• Non ci sono risultati.

(1)I Riferimenti bibliografici  Abood, M.E., Rizvi, G., Sallapudi, N., McAllister, S.D., 2001

N/A
N/A
Protected

Academic year: 2021

Condividi "(1)I Riferimenti bibliografici  Abood, M.E., Rizvi, G., Sallapudi, N., McAllister, S.D., 2001"

Copied!
25
0
0

Testo completo

(1)

I

Riferimenti bibliografici

 Abood, M.E., Rizvi, G., Sallapudi, N., McAllister, S.D., 2001. Activation of the CB1 cannabinoid receptor protects cultured mouse spinal neurons against excitotoxicity. Neurosci.

Lett. 309, 197–201.

 Abrahamsen B., Zhao J., Asante C.O., Cendan C.M., Marsh S., Martinez- Barbera J.P., Nassar M.A., Dickenson A.H., and Wood, J.N.:. The cell and molecular basis of mechanical, cold, and inflammatory pain. Science 321, 702–705. of fractalkine. Brain Behav. Immun. 21, 642–651(2008)

 Abramo A., Di Salvo C., Fortran F., Forfori F., Anselmino M., Giunta F.: Xenon anesthesia improves respiratory gas exchanges in morbidly obese patients. Journal of Obesity Volume 2010, Article ID 421593, 5 pages doi:10.1155/2010/421593

 Adolph O., Köster S.,Georgieff M.,Bäder S.,Föhr K .J.,Kammer T., Herrnberger B., Grön G.: Xenon-induced changes in CNS sensitization to pain. NeuroImage 49, 2010 720–730

 Agnati, L.F., Fuxe, K., Nicholson, C., Sykova’, E. (Eds.), 2000. Progress in Brain Research, Vol. 125. Elsevier, Amsterdam, p. 624

 Akopian A.N., Souslova V., England S., Okuse K., Ogata N., Ure J., Smith A., Kerr B.J., McMahon S.B., Boyce, S., et al.:The tetrodotoxin- resistant sodium channel SNS has a specialized function in pain pathways. Nat. Neurosci. 2, 541–548. (1999)

 Albarran, F.A., Roa, J.P., Navarrete, R., Castillo, R., Nualart, F., Aguayo, L.G., 2001. Effect of protein kinase C activation on the glycine evoked Cl current in spinal cord neurons. Brain Res. 902, 1–10.

 Albarran, F.A., Roa, J.P., Navarrete, R., Castillo, R., Nualart, F., Aguayo, L.G., 2001. Effect of protein kinase C activation on the glycine evoked Cl current in spinal cord neurons. Brain Res. 902, 1–10.

 Alessandri-Haber N., Dina O.A., Joseph E.K., Reichling D., Levine J.D.: A transient receptor potential vanilloid 4-dependent mechanism of hyperalgesia is engaged by concerted action of inflammatory mediators. J. Neurosci. 26, 3864–3874 (2006)

 Aley KO, Levine JD. Different peripheral mechanisms mediate enhanced nociception in metabolic/toxic and traumatic painful peripheral neuropathies in the rat. Neuroscience 2002;111: 389–97.

 Alfredson H, Forsgren S, Thorsen K, Lorentzon R. In vivo microdialysis and

immunohistochemical analyses of tendon tissue demonstrated high amounts of free glutamate and glutamate NMDAR1 receptors, but no signs of inflammation, in Jumper’s knee. J Orthop Res 2001;19:881–6.

 Allen H.L., Iversen L.L.: Phencyclidine, dizocilpine, and cerebrocortical neurons (letter).

Science 1990; 247:221

 Almeida, A., Storkson, R., Lima, D., Hole, K., Tjolsen, A., 1999. The medullary dorsal reticular nucleus facilitates pain behaviour inducedby formalin in the rat. Eur. J. Neurosci.

11, 110–122.

 Almeida, A., Tavares, I., Lima, D., Coimbra, A., 1993. Descending projections from the medullary dorsal reticular nucleus make synaptic contacts with spinal cord lamina I cells projecting to that nucleus: an electron microscopic tracer study in the rat. Neuroscience 55, 1093– 1106.

 Almeida, A., Tjolsen, A., Lima, D., Coimbra, A., Hole, K., 1996. The medullary dorsal reticular nucleus facilitates acute nociception in the rat. Brain Res. Bull. 39, 7–15.

 Ameri, A., 1999. The effects of cannabinoids on the brain. Prog. Neurobiol. 58, 315–348.

 Apkarian, A.V., Thomas, P.S., Krauss, B.R., Szeverenyi, N.M., 2001. Prefrontal cortical hyperactivity in patients with sympathetically mediated chronic pain. Neurosci. Lett. 311, 193–197.

 Attal N., Bouhassira D., Gautron M., Vaillant J.N., Mitry E., Lepere C., Rougier P., Guirimand, F.:Thermal hyperalgesia as a marker of oxaliplatin neurotoxicity: a prospective quantified sensory assessment study. Pain 144, 245–252 (2009)

 Azami, J., Green, D.L., Roberts, M.H.T., Monhemius, R., 2001. The behavioural importance of dynamically activated descending inhibitionfrom the nucleus reticularis gigantocellularis pars alpha. Pain 92, 53–62.

(2)

II

 Azami, J., Green, D.L., Roberts, M.H.T., Monhemius, R., 2001. The behavioural importance of dynamically activated descending inhibition from the nucleus reticularis gigantocellularis pars alpha. Pain 92, 53–62.

 Azkue JJ, Zimmermann M, Hsieh TF, Herdegen T. Peripheral nerve insult induces NMDA receptor-mediated, delayed degeneration in spinal neurons. Eur J Neurosci 1998;10:2204–6.

 Baba, H., Khono, T., Okamoto, M., Goldstein, P.A., Shimoji, K., Yoshimura, M., 1998.

Muscarinic facilitation of GABA release in substantia gelatinosa of the rat spinal dorsal horn.

J. Physiol. 508, 83-93.

 Baddeley A: Working memory: Looking back and looking forward. Nat Rev Neurosci 2003;

4:829–39

 Baier PC, Ondo WG, Winkelmann J. Animal studies in restless legs syndrome. Mov Disord 2007;2(S18):S459–S465.

 Baines C.P., Wang L., Cohen M.V., Downey J.M.: Protein tyrosine kinase is downstream of protein kinase C for ischemic preconditioning’s anti-infarct effect in the rabbit heart. J Mol Cell Cardiol 1998; 30:383–92

 Bandell, M., Story, G.M., Hwang, S.W., Viswanath, V., Eid, S.R., Petrus, M.J., Earley, T.J., and Patapoutian, A. : Noxious cold ion channel TRPA1 is activated by pungent compounds and bradykinin. Neuron 41, 849–857 (2004)

 Barberis, C., Morin, D., Durroux, T., Mouillac, B., Guillon, G., Seyer, R., Hibert, M., Tribollet, E., Manning, M., 1999. Molecular pharmacology of AVP and OT receptors and therapeutic potential. Drugs News Perspect. 12, 279–292.

 Barnard, E.A., 1996. The transmitter-gated channels: a range of receptor types and structures.

Trends Pharmacol. Sci. 17, 305–308.

 Barnard, E.A., 2000. The molecular architecture of the GABAA receptors. In: Mohler, H.

(Ed.), Handbook of Experimental Pharmacology: Pharmacology of Amino Acid Transmitters. Springer, Berlin,pp. 79–99.

 Barnes, N.M., Sharp, T., 1999. A review of central 5-HT receptors and their function.

Neuropharmacology 38, 1083–1152.

Basbaum A.I., D.M. Bautista, G. Scherrer, D. Julius : Molecular mechanisms of pain. Cell 139, 267-284 (2009)

 Basbaum A.I., Jessell T.: The perception of pain. In Principles of Neuroscience, E.R. Kandel, J. Schwartz, and T. Jessell, eds. (New York:Appleton and Lange), pp. 472–491 (2000).

 Basbaum, A.I., Fields, H.L., 1984. Endogenous pain control systems: brainstem spinal pathways and endorphin circuitry. Annu. Rev. Neurosci. 7, 309–338.

 Bautista D.M., Jordt S.E., Nikai T., Tsuruda P.R., Read A.J., Poblete J., Yamoah E.N., Basbaum A.I., Julius, D.: TRPA1 mediates the inflammatory actions of environmental irritants and proalgesic agents. Cell 124, 1269-1282 (2006)

 Bautista D.M., Sigal Y.M., Milstein A.D., Garrison J.L., Zorn J.A., Tsuruda P.R., Nicoll R.A., Julius, D. :Pungent agents from Szechuan peppers excite sensory neurons by inhibiting two-pore potassium channels. Nat. Neurosci. 11, 772–779 (2008)

 Bautista, D.M., Siemens, J., Glazer, J.M., Tsuruda, P.R., Basbaum, A.I., Stucky, C.L., Jordt, S.E., and Julius, D.: The menthol receptor TRPM8 is the principal detector of environmental cold. Nature 448, 204–208 (2007)

 Bayer KU, De Koninck P, Leonard AS, et al. Interaction with the NMDA receptor locks CaMKII in an active conformation. Nature 2001;411:801–5.

 Bedi A., McBride W.T., Armstrong M.A., Murray J.M., Fee J.P.H.: Xenon has no effect on cytokine balance and adhesion molecule expression within an isolated cardiopulmonary bypass system. Br J Anaesth 2002; 89:546–50

 Beirith A, Santos AR, Calixto JB. Mechanisms underlying the nociception and paw oedema caused by injection of glutamate into the mouse paw. Brain Res 2002;924:219–28.

 Benedetti, F., 1997. Cholecystokinin type A and type B receptors and their modulation of opioid analgesia. News Physiol. Sci. 12, 263–268.

 Benrath J., Kempf C., Georgieff M., Sandkühler J.: Xenon blocks the induction of synaptic long term-potentiation in pain pathways in rat spinal cord in vivo. Anesthesia Analgesia 2007;104:106 –11

 Bernard S.A., Gray T.W., Buist M.D., Jones B.M., Silvester W., Gutteridge G., Smith K.:

Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia.

