugler S, Lingor P, Scholl U, Zolotukhin S, Bahr M: Differential transgene expression in brain cells in vivo and in vitro from AAV-2 vectors with small transcriptional control units. Virology 2003, 311: 89–95. 10.1016/S0042-6822(03)00162-4PubMedCrossRef Kugler S, Lingor P, Scholl U, Zolotukhin S, Bahr M: Differential transgene expression in brain cells in vivo and in vitro from AAV-2 vectors with small transcriptional control units. Virology 2003, 311: 89–95. 10.1016/S0042-6822(03)00162-4PubMedCrossRef
53.
go back to reference Burger C, Gorbatyuk OS, Velardo MJ, Peden CS, Williams P, Zolotukhin S, Reier PJ, Mandel RJ, Muzyczka N: Recombinant AAV viral vectors pseudotyped with viral capsids from serotypes 1, 2, and 5 display differential efficiency and cell tropism after delivery to different regions of the central nervous system. Mol Ther 2004, 10: 302–317. 10.1016/j.ymthe.2004.05.024PubMedCrossRef Burger C, Gorbatyuk OS, Velardo MJ, Peden CS, Williams P, Zolotukhin S, Reier PJ, Mandel RJ, Muzyczka N: Recombinant AAV viral vectors pseudotyped with viral capsids from serotypes 1, 2, and 5 display differential efficiency and cell tropism after delivery to different regions of the central nervous system. Mol Ther 2004, 10: 302–317. 10.1016/j.ymthe.2004.05.024PubMedCrossRef
54.
go back to reference Zolotukhin S, Byrne BJ, Mason E, Zolotukhin I, Potter M, Chestnut K, Summerford C, Samulski RJ, Muzyczka N: Recombinant adeno-associated virus purification using novel methods improves infectious titer and yield. Gene Ther 1999, 6: 973–985. 10.1038/sj.gt.3300938PubMedCrossRef Zolotukhin S, Byrne BJ, Mason E, Zolotukhin I, Potter M, Chestnut K, Summerford C, Samulski RJ, Muzyczka N: Recombinant adeno-associated virus purification using novel methods improves infectious titer and yield. Gene Ther 1999, 6: 973–985. 10.1038/sj.gt.3300938PubMedCrossRef
55.
go back to reference Matsushita T, Elliger S, Elliger C, Podsakoff G, Villarreal L, Kurtzman GJ, Iwaki Y, Colosi P: Adeno-associated virus vectors can be efficiently produced without helper virus. Gene Ther 1998, 5: 938–945. 10.1038/sj.gt.3300680PubMedCrossRef Matsushita T, Elliger S, Elliger C, Podsakoff G, Villarreal L, Kurtzman GJ, Iwaki Y, Colosi P: Adeno-associated virus vectors can be efficiently produced without helper virus. Gene Ther 1998, 5: 938–945. 10.1038/sj.gt.3300680PubMedCrossRef
56.
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 Meth 1994, 53: 55–63. 10.1016/0165-0270(94)90144-9CrossRef Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL: Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Meth 1994, 53: 55–63. 10.1016/0165-0270(94)90144-9CrossRef
57.
go back to reference Milligan ED, O'Connor KA, Nguyen KT, Armstrong CB, Twining C, Gaykema R, Holguin A, Martin D, Maier SF, Watkins LR: Intrathecal HIV-1 envelope glycoprotein gp120 enhanced pain states mediated by spinal cord proinflammatory cytokines. J Neurosci 2001, 21: 2808–2819.PubMed Milligan ED, O'Connor KA, Nguyen KT, Armstrong CB, Twining C, Gaykema R, Holguin A, Martin D, Maier SF, Watkins LR: Intrathecal HIV-1 envelope glycoprotein gp120 enhanced pain states mediated by spinal cord proinflammatory cytokines. J Neurosci 2001, 21: 2808–2819.PubMed
58.
go back to reference Milligan ED, Mehmert KK, Hinde JL, Harvey LOJ, Martin D, Tracey KJ, Maier SF, Watkins LR: Thermal hyperalgesia and mechanical allodynia produced by intrathecal administration of the Human Immunodeficiency Virus-1 (HIV-1) envelope glycoprotein, gp120. Brain Res 2000, 861: 105–116. 10.1016/S0006-8993(00)02050-3PubMedCrossRef Milligan ED, Mehmert KK, Hinde JL, Harvey LOJ, Martin D, Tracey KJ, Maier SF, Watkins LR: Thermal hyperalgesia and mechanical allodynia produced by intrathecal administration of the Human Immunodeficiency Virus-1 (HIV-1) envelope glycoprotein, gp120. Brain Res 2000, 861: 105–116. 10.1016/S0006-8993(00)02050-3PubMedCrossRef
59.
go back to reference Treutwein B, Strasburger H: Fitting the psychometric function. Percept Psychophys 1999, 61: 87–106.PubMedCrossRef Treutwein B, Strasburger H: Fitting the psychometric function. Percept Psychophys 1999, 61: 87–106.PubMedCrossRef
60.
