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

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, 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

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.

Figures

Figure 1
Figure 1
Photomicrographs of beta-Galactosidase histochemistry; expression in spinal cord meninges. Spinal cords were removed from control, non-injected rat (A) or the AAV2-LacZ injected rat (B) 8 days after intrathecal injection. Beta-Galactosidase histochemistry was conducted with X-gal staining procedures. No sections are counter stained. Magnification in Panels A and B are identical, scale bar, 100 microns.
Figure 2
Figure 2
Adeno-associated viral IL-10 blocks development of chronic sciatic inflammatory neuropathy (SIN) induced mechanical allodynia. After baseline (BL) assessment on the von Frey test, all rats received intrathecal AAV2-GFP (Control, encoding green fluorescent protein) or AAV2-r-IL-10. Behavior was reassessed Day 3 after intrathecal AAV, confirming that neither AAV2-GFP (Control) nor AAV2-r-IL-10 affected behavior prior to peri-sciatic injections (F 7,88 = 0.686, p > 0.68). After this Day 3 assessment, unilateral peri-sciatic injections of 0 (vehicle control; Panels A, B), 4 ug zymosan (to induce ipsilateral allodynia; Panels C, D), or 160 ug zymosan (to induce bilateral allodynia; Panels E, F) were delivered, with repeated re-administration across days to induce a chronic neuropathic state. Repeated measures ANOVA revealed reliable main effects of peri-sciatic zymosan dose (F 1,40 = 12.093, p < 0.002), IL-10 (F 1,40 = 69.829, p < 0.0001), and laterality (F 1,40 = 22.315, p < 0.0001), and interactions between zymosan dose and IL-10 (F 1,40 = 6.161, p < 0.02) and between IL-10 and laterality (F 1,40 = 15.412, p < 0.001). The construct pTR2-CB-r-IL-10 employed in an AAV vector for behavioral testing induced the production and release of rat IL-10 from transfected IB3 cells in culture. Increases in rat IL-10 protein were detected in supernatants of transfected cells versus untransfected vehicle control cultures (Panel A Inset). Neither AAV2-GFP nor AAV2-r-IL-10 affected the behavioral responses of rats receiving chronic peri-sciatic vehicle, as illustrated by data obtained from the hindpaws ipsilateral (Panel A) or contralateral (Panel B) to the peri-sciatic injections. Allodynia was induced in the ipsilateral hindpaw of intrathecal AAV2-GFP rats receiving 4 ug peri-sciatic zymosan (Panel C). This allodynia was largely blocked by AAV2-r-IL-10 (p > 0.045 through Day 11 compared to BL) with allodynia reappearing on Day 13 after AAV; that is, 10 days after initiation of chronic zymosan. Again, neither AAV2-GFP nor AAV2-r-IL-10 affected behaviors obtained from the contralateral, non-allodynic hindpaws (Panel D). Allodynia was induced in both the ipsilateral (Panel E) and contralateral (Panel F) hindpaws of intrathecal AAV2-GFP rats receiving 160 ug peri-sciatic zymosan. These ipsilateral and contralateral allodynias were largely blocked by AAV2-r-IL-10 (p > 0.15 through Day 11 compared to BL), until allodynia reappeared on Day 13 after AAV; that is, 10 days after initiation of chronic zymosan.
Figure 3
Figure 3