N Engl J Med 2002; 346:557–63

(3)

III

 Bernardi M, Bertolini A, Szczawinska K, Genedani S. Blockade of the polyamine site of NMDA receptors produces antinociception and enhances the effect of morphine, in mice. Eur J Pharmacol 1996;298:51–5.

 Bessac B.F., Jordt S.E.: Breathtaking TRP channels: TRPA1 and TRPV1 in airway chemosensation and reflex control. Physiology (Bethesda) 23, 360-370 (2008)

 Bester, H., De Felipe, C., Hunt, S.P., 2001. The NK1 receptor is essential for the full expression of noxious inhibitory controls in the mouse. J. Neurosci. 21, 1039–1046.

 Bhattacharya M.R., Bautista D.M., Wu K., Haeberle H., Lumpkin E.A., Julius, D.: Radial stretch reveals distinct populations of mechanosensitive mammalian somatosensory neurons.

Proc. Natl. Acad. Sci. USA 105, 20015–20020 (2008)

 Bie B, Fields HL, Williams JT, Pan ZZ. Roles of alpha1- andalpha2-adrenoceptors in the nucleus raphe magnus in opioid analgesia and opioid abstinence-induced hyperalgesia.

JNeurosci. 2003;23:7950–7957.

 Bie, B.H., Zhao, Z.Q., 2001. Nitric oxide inhibits GABA-evoked current in dorsal root ganglion neuron via PKG-dependent pathway. Brain Res. Bull. 55, 335–339.

 Bodner M, Shafi M, Zhou YD, Fuster JM: Patterned firing of parietal cells in a haptic working memory task. Eur J Neurosci 2005; 21:2538–46

 Bosnjak Z.J., Supan F.D., Rusch N.J.: The effects of halothane, enflurane, and isoflurane on calcium current in isolated canine ventricular cells. Anesthesiology 1991; 74:340–5

 Bourgeais, L., Monconduit, L., Villanueva, L., Bernard, J.F., 2001. Parabrachial internal lateral neurons convey nociceptive messages from the deep laminae of the dorsal horn to the intralaminar thalamus. J. Neurosci. 21, 2159–2165.

 Boyce S, Wyatt A, Webb JK, et al. Selective NMDA NR2B antagonists induce

antinociception without motor dysfunction: correlation with restricted localisation of NR2B subunit in dorsal horn. Neuropharmacology 1999;38:611–23.

 Brandao, M.L., Anseloni, V.Z., Pandossio, J.E., De Araujo, J.E., Castilho, V.M., 1999.

Neurochemical mechanisms of the defensive behavior in the dorsal midbrain. Neurosci.

Biobehav. 23, 863–875.

 Breivogel, C.S., Griffin, G., Di Marzo, V., Martin, B.R., 2001. Evidence for a new G protein- coupled cannabinoid receptor in mouse brain.Mol. Pharmacol. 60, 155–163

 Bridges, D., Ahmad, K., Rice, A.S.C., 2001. The synthetic cannabinoid WIN55,212-2 attenuates hyperalgesia and allodynia in a rat model of neuropathic pain. Br. J. Pharmacol.

133, 586–594.

 Brown D.A., Passmore G.M.:Neural KCNQ (Kv7) channels. Br. J. Pharmacol. 156, 1185–

1195 (2009)

 Brown, R.E., Stevens, D.R., Haas, H.L., 2001. The physiology of brain histamine. Prog.

Neurobiol. 63, 637–672.

 Bruggemann I, Schulz S, Wiborny D, Hollt V. Colocalization of the mu-opioid receptor and calcium/calmodulin-dependent kinase II in distinct pain-processing brain regions. Brain Res Mol Brain Res 2000;85:239–50.

 Bryant B.P., Mezine I.: Alkylamides that produce tingling paresthesia activate tactile and thermal trigeminal neurons. Brain Res. 842, 452–460 (1999)

 Budal, D., Harasawa, I., Fields, H.L., 1998. Midbrain periaqueductal gray (PAG) inhibits nociceptive inputs to sacral dorsal horn nociceptive neurons through _2-adrenergic receptors.

J. Neurophysiol. 80, 2244–2254.

 C. L. Renn, S. G. Dorsey: The physiology and processing of pain: a review. AACN, volume 16, numero 3, 277-90 (2005)

 Caceres A.I., Brackmann M., Elia M.D., Bessac B.F., del Camino D., D’Amours M., Witek J.S., Fanger C.M. Chong J.A., Hayward N.J.,: A sensory neuronal ion channel essential for airway inflammationand hyperreactivity in asthma. Proc. Natl. Acad. Sci. USA 106, 9099- 9104 (2009)

 Caceres, A.I., Brackmann, M., Elia, M.D., Bessac, B.F., del Camino, D., D’Amours, M., Witek, J.S., Fanger, C.M., Chong, J.A., Hayward, N.J., et al. : A sensory neuronal ion channel essential for airway inflammationand hyperreactivity in asthma. Proc. Natl. Acad.

Sci. USA 106, 9099–9104 (2009)

 Calejesan, A.A., Kim, S.J., Zhou, M., 2000. Descending facilitatory modulation of a behavioral nociceptive response by stimulation in the adult rat anterior cingulate cortex. Eur.

J. Pain 4, 83–96.

(4)

IV

 Cambier J., Masson M., Dehen H.: Neurologia (decima edizione italiana). Casa editrice Elsevier (2003)

 CaoY.Q.: Voltage-gated calcium channels and pain. Pain 126, 5–9 (2006)

 Carlton S.M., Westlund K.N., Zhang D. et al.: Calcitonin gene-related peptide containing primary afferent fibers synapse on primate spinothalamic tract cells. Neurosci. Lett. 109, 76- 81 (1990)

 Carlton SM, Coggeshall RE. Inflammation-induced changes in peripheral glutamate receptor populations. Brain Res 1999;820: 63–70.

 Carlton SM, Hargett GL, Coggeshall RE. Localization and activation of glutamate receptors in unmyelinated axons of rat glabrous skin. Neurosci Lett 1995;197:25–8.

 Carlton SM. Localization of CaMKIIalpha in rat primary sensory neurons: increase in inflammation. Brain Res 2002;947: 252–9.

 Carruthers, A.M., Sellers, L.A., Jenkins, D.W., Jarvie, E.M., Feniuk, W., Humphrey, P.P.A., 2001. Adenosine A1 receptor-mediated inhibition of protein kinase A-induced calcitonin gene-related peptide release from rat trigeminal neurons. Mol. Pharmacol. 59, 1533–1541.

 Cason B.A., Gamperl A.K., Slocum R.E., Hickey R.F.: Anesthetic-induced preconditioning:

Previous administration of isoflurane decreases myocardial infarct size in rabbits.

Anesthesiology 1997; 87:1182–90

 Castro-Lopes JM, Tavares I, Coimbra A. GABA decreases in the spinal cord dorsal horn after peripheral neurectomy. Brain Res 1993;620:287–91.

 Castroman PJ, Ness TJ. Ketamine, an N-methyl-d-aspartate receptor antagonist, inhibits the reflex responses to distension of the rat urinary bladder. Anesthesiology 2002;96:1401–9.

 Castroman PJ, Ness TJ. Ketamine, an N-methyl-d-aspartate receptor antagonist, inhibits the spinal neuronal responses to distension of the rat urinary bladder. Anesthesiology 2002;96:

1410–9.

 Caterina M.J., Rosen T.A., Tominaga M., Brake A.J., Julius, D : A capsaicin-receptor homologue with a high threshold for noxious heat. Nature 398, 436–441 (1999)

 Caterina M.J., Schumacher M.A., Tominaga M., Rosen T.A., Levine J.D., Julius D.: The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 389, 816-824 (1997)

 Caterina, M.J., Leffler A., Malmberg A.B., Martin W.J., Trafton J., Petersen-Zeitz K.R., Koltzenburg M., Basbaum A.I., Julius, D.: Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science, 288, 306-313 (2000)

 Cattano D., Valleggi S., Daqing M., Kastsiuchenka O., Abramo A., Sun P., Cavazzana A.O., Natale G., Maze M., Giunta F.: Xenon induces transcription of ADNP in neonatal rat brain.

Neuroscience letters 440, 217-221 (2008)

 Celuch, S.M., 1995. Possible participation of histamine H3 receptors in the modulation of noradrenaline release from rat spinal cord slices. Eur. J. Pharmacol. 287, 127–133.

 Chahl LA. Opioids – mechanism of action. Aust Prescr 1996; 19:63-65.

 Chalfie M.: Neurosensory mechanotransduction. Nat. Rev. Mol.Cell Biol. 10, 44–52 (2009)

 Chao, M.V.: Neurotrophins and their receptors: a convergence point for many signalling pathways. Nat. Rev. Neurosci. 4, 299–309 (2003)

 Charles, K.J., Evans, M.L., Robbins, M.J., Calver, A.R., Leslie, R.A., Pangalos, M.N., 2001.

Comparative immunohistochemical localization

 Chatterton JE, Awobuluyi M, Premkumar LS, et al. Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 2002;415:793–8.

 Chen X., Alessandri-Haber N., Levine J.D.: Marked attenuation of inflammatory mediator- induced C-fiber sensitization for mechanical and hypotonic stimuli in TRPV4−/− mice. Mol.

Pain 3, 31 (2007)

 Chizh BA, Headley PM, Tzschentke TM. NMDA receptor antagonists as analgesics: focus on the NR2B subtype. Trends Pharmacol Sci 2001;22:636–42.

 Chizh BA, Reissmuller E, Schlutz H, et al. Supraspinal vs spinal sites of the antinociceptive action of the subtype-selective NMDA antagonist ifenprodil. Neuropharmacology 2001;40:

212–20.