go back to reference Harvey LOJ: Efficient estimation of sensory thresholds. Behav Res Meth Instrum Comput 1986, 18: 623–632.CrossRef Harvey LOJ: Efficient estimation of sensory thresholds. Behav Res Meth Instrum Comput 1986, 18: 623–632.CrossRef
61.
go back to reference Hargreaves K, Dubner R, Brown F, Flores C, Joris J: A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 1998, 32: 77–88. 10.1016/0304-3959(88)90026-7CrossRef Hargreaves K, Dubner R, Brown F, Flores C, Joris J: A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 1998, 32: 77–88. 10.1016/0304-3959(88)90026-7CrossRef
62.
go back to reference Lockwood LL, Silbert LH, Laudenslager ML, Watkins LR, Maier SF: Anesthesia-induced modulation of in vivo antibody levels: a study of pentobarbital, chloral hydrate, methoxyflurane, halothane, and ketamine/xylazine. Anesthes Analg 1993, 77: 769–774.CrossRef Lockwood LL, Silbert LH, Laudenslager ML, Watkins LR, Maier SF: Anesthesia-induced modulation of in vivo antibody levels: a study of pentobarbital, chloral hydrate, methoxyflurane, halothane, and ketamine/xylazine. Anesthes Analg 1993, 77: 769–774.CrossRef
63.
go back to reference Sato W, Enzan K, Masaki Y, Kayaba M, Suzuki M: The effect of isoflurane on the secretion of TNF-alpha and IL-1 beta from LPS-stimulated human peripheral blood monocytes. Masui 1995, 44: 971–975.PubMed Sato W, Enzan K, Masaki Y, Kayaba M, Suzuki M: The effect of isoflurane on the secretion of TNF-alpha and IL-1 beta from LPS-stimulated human peripheral blood monocytes. Masui 1995, 44: 971–975.PubMed
64.
go back to reference Miller LS, Morita Y, Rangan U, Kondo S, Clemens MG, Bulkley GB: Suppression of cytokine-induced neutrophil accumulation in rat mesenteric venules in vivo by general anesthesia. Int J Microcirc Clin Exp 1996, 16: 147–154.PubMedCrossRef Miller LS, Morita Y, Rangan U, Kondo S, Clemens MG, Bulkley GB: Suppression of cytokine-induced neutrophil accumulation in rat mesenteric venules in vivo by general anesthesia. Int J Microcirc Clin Exp 1996, 16: 147–154.PubMedCrossRef
65.
go back to reference Tas PW, Kress HG, Koschel K: General anesthetics can competitively interfere with sensitive membrane proteins. Proc Natl Acad Sci U S A 1987, 84: 5972–5975.PubMedCentralPubMedCrossRef Tas PW, Kress HG, Koschel K: General anesthetics can competitively interfere with sensitive membrane proteins. Proc Natl Acad Sci U S A 1987, 84: 5972–5975.PubMedCentralPubMedCrossRef
66.
go back to reference Feinstein DL, Murphy P, Sharp A, Galea E, Gavrilyuk V, Weinberg G: Local anesthetics potentiate nitric oxide synthase type 2 expression in rat glial cells. J Neurosurg Anesthesiol 13: 99–105. 10.1097/00008506-200104000-00006 Feinstein DL, Murphy P, Sharp A, Galea E, Gavrilyuk V, Weinberg G: Local anesthetics potentiate nitric oxide synthase type 2 expression in rat glial cells. J Neurosurg Anesthesiol 13: 99–105. 10.1097/00008506-200104000-00006
67.
go back to reference Mantz J, Cordier J, Giaume C: Effects of general anesthetics on intercellular communications mediated by gap junctions between astrocytes in primary culture. Anesthesiology 1993, 78: 892–901.PubMedCrossRef Mantz J, Cordier J, Giaume C: Effects of general anesthetics on intercellular communications mediated by gap junctions between astrocytes in primary culture. Anesthesiology 1993, 78: 892–901.PubMedCrossRef
68.
go back to reference Miyazaki H, Nakamura Y, Arai T, Kataoka K: Increase of glutamate uptake in astrocytes: a possible mechanism of action of volatile anesthetics. Anesthesiology 1997, 86: 1359–1366. 10.1097/00000542-199706000-00018PubMedCrossRef Miyazaki H, Nakamura Y, Arai T, Kataoka K: Increase of glutamate uptake in astrocytes: a possible mechanism of action of volatile anesthetics. Anesthesiology 1997, 86: 1359–1366. 10.1097/00000542-199706000-00018PubMedCrossRef
Metadata
Title
Controlling neuropathic pain by adeno-associated virus driven production of the anti-inflammatory cytokine, interleukin-10
Authors
Erin D Milligan
Evan M Sloane
Stephen J Langer
Pedro E Cruz
Marucia Chacur
Leah Spataro
Julie Wieseler-Frank
Sayamwong E Hammack
Steven F Maier
Terence R Flotte
John R Forsayeth
Leslie A Leinwand
Raymond Chavez
Linda R Watkins
Publication date
01-12-2005
Publisher
BioMed Central
Published in
Molecular Pain / Issue 1/2005
Electronic ISSN: 1744-8069
DOI
https://doi.org/10.1186/1744-8069-1-9

Other articles of this Issue 1/2005

Molecular Pain 1/2005 Go to the issue