Adeno-associated viral IL-10 reverses established CCI-induced thermal hyperalgesia. After predrug (baseline; BL) assessment on the Hargreaves test, sham (Panels A, B) or CCI (Panels C, D) surgery was performed (timing denoted by the first vertical dotted line). Behavioral assessments were reassessed Days 3 and 10 after surgery to document the lack of thermal hyperalgesia in sham operated rats and development of unilateral allodynia in CCI groups ipsilateral to sciatic surgery. ANOVA revealed reliable main effects of CCI (F 1,40 = 140.740, p < 0.0001) and laterality (F 1,38 = 48.901, p < 0.0001), and an interaction between CCI and laterality (F 1,40 = 104.295, p < 0.0001). After the Day 10 assessment, rats received intrathecal injections of either AAV2-GFP (Control) or AAV2-r-IL-10 (timing denoted by the second vertical dotted line). Behavioral assessments were again recorded on Days 13, 15, 17, 19, 21, 24, 26, and 30 after surgery; that is, Days 3, 5, 7, 9, 11, 14, 16, and 20 days after AAV. While neither AAV2-GFP nor AAV2-r-IL-10 exerted marked effects in sham operated animals (Panels A, B) or non-allodynic hindpaws of CCI-operated animals (Panel D), AAV2-r-IL-10 transiently reversed ipsilateral CCI allodynia compared to CCI operated AAV2-GFP treated animals (Panel C). For Days 13–26, ANOVA revealed reliable main effects of CCI (F 1,39 = 134.036, p < 0.0001), IL-10 (F 1,39 = 12.047, p < 0.01) and laterality (F 1,39 = 66.284, p < 0.0001), and interactions between CCI and AAV2-r-IL-10 (F 1,39 = 24.486, p < 0.0001), CCI and laterality (F 1,39 = 91.956, p < 0.0001), and IL-10 and laterality (F 1,39 = 17.392, p < 0.0001). At Day 30, behavioral responses were not significantly different from Day 10 preinjection levels (F 1,39 = 7.824, p > 0.10).
Figure 4
Figure 4
Adeno-associated viral IL-10 attenuates established CCI-induced mechanical allodynia. After predrug (baseline; BL) assessment on the von Frey test, sham (Panels A, B) or CCI (Panels C, D) surgery was performed (timing denoted by the first vertical dotted line). Behavioral assessments were reassessed Days 3 and 10 after surgery to document the lack of allodynia in sham operated rats and development of bilateral allodynia in CCI groups. ANOVA revealed reliable main effects of CCI (F 1,40 = 197.446, p < 0.0001) and laterality (F 1,40 = 6.356, p < 0.05). After the Day 10 assessment, rats received intrathecal (i.t.) injections of either AAV2-GFP (Control) or AAV2-r-IL-10 (timing denoted by the second vertical dotted line). Behavioral assessments were again recorded on Days 13, 15, 17, 19, 21, 24, 26, and 30 after surgery; that is, Days 3, 5, 7, 9, 11, 14, 16, and 20 days after AAV. While neither AAV2-GFP nor AAV2-r-IL-10 exerted marked effects in sham operated animals (Panels A, B), AAV2-r-IL-10 transiently attenuated bilateral CCI allodynia compared to CCI operated AAV2-GFP treated animals (Panels C, D). For Days 13–26, ANOVA revealed reliable main effects of CCI (F 1,40 = 496.336, p < 0.0001), IL-10 (F 1,40 = 59.636, p < 0.0001), and laterality (F 1,40 = 28.565, p < 0.0001), and interactions between CCI and IL-10 (F 1,40 = 72.988, p < 0.0001) and CCI and laterality (F 1,40 = 9.325, p < 0.01). At Day 30, behavioral responses were not significantly different from Day 10 preinjection levels (F 1,40 = 0.696, p > 0.40).