 Chuang, H.H., Prescott, E.D., Kong, H., Shields, S., Jordt, S.E., Basbaum, A.I., Chao, M.V., Julius, D.: Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2-mediated inhibition Nature 411, 957–962 (2001)

(5)

V

 Chung, M.K., Lee, H., Caterina, M.J. : Warm temperatures activate TRPV4 in mouse 308 keratinocytes. J. Biol. Chem. 278, 32037–32046 (2003)

 Ciabarra AM, Sullivan JM, Gahn LG, et al. Cloning and characterization of chi-1: a developmentally regulated member of a novel class of the ionotropic glutamate receptor family. J Neurosci 1995;15:6498–508.

 Coggeshall RE, Carlton SM. Ultrastructural analysis of NMDA, AMPA, and kainate receptors on unmyelinated and myelinated axons in the periphery. J Comp Neurol 1998;391:78–86.

 Coggeshall, R.E., Carlton, S.M., 1997. Receptor localization in the mammalian dorsal horn and primary afferent neurons. Brain Res. Rev. 24, 28–66.

 Collins J. C., Ren K.: WDR response profiles of spinal dordal horn neurons may be unmasked by barbiturate anesthesia. Pain 28 369-378 (1987)

 Cordero-Erausquin, M., Changeux, J.P., 2001. Tonic nicotinic modulation of serotonergic transmission in the spinal cord. Proc. Natl. Acad. Sci. U.S.A. 98, 2803–2807.

 Cox J.J., Reimann F., Nicholas A.K., Thornton G., Roberts E., Springell K., Karbani G., Jafri H., Mannan J., Raashid Y., et al.: An SCN9A channelopathy causes congenital inability to experience pain. Nature 444, 894–898 (2006)

 Cravatt, B.F., Demarest, K., Patricelli, M.P., Bracey, M.H., Giang, D.K., Martin, B.R., Lichtman, A.H., 2001. Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase. Proc. Natl. Acad. Sci.

U.S.A. 98, 9371–9376.

 Crombez G., Van Damme S., Eccleston C.: Hypervigilance to pain: an experimental and clinical analysis. Pain 116, 4-7 (2005)

 Cull-Candy S, Brickley S, Farrant M. NMDA receptor subunits: diversity, development and disease. Curr Opin Neurobiol 2001;11:327–35.

 Cussac, D., Newman-Tancredi, A., Quentric, Y., Carpentier, N., Poissonnet, G., Parmentier, J.G., Goldstein, S., Millan, M.J., 2002. Characterization of phospholipase C activity at h5- HT2C compared with h5-HT2B receptors: influence of novel ligands upon membrane-bound levels of [3H]phosphatidylinositols. Naunyn Schmiedebergs Arch. Pharmacol. 365, 242–252.

 D. Regoli, A. Boudon, J.L. Fauchere, Receptors and antagonists for substance P and related peptides, Pharmacol. Rev. 46 (1994) 551–599.

 Daniels S., Roberts R.J.: Post-synaptic inhibitory mechanisms of anaesthesia: Glycine receptors. Toxicol Lett 1998; 101:71–6

 David H.N., Haelewyn B., Risso J.J., Colloc’h N., Abraini J.H.: Xenon is an inhibitor of tissue-plasminogen activator: adverse and beneficial effects in a ratmodel of thromboembolic stroke. Journal of Cerebral Blood Flow & Metabolism 30, 718–728 (2010)

 David H.N., Leveille F., Chazalviel L., MacKenzie E.T., Buisson A., Lemaire M., Abraini J.H.: Reduction of ischemic brain damage by nitrous oxide and xenon. J Cereb Blood Flow Metab 2003; 23:1168–73

 Davidson EM, Carlton SM. Intraplantar injection of dextrorphan, ketamine or memantine attenuates formalin-induced behaviors. Brain Res 1998;785:136–42.

 Davies A., Hendrich J., Van Minh A.T., Wratten J., Douglas L., Dolphin A.C.: Functional biology of the alpha(2)delta subunits of voltage-gated calcium channels. Trends Pharmacol.

Sci. 28, 220–228 (2008)

 Davis J.B., Gray J., Gunthorpe M.J., Hatcher J.P., Davey P.T., Overend P., Harries M.H., Latcham J., Clapham C., Atkinson K.: Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature 405, 183-187 (2000)

 Davis MP, LeGrand SB, Lagman R. Look before leaping. Support Care Cancer 2005;

13:769-774.

 De Biasi, Rustioni S. e A.: Glutamate and substance P coexist in primary afferent terminals in the superficial laminae of spinal cord. Proc. Natl. Acad. Sci. U.S.A. 85, 7820-7824 (1988)

 De Rossi L.W., Brueckmann M., Rex S., Barderschneider M., Buhre W., Rossaint R.: Xenon and isoflurane differentially modulate lipopolysaccharideinduced activation of the nuclear transcription factor KB and production of

 De Rossi L.W., Gott K., Horn N.A., Hecker K., Hutschenreuter G., Rossaint R.: Xenon preserves neutrophil and monocyte function in human whole blood. Can J Anesth 2002;

49:942–5

(6)

VI

 De Rossi L.W., Horn N.A., Baumert H.J., Gutensohn K., Hutschenreuter G., Rossaint R.:

Xenon does not affect human platelet function in vitro. Anesthesia Analgesia 2001; 93:635–

40

 De Rossi L.W., Horn N.A., Stevanovic A., Buhre W., Hutschenreuter G., Rossaint R.: Xenon modulates neutrophil adhesion molecule expression in vitro. Eur J Anaesthesiol 2004;

21:139–43

 De Sousa SLM, Dickinson R, Lieb WR, Franks NP: Contrasting synaptic actions of the inhalational general anesthetics isoflurane and xenon. Anesthesiology 2000; 92:1055–66

 De Vries B., Frants R.R., Ferrari M.D., Van den Maagdenberg, A.M.: Molecular genetics of migraine. Hum. Genet. 126, 115–132 (2009)

 Decker, M.W., Meyer, M.D., 1999. Therapeutic potential of neuronal nicotinic acetylcholine receptor agonists as novel analgesics. Biochem. Pharmacol. 58, 917–923.

 Di Piero V., Jones A.K., Iannotti F., Powell M., Perani, D., Lenzi, G. F., Frackowiak R.S.:

Chronic pain: a PET study of the central effects of percutaneous high cervical cordotomy.

Pain 46, 9–12(1991)

 Dias R, Robbins TW, Roberts AC: Dissociation in prefrontal cortex of affective and attentional shifts. Nature 1996; 380:69–72

 Dib-Hajj S.D., Yang Y., Waxman S.G.: Genetics and molecular pathophysiology of Na(v)1.7-related pain syndromes. Adv. Genet. 63,85–110 (2008)

 Dick I.E., Brochu R.M., Purohit Y., Kaczorowski G.J., Martin W.J., Priest B.T.: Sodium channel blockade may contribute to the analgesic efficacy of antidepressants. J. Pain 8, 315–

324 (2007)

 Dickenson, A., Suzuki, R., Reeve, A., 2000. Adenosine as a potential analgesic target in inflammatory and neuropathic pains. CNS Drugs 13, 77–85.

 Dickinson R. Peterson B.K., Banks P. Simillis C. Martin J.C., Valenzuela C.A. et al.:

competitive inhitibion at the glycine site of the N-methyl-D-aspartate by anesthetics xenon and isoflurane: evidence from molecular modeling and electrophysiology. Anesthesiology, 2007; 107: 756-67

 Dildy-Mayfield JE, Eger EI II, Harris RA: Anesthetics produce subunitselective actions on glutamate receptors. J Pharmacol Exp Ther 1996; 276: 1058–65

 Dinse A., Föhr K.J., Georgieff M., Beyer C., Bulling A., Weigt H.U.: Xenon reduces glutamate-, AMPA-, and kainate-induced membrane currents in cortical neurones. Br J Anaesth 2005; 94:479–85

 Dobler T., Springauf A., Tovornik S., Weber M., Schmitt A., Sedlmeier R., Wischmeyer E., Doring F.: TRESK two-pore-domain K+ channels constitute a significant component of background potassium currents in murine dorsal root ganglion neurones. J. Physiol. 585, 867–879 (2007)

 Doubell TP, Mannion RJ, Woolf CJ. The dorsal horn: statedependent sensory processing, plasticity and the generation of

 Dougherty, P.M., Staats, P.S., 1999. Intrathecal drug therapy for chronic pain.

Anesthesiology 91, 1891–1918.

 Doyle M.W., Bailey T.W., Jin Y.H., Andresen M.C.: Vanilloid receptors presynaptically modulate cranial visceral afferent synaptic transmission in nucleus tractus solitarius. J Neurosci 22, 8222-8229 (2002)

 Drew L.J., Rohrer D.K., Price M.P., Blaver K.E., Cockayne D.A., Cesare P., Wood J.N.:

Acid-sensing ion channels ASIC2 and ASIC3 do not contribute to mechanically activated currents in mammalian sensory neurones. J. Physiol. 556, 691–710 (2004)

 Duggan, A.W., 2000. Neuropeptide spread in the brain and spinal cord.Prog. Brain Res. 125, 369–380.

 Dundee J.W., Moore J.: Alterations in response to somatic pain associated with anaesthesia.

IV. The effect of subanaesthetic concentrations of inhalation agents. British Journal of Anaesthesia 1960; 32: 453-459.

 Edwards R., Bingham C.O., 3rd, Bathon J., Haythornthwaite J.A.:Catastrophizing and pain in arthritis, fibromyalgia, and other rheumatic diseases. Arthritis Rheum. 15, 325-332 (2006)

 Eger R.P., MacLeod B.A. Anaesthesia by intravenous emulsified isoflurane in mice. Can J Anaesth 1995;42:173– 6.

 Eisenach, J.C., 1999. Muscarinic-mediated analgesia. Life Sci. 64, 549– 554.

(7)

VII

 Estacion M., Dib-Hajj S.D., Benke P.J., Te Morsche, R.H., Eastman E.M., Macala L.J., Drenth J.P.,Waxman S.G. NaV1.7 gain-of-function mutations as a continuum: A1632E displays physiological changes associated with erythromelalgia and paroxysmal extreme pain disorder mutations and produces symptoms of both disorders. J.Neurosci. 28, 11079–11088 (2008)

 Fairbanks CA, Stone LS, Kitto KF, Nguyen HO, Posthumus IJ, Wilcox GL. Alpha (2C)- adrenergic receptors mediate spinal analgesia and adrenergic-opioid synergy. J Pharmacol Exp Ther. 2002;300:282–290. 54 Asano T, Dohi S, Ohta S, Shimonaka H, Iida H.

Antinociception

 Fairhurst M., Wiech K., Dunckley P., Tracey I.: Anticipatory brainstem activity predicts neural processing of pain in humans. Pain 128, 101-110 (2007)

 Fang L, Wu J, Lin Q, Willis WD. Calcium-calmodulindependent protein kinase II contributes to spinal cord central sensitization. J Neurosci 2002;22:4196–204.

 Fertleman C.R., Baker M.D., Parker K.A., Moffatt S., Elmslie F.V., Abrahamsen B., Ostman J., Klugbauer N., Wood, J.N., Gardiner, R.M., e al.: SCN9A mutations in paroxysmal extreme pain disorder: allelic variants underlie distinct channel defects and phenotypes.

Neuron 52, 767–774 (2006)

 Fields H. L.: Pain. New York: McGraw-Hill (1987)

 Fields, H.L., 2000. Pain modulation: expectation, opioid analgesia and virtual pain. Prog.

Brain Res. 122, 245–253

 Fields, H.L., Basbaum, A.I., 1999. Central nervous system mechanisms of pain modulation.

In: Wall, P.D., Melzack, R. (Eds.), Textbook of Pain, 4th Edition. Churchill Livingston, Edinburgh, pp. 309–329.

 Fields, H.L., Heinricher, M.M., Mason, P., 1991. Neurotransmitters in nociceptive modulatory circuits. Annu. Rev. Neurosci. 14, 219–245.

 Fillingim R. B., King C. D., Ribeiro-Dasilva M.D.,Rahim-Williams B.,Riley J. L.: Sex, gender and pain: a review of clinical and experimental findings. The Journal of Pain, 10, 447- 485 (2009)

 Fisher K, Coderre TJ, Hagen NA. Targeting N-methyl-daspartate receptor for chronic pain management: preclinical animal studies, recent clinical experience and future research directions. J Pain Symptom Manage 2000;20:358–73.

 Flood P., Ramirez-Latorre J., Role L.: a4b2 neuronal nicotinic acetylcholine receptors in the central nervous system are inhibited by isoflurane and propofol, but a7-type nicotinic acetylcholine receptors are unaffected. Anesthesiology 1997; 86:859–65

 Flor H.: Phantom limb pain:characteristics, causes and treatments. Lancet Neurology 1, 182- 189 (2002)

 Fomitcheva I., Kosk-Kosicka D.: Volatile anesthetics selectively inhibit the Ca2_-

transporting ATPase in neuronal and erythrocyte plasma membranes. Anesthesiology 1996;

84:1189–95

 Fonseca, M.I., Ni, Y.G., Dunning, D.D., Miledi, R., 2001. Distribution of serotonin2A, serotonin2C and serotonin3 receptor mRNA in spinal cord and medulla oblongata. Mol.

Brain Res. 89, 11–19.

 Fox, A., Kesingland, A., Gentry, C., McNair, K., Patel, S., Urban, L., James, I., 2001. The role of central and peripheral cannabinoid1 receptors in the antihyperalgesic activity of cannabinoids in a model of neuropathic pain. Pain 92, 91–100.

 Franks NP, Dickinson R, De Sousa SL, Hall AC, Lieb WR: How does xenon produce anaesthesia? Nature 1998; 396:324 Letter

 Franks NP, Lieb WR: Molecular and cellular mechanisms of general anesthesia. Nature 1994; 367:607–14

 Freitas da Rocha R, Pereira A Jr, Bezerra Coutinho FA: N-methyl-D-aspartate channel and consciousness: From signal coincidence detection to quantum computing. Prog Neurobiol 2001; 64:555–73

 Froeba G., Georgieff M., Linder E.M., Föhr K.J., Weight H.U., Holsträter T.F., Kölle M.A:, Adolph O.: Intranasal application of xenon: describing the pharmacokinetics in experimental animals and the increased pain tolerance withina placebo-controlled experimental human study. British Journal of Anaesthesia 104 (3): 351–8 2010

 Frot, M., Mauguière, F., 2003. Dual representation of pain in the operculo-insular cortexin humans. Brain 126, 438–450.

(8)

VIII

 Fujinaga M., Maze M.: Neurobiology of nitrous oxide induced antinociceptive effects. Mol Neurobiol 2002; 25:167–89

 Fukuda K. Intravenous opioid anesthetics In Miller RD (ed). Miller’s Anesthesia .6th Ed.

Elsevier, Philadelphia, 2005.

 Fukuda T., Nishimoto C., Hisano S., Miyabe M., Toyooka H.: The analgesic effect of Xenon on the formalin test in rats: a comparison with Nitrous Oxide. Anesthesia Analgesia 2002;

95:1300–4

 Gage, H.E., Gage, J.C., Tobin, J.R., Chiari, A., Tong, C., Xu, Z.M., Mach, R.H., Efange, S.M., Ehrenkaufer, R.L., Eisenach, J.C., 2001. Morphine-induced spinal cholinergic activation: in vivo imaging with positron emission tomography. Pain 91, 139–145.

 Galley H.F., Le Cras A.E., Logan S.D., Webster N.R.: Differential nitric oxide synthase activity, cofactor availability and cGMP accumulation in the central nervous system during anaesthesia. Br J Anaesth 2001; 86:388–94

 Gallo AM.: The fifth vital sign: implementation of the neonatal infant pain scale. J Obstet Gynecol Neonatal Nurse. Mar-Apr; 32 (2): 199-206 (2003)

 Gamse R., Molnar A., Lembeck F.: Substance P release from spinal cord slices by capsaicin.

Life Sci. 25 629-636 (1979)

 Gao, K., Mason, P., 2000. Serotonergic raphe magnus cells that respond to noxious tail heat are not on or off cells. J. Neurophysiol. 84, 1719–1725.

 Gao, X., Zhang, Y.Q., Wu, G.C., 2000. Effects of dopaminergic agents on carrageenan hyperalgesia in rats. Eur. J. Pharmacol. 406, 53–58.

 Garcia-Larrea L., Maarrawi J., Peyron R., Costes N., Mertens P., Magnin M., Laurent B.: On the relation between sensory deafferentation, pain and thalamic activity in Wallenberg’s syndrome: a PET-scan study before and after motor cortex stimulation. Eur. J. Pain 10, 677–

688 (2006)

 Garcia-Larrea, L., Peyron, R., Mertens, P., Gregoire, M.C., Lavene, F., LeBars, D., Convers, P., Mauguières, F., Sindou, M., Laurent, B., 1999. Electrical stimulation of motor cortex for pain control: a combined PET-scan and electrophysiological study. Pain 83, 259–273.

 Gass P., Herdegen T., Bravo R., Kiessling M.: Induction and suppression of immediate early genes in specific rat brain regions by the non-competitive N-methyl-D-aspartate receptor antagonist MK-801. Neuroscience 1993; 53:749–58

 Gaunitz C, Schuttler A, Gillen C, Allgaier C. Formalin-induced changes of NMDA receptor subunit expression in the spinal cord of the rat. Amino Acids 2002;23:177–82.

 Gear, R.W., Aley, K.O., Levine, J.D., 1999. Pain-induced analgesia mediated by mesolimbic reward circuits. J. Neurosci. 19, 7175–7181.

 Gebhart GF. Descending modulation of pain. Neurosi Biobehav Rev 2004;27:729–737.

 Geiger, J.D., Nagy, J.I., 1986. Distribution of adenosine deaminase activity in rat brain and spinal cord. J. Neurosci. 6, 2707–2714.

 Genzen, J.R., Van Cleve, W., McGehee, D.S., 2001. Dorsal root ganglion neurons express multiple nicotinic acetylcholine receptor subtypes. J. Neurophysiol. 86, 1773–1782.

 Geraghty D.P., Mazzone S.B.: Respiratory actions of vanilloid receptor agonists in the nucleus of the solitary tract: comparison of resiniferatoxin with non-pungent agents and anandamide. Br J Pharmacol 137 919-927 (2002)

 Gilbert, A.K., Franklin, K.B.J., 2001. GABAergic modulation of descending inhibitory systems from the rostral ventromedial medulla (RVM). Dose-response analysis of nociception and neurological deficits. Pain 90, 25–36.

 Giuffrida, A., Beltramo, M., Piomelli, D., 2001. Mechanisms of endocannabinoid inactivation: biochemistry and pharmacology. J. Pharmacol. Exp. Ther. 298, 7–14.

 Gobert, A., Rivet, J.M., Lejeune, F., Newman-Tancredi, A., Adhumeau-Auclair, A., Nicolas, J.P., Cistarelli, L., Melon, C., Millan, M.J., 2000. Serotonin2C receptors tonically suppress the activity of mesocortical dopaminergic and adrenergic, but not serotonergic, pathways: a combined dialysis and electrophysiological analysis in the rat. Synapse 36, 205–221.

 Goldberg M.P., Choi D.W.: Combined oxygen and glucose deprivation in cortical cell culture: Calcium-dependent and calcium-independent mechanisms of neuronal injury. J Neurosci 1993; 13:3510–24

 Goldman-Rakic PS: Cellular basis of working memory. Neuron 1995; 14: 477–85

(9)

IX

 Gracely R.H., Geisser M.E., Giesecke T., Grant M.A., Petzke F.,Williams D.A., Clauw D.J:

Pain catastrophizing and neural responses to pain among persons with fibromyalgia. Brain 127, 835-843 (2004)

 Grant, A.D., Cottrell G.S., Amadesi S., Trevisani M., Nicoletti P., Materazzi S., Altier C., Cenac N., Zamponi G.W., Bautista-Cruz F. et al.: Protease-activated receptor 2 sensitizes the transient receptor potential vanilloid 4 ion channel to cause mechanical hyperalgesia in mice.