References

    1. 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. doi: 10.1016/S0885-3924(00)00218-9.
    1. 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. doi: 10.1016/S0304-3959(96)03140-5.
    1. McQuay H, Carroll D, Jadad AR, Wiffen P, Moore A. Anticonvulsant drugs for management of pain: a systematic review. Brit Med J. 1995;311:1047–1052.
    1. DeLeo JA, Tanga FY, Tawfik V. Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. The Neuroscientist. 2004;10:40–52. doi: 10.1177/1073858403259950.
    1. Watkins LR, Maier SF. Glia: a novel drug discovery target for clinical pain. Nature Reviews Drug Discovery. 2003;2:973–985. doi: 10.1038/nrd1251.
    1. Watkins LR, Milligan ED, Maier SF. Glial activation: a driving force for pathological pain. Trends Neurosci. 2001;24:450–455. doi: 10.1016/S0166-2236(00)01854-3.
    1. Watkins LR, Maier SF. Targeting glia to control clinical pain: An idea whose time has come. Drug Discovery Today: Therapeutic Strategies. 2004.
    1. 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. doi: 10.1007/BF00227104.
    1. Hassel B, Paulsen RE, Johnson A, Fonnum F. Selective inhibition of glial cell metabolism by fluorocitrate. Brain Res. 1992;249:120–124. doi: 10.1016/0006-8993(92)90616-H.
    1. 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.
    1. 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. doi: 10.1002/jnr.10743.
    1. 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.
    1. 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. doi: 10.1124/jpet.103.052407.
    1. 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
    1. 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. doi: 10.1016/S0306-4522(00)00574-1.
    1. 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.
    1. 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.
    1. 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. doi: 10.1016/S0304-3959(00)00281-5.
    1. Moore KW, Ho ASY, Xu-Amano J. Molecular biology of interleukin-10 and its receptor. In: DeVries JE and de Waal Malefyt R, editor. Interleukin-10. , R.G. Landes Company; 1995. pp. 1–9.
    1. 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. doi: 10.1016/S0304-3959(01)00354-2.
    1. 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. doi: 10.1016/0304-3959(88)90209-6.
    1. 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. doi: 10.1016/j.jpain.2004.06.006.
    1. 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.
    1. 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. doi: 10.1111/j.1460-9568.2004.03709.x.
    1. 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. doi: 10.1016/S0006-8993(97)01422-4.
    1. Milligan ED, Maier SF, Watkins LR. Sciatic Inflammatory Neuropathy (SIN) in the rat: Surgical procedures, induction of inflammation and behavioral testing. In: Luo ZD, editor. Pain Research: Methods and Protocols. Vol. 99. Totowa, Humana Press; 2004. pp. 67–89.
    1. 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. doi: 10.1046/j.1529-8027.2001.006001111.x.
    1. Yoshimura A, Mori H, Ohishi M, Aki D, Hanada T. Negative regulation of cytokine signaling influences inflammation. Curr Opion Immunol. 2003;15:704–708. doi: 10.1016/j.coi.2003.09.004.
    1. Opal SM, DePalo VA. Anti-inflammatory cytokines. Chest. 2000;117:1162–1172. doi: 10.1378/chest.117.4.1162.
    1. Knight D. Leukaemia inhibitory factor (LIF): a cytokine of emerging importance in chronic airway inflammation. Pulm Pharmacol Ther. 2001;14:169–176. doi: 10.1006/pupt.2001.0282.
    1. 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. doi: 10.1096/fj.99-1003rev.
    1. 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.
    1. 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.
    1. 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. doi: 10.1046/j.1460-9568.2000.01348.x.
    1. 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. doi: 10.1111/j.1460-9568.2004.03514.x.
    1. Laughlin TM, Bethea JR, Yezierski RP, Wilcox GL. Cytokine involvement in dynorphin-induced allodynia. Pain. 2000;84:159–167. doi: 10.1016/S0304-3959(99)00195-5.
    1. 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. doi: 10.1016/j.pain.2003.12.023.
    1. 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. doi: 10.1006/exnr.2000.7604.
    1. 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. doi: 10.1054/jpai.2003.11.
    1. 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. doi: 10.1016/j.neuroscience.2004.01.004.
    1. 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. doi: 10.1016/S0165-5728(03)00031-6.
    1. 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. doi: 10.1016/S0306-4522(03)00398-1.
    1. 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.
    1. 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. doi: 10.1046/j.1460-9568.2000.00090.x.
    1. Brewer KL, Bethea JR, Yezierski RP. Neuroprotective effects of interleukin-10 folowing spinal cord injury. Exp Neurol. 1999;159:484–493. doi: 10.1006/exnr.1999.7173.
    1. Buchschacher GLJ, Wong-Staal F. Development of lentiviral vectors for gene therapy for human diseases. Blood. 2000;95:2499–2504.
    1. 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.
    1. Daly TM. Overview of adeno-associated viral vectors. Methods Mol Biol. 2004;246:157–165.
    1. 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. doi: 10.1016/j.expneurol.2004.05.014.
    1. 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. doi: 10.1089/104303403765701187.
    1. 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.
    1. 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. doi: 10.1016/S0042-6822(03)00162-4.
    1. 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. doi: 10.1016/j.ymthe.2004.05.024.
    1. 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. doi: 10.1038/sj.gt.3300938.
    1. 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. doi: 10.1038/sj.gt.3300680.
    1. 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. doi: 10.1016/0165-0270(94)90144-9.
    1. 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.
    1. 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. doi: 10.1016/S0006-8993(00)02050-3.
    1. Treutwein B, Strasburger H. Fitting the psychometric function. Percept Psychophys. 1999;61:87–106.
    1. Harvey LOJ. Efficient estimation of sensory thresholds. Behav Res Meth Instrum Comput. 1986;18:623–632.
    1. 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. doi: 10.1016/0304-3959(88)90026-7.
    1. 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.
    1. 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.
    1. 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.
    1. 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.
    1. 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. doi: 10.1097/00008506-200104000-00006.
    1. 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.
    1. 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. doi: 10.1097/00000542-199706000-00018.

Source: PubMed

3
Se inscrever