J. Physiol. 578, 715–733(2007)

 Gruss M., Bushell T.J., Bright D.P., Lieb W.R., Mathie A., Franks N.P.: Two-poredomain K_ channels are a novel target for the anesthetic gases xenon, nitrous oxide, and

cyclopropane. Mol Pharmacol 2004; 65:443–52

 Gu Y, Huang LY. Gabapentin potentiates N-methyl-d-aspartate receptor mediated currents in rat GABAergic dorsal horn neurons. Neurosci Lett 2002;324:177–80.

 Guler A.D., Lee H., Iida T., Shimizu I., Tominaga M., Caterina M.:. Heat-evoked activation of the ion channel, TRPV4. J. Neurosci. 22, 6408–6414 (2002)

 Guo H, Huang LY. Alteration in the voltage dependence of NMDA receptor channels in rat dorsal horn neurones following peripheral inflammation. J Physiol 2001;537:115–23.

 Guo W, Zou S, Guan Y, et al. Tyrosine phosphorylation of the NR2B subunit of the NMDA receptor in the spinal cord during the development and maintenance of inflammatory hyperalgesia. J Neurosci 2002;22:6208–17.

 Gutstein, H.B., Mansour, A., Watson, S.J., Fields, H.L., 1998. Mu and kappa opioid receptors in periaqueductal gray and rostral ventromedial medulla. Neuroreport 9, 1777–

1781.

 Haberberger, R., Scholz, A., Kummer, W., Kress, M., 2000. M2-receptor subtype does not mediate muscarine-induced increases in [Ca2+]i in nociceptive neurons of rat dorsal root ganglia. J. Neurophysiol. 84,1934–1941.

 Hajos, N., Ledent, C., Freunds, T.F., 2001. Novel cannabinoid-sensitive receptor mediates inhibition of glutamatergic synaptic transmission in the hippocampus. Neuroscience 106, 1–

4.

 Hallbeck, M., Larhammar, D., Blomqvist, A., 2001. Neuropeptide expression in rat paraventricular hypothalamic neurons that project to the spinal cord. J. Comp. Neurol. 433, 222–238.

 Hama, A.T., Menzaghi, F., 2001. Antagonist of nicotinic acetylcholine receptors (nAChR) enhances formalin-induced nociception in rats: tonic role of nAChRs in the control of pain following injury. Brain Res. 888, 102–106.

 Hamza M., Dionne R.A.: Mechanisms of Non-Opioid Analgesics Beyond Cyclooxygenase Enzyme Inhibition. Curr Mol Wang Y., Qin Z.: Molecular and cellular mechanisms of excitotoxic neuronal death. Apoptosis, 2010.

 Hapfelmeier G., Zieglgansberger W., Haseneder R., Schneck H., Kochs E.: Nitrous oxide and xenon increase the efficacy of GABA at recombinant mammalian GABAA receptors.

Anesthesia Analgesia 2000; 91:1542–9

 Hardingham G.E., Bading H.: The Yin and Yang of NMDA receptor signalling. Trends Neurosci 2003; 26:81–9

 Harris J.A. Using c-fos as a neural marker of pain. Brain Res Bull 1998; 45:1–8

 Haseneder R., Kratzer S., Kochs E., Mattusch C., Eder M., Rammes G: Xenon attenuates excitatory synaptic transmission in the rodent prefrontal cortex and spinal cord dorsal horn.

Anesthesiology 2009; 111:1297–1307

 Hernando, F., Schoots, O., Lolait, S.J., Burbach, J.P.H., 2001. Immunohistochemical localization of the vasopressin V1b receptor in the rat brain pituitary gland: anatomical support for its involvement inthe central effects of vasopressin. Endocrinology 142, 1659–

1668.

 Hewitt DJ. The use of NMDA-receptor antagonists in the treatment of chronic pain. Clin J Pain 2000;16:S73–9.

 Hinman A., Chuang H.H., Bautista D.M., Julius, D.: TRP channel activation by reversible covalent modification. Proc. Natl. Acad. Sci USA 108, 19564–19568 (2006)

 Hirai H, Kirsch J, Laube B, et al. The glycine binding site of the N-methyl-d-aspartate receptor subunit NR1: identification of novel determinants of co-agonist potentiation in the extracellular M3–M4 loop region. Proc Natl Acad Sci U S A 1996;93: 6031–6.

(10)

X

 Hirota K, Lambert DG. Ketamine: its mechanism(s) of action and unusual clinical uses. Br J Anaesth 1996;77:441–4.

 Hökfelt, T., Arvidsson, U., Cullheim, S., Millhorn, D., Nicholas, A.P., Pieribone, V., Seroogy, K., Ulfhake, B., 2000. Multiple messengers in descending serotonin neurons:

localization and functional implications. J. Chem. Neuroanatomy 18, 75–86.

 Hökfelt, T., Arvidsson, U., Cullheim, S., Millhorn, D., Nicholas, A.P., Pieribone, V., Seroogy, K., Ulfhake, B., 2000. Multiple messengers in descending serotonin neurons:

localization and functional implications. J. Chem. Neuroanatomy 18, 75–86.

 Holden, J.E., Naleway, E., 2001. Microinjection of carbachol in the lateral hypothalamus produces opposing actions on nociception mediated by alpha1- and alpha2-adrenoceptors.

Brain Res. 911, 27–36.

 Holden, J.E., Schwartz, E.J., Proudfit, H.K., 1999. Microinjection of morphine in the A7 catecholamine cell group produces opposing effects on nociception that are mediated by alfa1- and alfa2-adrenoceptors. Neuroscience 91, 979–990.

 Holstege G, Kuypers HG. The anatomy of brain stem pathways to the spinal cord in cat: a labeled amino acid tracing study. Prog Brain Res 1982;57:145–175.

 Holzer P.:Capsaicin: cellular targets, mechanisms of action, and selectivity for thin sensory neurons. Pharmacol. Rev. 43, 143-201 (1991)

 Homi H.M., Yokoo N., Ma D., Warner D.S., Franks N.P., Maze M., Grocott H.P.: The neuroprotective effect of xenon administration during transient middle cerebral artery occlusion in mice. Anesthesiology 2003; 99:876–81

 Horn J-L, Janicki P.K., Franks J.J.: Nitrous oxide and xenon enhance phospholipid- N- methylation in rat brain synaptic plasma membranes. Life Sci 1995; 56:PL455–60

 Hough, L.B., 2001. Genomics meets histamine receptors: new subtypes, new receptors. Mol.

Pharmacol. 59, 415–419.

 Hsieh JC, Belfrage M, Stone-Elander S, et al. Central representation of chronic ongoing neuropathic pain studied by positron emission tomography. Pain 1995;63:225–36.

 Hu¨neke R., Ju¨ngling E., Skasa M., Rossaint R., Lu¨ckhoff A.: Effects of the anesthetic gases xenon, halothane, and isoflurane on calcium and potassium currents in human atrial cardiomyocytes. Anestesiology 2001; 95:999–1006

 Hurley R. W., Adams M. C. D.: Sex, gender and pain: an overview of a complex field.

Aneshesia Analgesia 107, 309-317 (2008)

 Hwang, J.-H., Hwang, K.-S., Kim, J.-U., Choi, I.-C., Park, P.-H., Han, S.-M., 2001. The interaction between intrathecal neostigmine and GABA receptor agonists in rats with nerve ligation injury. Anesth. Analg. 93, 1297–1303.

 Ishiguro Y., Kikuchi T., Etsuki H., Niimi Y., Goto T., Morita S., Irie T.: Does xenon anesthesia inhibit cholinesterase? An in vitro radiometric assessment. Anesthesiology 2003;

98:791–2

 Jackson DL, Graff CB, Richardson JD, Hargreaves KM. Glutamate participates in the peripheral modulation of thermal hyperalgesia in rats. Eur J Pharmacol 1995;284:321–5.

 Jevtovic-Todorovic V., 'I'odorovic S.M., Mennerick S., Powell S., Dikranian K., Benshoff N., Zorumski C.F., Olneyp: Nitrous oxide (laughing gas) is an NMDA antagonist, neuroprotectant and neurotoxin. Nat Med 1998; 4383-4

 Jevtovic-Todorovic V., Todorovic S.M., Mennerick S., Powell S., Dikranian K., Benshoff N., Zorumski C.F., Olney J.W.: Nitrous oxide (laughing gas) is a NMDA antagonist,

neuroprotectant and neurotoxin. Nature Med 1998; 4:460–3

 Ji R.R., Samad T.A., Jin S.X., Schmoll R., Woolf C.J.: p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 36, 57-68 (2002)

 Jiang, M., Behbehani, M.M., 2001. Physiological characteristics of the projection pathway from the medial preoptic to the nucleus raphemagnus of the rat and its modulation by the periaqueductal gray. Pain 94, 147–1369.

 Jo, Y.H., Stoeckel, M.E., Freund-Mercier, M.J., Schlichter, R., 1998. Oxytocin modulates glutamatergic synaptic transmission between cultured neonatal spinal cord dorsal horn neurons. J. Neurosci. 18, 2377–2386.

 Johanek, L.M., Heitmiller, D.R., Turner, M., Nader, N., Hodges, J., Simone, D.A., 2001.

Cannabinoids attenuate capsaicin-evoked hyperalgesia through spinal and peripheral mechanisms. Pain 93, 303–315

(11)

XI

 Johansen, J.P., Fields, H.L., Manning, B.H., 2001. The affective component of pain in rodents: direct evidence for a contribution of the anterior cingulate cortex. Proc. Natl. Acad.

Sci. U.S.A. 98, 8077–8082.

 Johns R.A.: Nitric oxide, cyclic guanosine monophosphate, and the anesthetic state.

Anesthesiology 1996; 58:4579

 Julius, D., Basbaum, A.I.: Molecular mechanisms of nociception. Nature 413, 203–210 (2001)

 Kandel E. R., Schwartz J. H., Jessel T. M.: Principi di neuroscienze. Casa editrice Ambrosiana (2003).

 Karlsson U, Sjodin J, Angeby Moller K, et al. Glutamate-induced currents reveal three functionally distinct NMDA receptor populations in rat dorsal horn: effects of peripheral nerve lesion and inflammation. Neuroscience 2002;112:861–8

 Kashiba, H., Fukui, H., Morikawa, Y., Senba, E., 1999. Gene expression

 Kashiba, H., Fukui, H., Senba, E., 2001. Histamine H1 receptor mRNA is expressed in capsaicin-insensitive sensory neurons with neuropeptide Y-immunoreactivity in guinea pigs.

Brain Res. 901, 85–93.

 Kehl F., Pagel P.S., Krolikowski J.G., Gu W., Toller W.G., Warltier D.C., Kersten J.R.:

Isoflurane does not produce a second window of preconditioning against myocardial infarction in vivo. Anesth Analg 2002; 95:1162–8

 Kendig J.J.: In vitro networks: subcortical mechanisms of anaesthetic action. British Journal Anaesthesia 89, 91– 101 2002

 Khan, I.M., Stanislaus, S., Zhang, L., Taylor, P., Yaksh, T.L., 2001. A-85380 and epibatidine each interact with disparate spinal nicotinic receptor subtypes to achieve analgesia and nociception. J. Pharmacol. Exp. Ther. 297, 230–239.

 Kim S.R., Lee D.Y., Chung E.S., Oh U.T., Kim S.U., Jin B.K.: Transient receptor potential vanilloid subtype 1 mediates cell death of mesencephalic dopaminergic neurons in vivo and in vitro. J Neurosc 25, 662-671 (2005)

 Kirsch G.E., Codina J., Birnbaumer L., Brown A.M.: Coupling of ATP-sensitive K_

channels to A1 receptors by G proteins in rat ventricular myocytes. Am J Physiol 1990;

259:H820–26

 Kiss, J.P., Vizi, E.S., 2001. Nitric oxide: a novel link between synaptic and nonsynaptic transmission. Trends Neurosci. 24, 211–215.

 Kitano H., Young J.M., Cheng J., Wang L., Hurn P.D., Murphy S.J.:Gender-specific response to isoflurane preconditioning in focal cerebral ischemia. J Cereb Blood Flow Metab 27:1377–1386 (2007).

 Kjorsvik, A., Tjolsen, A., Hole, K., 2001. Activation of spinal serotonin2A/2C receptors augments nociceptive responses in the rat. Brain Res. 910, 179–181.

 Kobayashi S., Kato T., Bito H., Sato S.: Effects of Xenon on catecolamine and hemodynamic response to surgical noxious stimulation in humans. Journal Clinical Anesthesia 2006; 18 (5):

343-8

 Kolhekar R, Gebhart GF. NMDA and quisqualate modulation of visceral nociception in the rat. Brain Res 1994;651:215–26.

 Kowaluk, E.A., Kohlhaas, K.L., Bannon, A., Gunther, K., Lynch, J.J., Jarvis, M.F., 1999.

Characterization of the effects of adenosine kinase inhibitors on acute thermal nociception in mice. Pharmacol. Biochem. Behav. 63, 83–91.

 Krasowski MD, Harrison NL: General anaesthetic actions on ligand-gated ion channels. Cell Mol Life Sci 1999; 55:1278–303

 Kuroda, R., Kawao, N., Yoshimura, H., Umeda, W., Takemura, M.,Shigenaga, Y., Kawabata, A., 2001. Secondary somatosensory cortex stimulation facilitates the

antinociceptive effect of the NO synthase inhibitor through suppression of spinal nociceptive neurons in the rat.Brain Res. 903, 110–116.

 LaBella F.S., Stein D., Queen G.: The site of general anesthesia and cytochrome P450 monooxygenases: Occupation of the enzyme heme pocket by xenon and nitrous oxide. Eur J Pharmacol 1999; 381:R1–3

 Lachmann B, Armbruster S, Schairer W, Landstra M, Trouwborst A, Van Daal GJ, Kusuma A, Erdmann W: Safety and efficacy of xenon in routine use as an inhalational anaesthetic.

Lancet 1990; 335:1413–5

(12)

XII

 Lambe EK, Olausson P, Horst NK, Taylor JR, Aghajanian GK: Hypocretin and nicotine excite the same thalamocortical synapses in prefrontal cortex: Correlation with improved attention in rat. J Neurosci 2005; 25:5225–9

 LaMotte R.H., Lundberg L.E: R., Torrbjörk H. E.:Pain, hyperalgesia, and activity in nociceptive C units in humans after intradermal injection of capsaicin. J. Physiol.

(Lond.) 448, 749-764 (1992)

 Lau HC, Passingham RE: Relative blindsight in normal observers and the neural correlate of visual consciousness. Proc Natl Acad Sci U S A 2006; 103: 18763–8

 Laube B, Hirai H, Sturgess M, et al. Molecular determinants of agonist discrimination by NMDA receptor subunits: analysis of the glutamate binding site on the NR2B subunit.

Neuron 1997; 18:493–503.

 Lawand NB, Willis WD, Westlund KN. Excitatory amino acid receptor involvement in peripheral nociceptive transmission in rats. Eur J Pharmacol 1997;324:169–77.

 Le Grand, B., Panissié, A., Perez, M., Pauwels, P.J., John, G.W., 2000. Zolmitriptan stimulates a Ca2+-dependent K+ current in C6 glioma cells stably expressing recombinant human 5-HT1B receptors. Eur. J. Pharmacol. 397, 297–302.

 Leem J. W., Lee B. H., Willis W. D., Chung J. M.: Grouping of somatosensory neurons in the spinal cord and the gracile nucleus of the rat by cluster analysis. J Neurophysiol 72 2590- 2597 (1994)

 Leem JW, Hwang JH, Hwang SJ, et al. The role of N-methyl-daspartate receptors in Freund’s complete adjuvant induced mechanical hyperalgesia in rats. Neurosci Lett 2001;297:155–8

 Leung A, Wallace MS, Ridgeway B, Yaksh T. Concentrationeffect relationship of intravenous alfentanil and ketamine on peripheral neurosensory thresholds, allodynia and hyperalgesia of neuropathic pain. Pain 2001;91:177–87.

 Levine, R.R., Birdsall, N.J.M., Nathanson, N.M., 1999. Subtypes of muscarinic receptors:

Proceedings of the Eighth International Symposium on Subtypes of Muscarinic Receptor.

Life Sci. 67, 355–596.

 Levine, R.R., Birdsall, N.J.M., Nathanson, N.M., 1999. Subtypes of muscarinic receptors:

Proceedings of the Eighth International Symposium on Subtypes of Muscarinic Receptor.

Life Sci. 67, 355–596.

 Liao GY, Wagner DA, Hsu MH, Leonard JP. Evidence for direct protein kinase-C mediated modulation of N-methyl-d-aspartate receptor current. Mol Pharmacol 2001;59:960–4.

 Liapi A., Wood J.N.: Extensive co-localization and heteromultimer formation of the vanilloid receptor-like protein TRPV2 and the capsaicin receptor TRPV1 in the adult ratcerebral cortex. Eur J Neurosci 22, 825-834 (2005)

 Liedtke W., Choe Y., Marti-Renom M.A., Bell A.M., Denis C.S., Sali A., Hudspeth, A.J., Friedman J.M., Heller S.: Vanilloid receptorrelated osmotically activated channel (VR- OAC), a candidate vertebrate osmoreceptor. Cell 103, 525–535 (2000)

 Liedtke W.,Friedman, J.M.: Abnormal osmotic regulation in trpv4−/− mice. Proc. Natl.

Acad. Sci. USA 100, 13698–13703(2003)

 Limatola V., Ward P., Cattano D., Gu J., Giunta F., Maze M., Ma D.: Xenon preconditioning confers neuroprotection regardless of gender in a mouse model of transient middle cerebral artery occlusion. Neuroscience 165 (2010) 874–881

 Liu H, Mantyh PW, Basbaum AI. NMDA-receptor regulation of substance P release from primary afferent nociceptors. Nature 1997;386:721–4.

 Logothetis, N.K., Pauls, J., Augath, M., Trinath, T., Oeltermann, A., 2001.

Neurophysiological investigation of the basis of the fMRI signal. Nature 412, 150–157.

 Loomis, C.W., Khandwala, H., Osmond, G., 2001. Co-administration of intrathecal

strychnine and bicuculline effects synergistic allodynia in the rat: an isobolographic analysis.

J. Pharmacol. Exp. Ther. 296, 756–771

 Lu WY, Xiong ZG, Lei S, et al. G-protein-coupled receptors act via protein kinase C and Src to regulate NMDA receptors. Nat Neurosci 1999;2:331–8.

 Luo Z.D., Chaplan S.R., Higuera E.S., Sorkin L.S., Stauderman K.A., Williams M.E., and Yaksh, T.L.:Upregulation of dorsal root ganglion (alpha)2(delta) calcium channel subunit and its correlation with allodynia in spinal nerve-injured rats. J. Neurosci. 21, 1868–1875 (2001)

(13)

XIII

 Lutz M.R.., Liu H.: Sevoflurane produces a delayed window of protection in young rat myocardium and fails to in aged rat myocardium (abstract). ANESTHESIOLOGY 2004; 101 (suppl):A-732

 Lynch M.: Pain as the fifth vital sign. J. Intraven Nurs. Mar-Apr; 24 (2): 85-94 (2001)

 Ma D., Hossain M., Chow A., Arshad M., Battson R.M., Sanders R.D., Mehmet H., Edwards D.D., Franks N.P., Maze M.: Xenon and hypothermia combine synergistically to provide neuroprotection from neonatal asphyxia. Ann Neurol 2005; 58:182–93

 Ma D., Hossain M., Pettet G.K.J., Luo Y., Lim T., Akimov S., Sanders R.D., Franks N.P., Maze M.: Xenon preconditioning reduces brain damage from neonatal asphyxia in rats. J Cereb Blood Flow Metab 2006; 26:199–208

 Ma D., Hossain M., Rajakumaraswamy N., Franks N.P., Maze M.: Combination of xenon and isoflurane produces a synergistic protective effect against oxygenglucose deprivation injury in a neuronal-glial co-culture model. Anesthesiology 2003; 99:748–51

 Ma D., Sanders R.D., Halder S., Rajakumaraswamy N. Frank N.P., Maze M.: Xenon exerts age-independent antinociception in Fischer rats. Anesthesiology 2004; 100100, 1313-8.

 Ma D., Wilhelm S., Maze M., Franks N.P.: Neuroprotective and neurotoxic properties of the inert gas xenon. British Journal Anaesthesia 2002; 89:739–46

 Ma D., Yang H., Lynch J., Franks N.P., Maze M., Grocott H.P.: Xenon attenuates cardiopulmonary bypass-induced neurologic and neurocognitive dysfunction in the rat.

Anesthesiology 2003; 98:690–8

 Ma QP, Hargreaves RJ. Localization of N-methyl-d-aspartate NR2B subunits on primary sensory neurons that give rise to small-caliber sciatic nerve fibers in rats. Neuroscience 2000;101: 699–707.

 Ma, H.C., Dohi, S., Wang, Y.F., Ishizawa, Y., Yanagidate, F., 2001. The antinociceptive and sedative effects of carbachol and oxycodone administered into brainstem pontine reticular formation and spinal subarachnoid space in rats. Anesth. Analg. 92, 1307–1315.

 Macpherson L.J., Dubin, A.E. Evans M.J., Marr F., Schultz P.G., Cravatt B.F., Patapoutian A.: Noxious compounds activate TRPA1 ion channels through covalent modification of cysteines. Nature 445, 541-545 (2007)

 Madrid R., de la Pena E., Donovan-Rodriguez T., Belmonte C., Viana F.: Variable threshold of trigeminal cold-thermosensitive neurons is determined by a balance between TRPM8 and Kv1 potassium channels. J. Neurosci. 29, 3120–3131 (2009)

 Maione S., Bisogno T., De Novellis V., Palazzo E., Cristino L., Valenti M., Petrosino S., Guglielmotti V., Rossi F., Di Marzo V.: Elevation of endocannabinoid levels in the ventrolateral periaqueductal grey through inhibition of fatty acid amide hydrolase affects descending nociceptive pathways via both cannabinoid receptor type 1 and transient receptor potential vanilloid type-1 receptors. J Pharmacol Exp Ther 316, 969-982 (2006)

 Malan, T.P., Ibrahim, M.M., Deng, H., Liu, Q., Mata, H.P., Vanderah, T., Porreca, F., Makriyannis, A., 2001. CB2 cannabinoid receptor-mediatedperipheral antinociception. Pain 93, 239–245.

 Malmberg AB, Gilbert H, McCabe RT, Basbaum AI. Powerful antinociceptive effects of the cone snail venom-derived subtypeselective NMDA receptor antagonists conantokins G and T. Pain 2003;101:109–16.

 Malmberg, A.B., Chen, C., Tonegawa, S., and Basbaum, A.I.: Preserved acute pain and reduced neuropathic pain in mice lacking PKCgamma. Science 278, 279–283 (1997)

 Marinelli S., Di Marzo V., Berretta N., Matias I., Maccarrone M., Bernardi G., Mercuri N.B.:

Presynaptic facilitation of glutamatergic synapses to dopaminergic neurons of the rat substantia nigra by endogenous stimulation of vanilloid receptors. J Neurosci 23, 3136-3144 (2003)

 Marvizon JC, McRoberts JA, Ennes HS, et al. Two N-methyl-daspartate receptors in rat dorsal root ganglia with different subunit composition and localization. J Comp Neurol 2002;446: 325–41.

 Mason P, Gao K, Genzen JR. Serotonergic raphe magnus cell discharge reflects ongoing autonomic and respiratory activities. J Neurophysiol 2007;98:1919–1927.

 Mason P. Deconstructing endogenous pain modulations. J Neurophysiol 2005;94:1659–1663.

 Mason, D.J., Lowe, J., Welch, S.P., 1999. Cannabinoid modulation of dynorphin A:

correlation to cannabinoid-induced antinociception. Eur.J. Pharmacol. 378, 237–248.

(14)

XIV

 Mason, P., 1999. Central mechanisms of pain modulation. Curr. Opin. Neurobiol. 9, 436–

441.

 Matta J.A., Cornett P.M., Miyares R.L., Abe K., Sahibzada N., Ahern G.P.: General anesthetics activate a nociceptive ion channel to enhance pain and inflammation. Proc. Natl.

Acad. Sci. USA 105, 8784-8789 (2008)

 May A: Neuroimaging: visualizing brain in pain. Neurol Sci 28, 101-107 (2007)

 Mayadevi M, Praseeda M, Kumar KS, Omkumar RV. Sequence determinants on the NR2A and NR2B subunits of NMDA receptor responsible for specificity of phosphorylation by CaMKII. Biochim Biophys Acta 2002;1598:40–5.

 Mayer E.A., Berman S., Suyenobu B., Labus J., Mandelkern M.A.,Naliboff B.D., Chang, L.:Differences in brain responses to visceral pain between patients with irritable bowel syndrome and ulcerative colitis. Pain 115, 398-409 (2005)

 McCleane G, Smith H. Opioids for persistent noncancer pain. Med Clin N Am 2007; 91:177- 197.

 McCormack K. Signal transduction in neuropathic pain, with special emphasis on the analgesic role of opioids. Part I: The basic science of phenotype expression in normal and regeneratingnerve. Pain Rev 1999;6:3–33.

 McGaraughty S., Chu K.L., Bitner R.S., Martino B., El Kouhen R., Han P., Nikkel A.L., Burgard E.C., Faltynek C.R., Jarvis M.F.: Capsaicin infused into the PAG affects rat tail flick responses to noxious heatand alters neuronal firing in the RVM. J Neurophysiol 2003, 90, 2702-2710 (2003)

 McRoberts JA, Coutinho SV, Marvizon JC, et al. Role of peripheral N-methyl-d-aspartate (NMDA) receptors in visceral nociception in rats. Gastroenterology 2001;120:1737–48.

 Meen M, Coudore-Civiale MA, Parry L, et al. Involvement of N-methyl-d-aspartate receptors in nociception in the cyclophosphamide-induced vesical pain model in the conscious rat. Eur J Pain 2002;6:307–14.

 Meldrum ML. A capsule history of pain management. JAMA 2003; 290:2470-2475.

 Melzack, R.: From the gate to the neuromatrix. Pain Suppl. 6, 121-126 (1999)

 Meng, I.D., Manning, B.H., Martin, W.J., Fields, H.L., 1998. An analgesia circuit activated by cannabinoids. Nature 395, 381–383.

 Merborth MK, Barnason S.: Managing pain: the fifth vital sign. Nurs Clin North Am Jun; 35 (2): 375-83 (2000)

 Merksey H., Bogduk N.: Classificazion of cronic pain. Seattle: IASP Press (1994)

 Messinger R.B., Naik A.K., Jagodic M.M., Nelson M.T., Lee W.Y., Choe W.J., Orestes P., Latham, J.R., Todorovic, S.M., and Jevtovic-Todorovic, V.: In vivo silencing of the Ca(V)3.2 T-type calcium channels in sensory

 Meyer, R.A., Ringkamp, M., Campbell, J.N., and Raja, S.N.: Peripheral mechanisms of cutaneous nociception. In Wall and Melzack’s Textbook of Pain, S.B. McMahon and M.

Koltzenburg, eds. (Philadelphia: Elsevier), pp. 3–34 (2008)

 Mezey E., Toth Z.E., Cortright D.N., Arzubi M.K., Krause J.E., Elde R., Guo A., Blumberg P.M., Szallasi A.: Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1- like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci U S A 97, 3655-3660 (2000)

 Miao, F.J.P., Benowitz, N.L., Levine, J.D., 2001. Endogenous opioids suppress activation of nociceptors by sub-nanomolar nicotine. Br. J. Pharmacol. 133, 23–28.

 Miki K, Zhou QQ, Guo W, et al. Changes in gene expression and neuronal phenotype in brain stem pain modulatory circuitry after inflammation. J Neurophysiol 2002;87:750–60.

 Millan M.J. Descending control of pain. Prog Neurobiol 2002;66:355–474.

 Millan M.J.Serotonin (5-HT) and pain: a reappraisal of its role in the light of receptor multiplicity. Semin.Neurosci. 1995 7, 409–419

 Millan, M.J., 1997. The role of descending noradrenergic and serotoninergic pathways in the modulation of nociception: focus on receptor multiplicity. In: Dickenson, A., Besson, J.M.

(Eds.), The

 Millan, M.J., 1999. The induction of pain: an integrative review. Prog. Neurobiol. 57, 1–164.

 Millan, M.J., Lejeune, F., Gobert, A., 2000. Reciprocal autoreceptor and hereroreceptor control of serotonergic, dopaminergic and noradrenergic transmission in the frontal cortex:

relevance to the actions of antidepressant agents. J. Psychopharmacol. 14, 114–136.

(15)

XV

 Miyazaki Y., Adachi T., Utsumi J., Shichino T., Segawa H.: Xenon has greater inhibitory effects on spinal dorsal horn neurons than nitrous oxide in spinal cord transected cats.

Anesthesia Analgesia 1999; 88:893–7

 Mobarakeh, J.I., Sakurada, S., Katsuyama, S., Kutsuwa, M., Kuramasu, A., Lin, Z.Y., Watanabe, T., Hashimoto, Y., Watanabe, T., Yanai, K., 2000. Role of histamine H1 receptor in pain perception: a study ofthe receptor gene knockout mice. Eur. J. Pharmacol. 391, 81–

89.

 Mochizuki T., Sokabe T., Araki I., Fujishita K., Shibasaki K., Uchida K., Naruse K., Koizumi S., Takeda M., Tominaga, M.: The TRPV4 cation channel mediates stretch-evoked Ca2+ influx and ATP release in

 model. Circ Res 1995; 77:1030–5

 Momiyama A. Distinct synaptic and extrasynaptic NMDA receptors identified in dorsal horn neurones of the adult rat spinal cord. J Physiol 2000;523 Pt 3:621–8.

 Montes C., Magnin M., Maarrawi J., Frot M., Convers P., Mauguiere F., Garcia-Larrea L.:

Thalamic thermo-algesic transmission: ventral posterior (VP) complex versus VMpo in the light of a thalamic infarct with central pain. Pain 113, 223–232 (2005)

 Moore KA, Baba H, Woolf CJ. Gabapentin–actions on adult superficial dorsal horn neurons.

Neuropharmacology 2002;43: 1077–81.

 Moore KA, Baba H, Woolf CJ: Synaptic transmission and plasticity in the superficial dorsal horn. Prog Brain Res 2000; 129:63–80

 Morales, M.A., McCollum, N., Kirkness, E.F., 2001b. 5-HT3-receptor subunits A and B are co-expressed in neurons of the dorsal root ganglion. J. Comp. Neurol. 438, 163–172.

 Mori.T, Zhao X., Zuo Y., Aistrup G.L., Nishikawa K., Marszalec W., Yeh J.Z., Narahashi T.:

Modulation of neuronal nicotinic acetylcholine receptors by halothane in rat cortical neurons.

Mol Pharmacol 2001; 59:732–43

 Mott DD, Doherty JJ, Zhang S, et al. Phenylethanolamines inhibit NMDA receptors by enhancing proton inhibition. Nat Neurosci 1998;1:659–67.

 Mott, D.D., Erreger, K., Banke, T.G., Traynelis, S.F., 2001. Open probability of homomeric murine 5-HT3A serotonin receptors depends on subunit occupancy. J. Physiol. 535, 427–443.

 Mu¨llenheim J., Ebel D., Bauer M., Otto F., Heinen A., Fra¨ßdorf J., Preckel B., Schlack W.:

Sevoflurane confers additional cardioprotection after ischemic late preconditioning in rabbits.

Anesthesiology 2003; 99:624–31

 Mu¨llenheim J., Ebel D., Fra¨ßdorf J., Preckel B., Tha¨mer V., Schlack W.: Isoflurane preconditions myocardium against infarction via release of free radicals. Anesthesiology 2002; 96:934–40

 Mukamel, R., Gelbhard, H., Arieli, A., Hasson, U., Fried, I., Malach, R., 2005. Coupling between neuronal firing, field potentials, and f-MRI in human auditory cortex. Science 309, 951–954.

 Muraki K., Iwata Y., Katanosaka Y., Ito T., Ohya S., Shigekawa M., Imaizumi Y.: TRPV2 is a component of osmotically sensitive cation channels in murine aortic myocytes. Circ. Res.

93, 829–838 (2003)

 Murphy, A.Z., Rizvi, T.A., Ennis, M., Shipley, M.T., 1999. Theorganization of preoptic- medullary circuits in the male rat: evidence forinterconnectivity of neural structures involved in reproductive behaviorantinociception and cardiovascular regulation. Neuroscience 91, 1103–1116.

 Musser J.B., Fontana J.L., Mongan P.D.: The anesthetic and physiologic effects of an intravenous administration of a halothane lipid emulsion (5% vol/vol). Anesthesia Analgesia 1999;88:671–5.

 N’guyen, J.P., Lefaucheur, J.P., Decq, P., Uchiyama, T., Carpentier, A., Fontaine, D., Brugières, P., Pollin, B., Fève, A., Rostaing, S., Cesaro, P., Keravel, Y., 1999. Chronic motor cortex stimulation in the treatment of central and neuropathic pain. Correlations between clinical, electrophysiological and anatomical data, electrophysiological and anatomical data.

Pain 82, 245–251.

 Nagata A., Nakao S., Nishizawa N., Masuzawa M., Inada T., Murao K., Miyamoto E., Shingu K.: Xenon inhibits but N2O enhances ketamine-induced c-Fos expression in the rat posterior cingulate and retrosplenial cortices. Anesthesia Analgesia 2001; 92:362–8

(16)

XVI

 Nagele P, Metz LB, Crowder CM: Nitrous oxide (N2O) requires the Nmethyl-D-aspartate receptor for its action in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2004; 101:8791–

6

 Nagele P, Metz LB, Crowder CM: Xenon acts by inhibition of non–Nmethyl-D-aspartate receptor mediated glutamatergic neurotransmission in Caenorhabditis elegans.

Anesthesiology 2005; 103:508–13

 Nagy I., Santha. P, Jancso G., Urban L.: The role of the vanilloid (capsaicin) receptor (TRPV1) in physiology and pathology. Eur J Pharmacol 500, 351-369 (2004)

 Nakata Y, Goto T, Saito H, Ishiguro Y, Terui K, Kawakami H, Tsuruta Y, Niimi Y, Morita S: Plasma concentration of fentanyl with xenon to block somatic and hemodynamic responses to surgical incision. Anesthesiology 2000; 92:1043–8

 Nakata Y., Goto T., Ishiguro Y., Terui K., Niimi Y., Morita S.: Anesthetic doses of sevoflurane to block cardiovascular responses to incision when administered with xenon or nitrous oxide. Anesthesiology 1999; 91:369-73

 Nakata Y., Goto T.., Saito H., Ishiguro Y, Terui K., Kawakami H., Tsuruta Y., Niimi Y., Morita S: Plasma concentration of Fentanyl with Xenon to block somatic and hemodynamic responses to surgical incision. Anesthesiology 2000, 92: 1043- 8

 Nassar M.A., Levato A., Stirling L.C., Wood J.N.: Neuropathic pain develops normally in mice lacking both Na(v)1.7 and Na(v)1.8. Mol. Pain 1, 24. (2005)

 Nassar M.A., Stirling L.C., Forlani G., Baker M.D., Matthews E.A., Dickenson A.H., and Wood, J.N.:.Nociceptor-specific gene deletion reveals a major role for Nav1.7 (PN1) in acute and inflammatory pain. Proc. Natl. Acad. Sci. USA 101, 12706–12711 (2004)

 Natale G., Cattano D., Abramo A., Forfori F., Fulceri F., Fornai F., Paparelli A., Giunta F.:

Morfological evidence that Xenon neuroprotects against N-Methyl-DL-Aspartic-induced damage in the rat arcuate nucleus. A time-dependent study. Ann. N. Y. Acad. Sci. 2006 1074, 650-658

 Nestler EJ. Molecular basis on longterm longterm plasticity underlying addiction. Nat Rev Neurosci 2001; 2:119-128.

 neurons alleviates hyperalgesia in rats with streptozocin-induced diabetic neuropathy. Pain 145, 184–195(2009)

 Nicoll R.A., Madison D.V.: General anesthetics hyperpolarize neurons in the vertebrate central nervous system. Science 1982; 217:1055–7

 Niessing, J., Ebisch, B., Schmidt, K.E., Niessing, M., Singer, W., Galuske, R.A., 2005.

Hemodynamic signals correlate tightly with synchronized gamma oscillations. Science 309, 48–951.

 Nishi M, Hinds H, Lu HP, et al. Motoneuron-specific expression of NR3B, a novel NMDA- type glutamate receptor subunit that works in a dominant-negative manner. J Neurosci 2001;21: RC185.

 Noble, F., Roques, B.P., 1999. CCK-B receptors: chemistry, molecular biology, biochemistry and pharmacology. Prog. Neurobiol. 56, 1–31.

 Noel J., Zimmermann K., Busserolles J., Deval E., Alloui A., Diochot S., Guy N., Borsotto M., Reeh P., Eschalier A.: The mechanoactivated K+ channels TRAAK and TREK-1 control both warm and coldperception. EMBO J. 28, 1308–1318 (2009)

 Nuseir, K., Proudfit, H.K., 2000. Bidirectional modulation of nociception by GABA neurons in the dorsolateral pontine tegmentum that tonically inhibit spinally projecting noradrenergic A7 neurons. Neuroscience 96,773–783.

 Odeh, F., Antal, M., 2001. The projections of the midbrain periaqueductal grey to the pons and medulla oblongata in rats. Eur. J. Neurosci. 14, 1275–1286.

 of GABAB1A, GABAB1B, and GABAB2 subunits in rat brain, spinal cord and dorsal root ganglion. Neuroscience 106, 447–467.

 of histamine H1 receptor in guinea pig primary sensory neurons: relationship between H1 receptor mRNA-expressing neurons andpeptidergic neurons. Mol. Brain Res. 66, 24–34.

 Ohara A., Mashimo T., Zhang P., Inagaki Y., Shibuta S., Yoshiya I.: A comparative study of the antinociceptive action of xenon and nitrous oxide in rats. Anesthesia Analgesia 1997;

85:931–6

 Okada, M., Nutt, D.J., Murakami, T., Zhu, G., Kamata, A., Kawata, Y., Kaneko, S., 2001.

Adenosine receptor subtypes modulate two major functional pathways to hippocampal serotonin release. J. Neurosci. 15, 628–640.

Riferimenti

Documenti correlati

Il Consiglio Superiore della Magistratura con delibera 17 luglio 1991 ha segnalato ai presidenti delle Corti d'Appello ed ai presidenti dei Tribunali l'opportunità di nominare ai

E’ una lampada di grande qualità, costruita con materiali nobili e inalterabili (alluminio pressofuso ed alluminio estruso), ed è verniciata con polveri epossidiche

La lampada è costruita da materiali riciclabili (alluminio, acciaio, tecnopolimero) The lamp is made of recyclable materials (aluminum, steel, technopolymer). Vassoio

Le attività di comunicazione saranno intraprese già nel mese di dicembre 2021, in preparazione alla Settimana delle Marche, e condotte contemporaneamente in Italia

Insert the little plate in the groove on the vertical profile, move it under the shelf then lock it in postion using an hex

Rivisitata di recente, la bilancia lineare è indispensabile per prodotti fragili ed è abbinabile a tutte le VFFS e macchine inclinate Pentavac.. Indicata per clienti che

"Come questo Consiglio ha affermato nella risoluzione del 1 dicembre 1994, adottata con il consenso di quasi tutti i suoi componenti e sotto la Presidenza e con il contributo

75% poliestere, 20% rayon, 5% elastane 330g Fodera Insulatex 120g Blu Metro, Grigio