Purkinje-neuron-specific down-regulation of p38 protects motoric function from the repeated use of benzodiazepine

Abstract

Benzodiazepine (BZD) is the most prescribed CNS depressant in America to treat hyper-excitatory disorders such as anxiety and insomnia. However, the chronic use of BZD often creates adverse effects including psychomotor deficit. In this study, we investigated a novel mechanism by which chronic BZD impedes motoric function in female mice. We used female mice because BZD use is much more prevalent in female than male populations. We tested the hypothesis that the accumulation of p38 (stress-activated protein) in cerebellar Purkinje neurons mediates motoric deficit induced by chronic BZD. To test this hypothesis, we generated transgenic mice that lack p38 incerebellar Purkinje neurons by crossing Pcp2 (Purkinje cell protein 2)-Cre mice with p38loxP/loxP mice. p38-knockdown mice and wild-type mice received BZD (lorazepam, 0.5 mg/kg) for 14 days. During this period, they were tested for motoric performance using Rotarod assay in which a quicker fall from rotating rod indicates poorer motoric performance. Cerebellum was then collected to detect p38 inPurkinje neurons and to measure mitochondrial respiration using immunohistochemistry and real-time XF respirometry, respectively. Compared to vehicletreated mice, BZD-treated mice showed poorer motoric performance, a higher number of Purkinje neurons containing p38, and lower mitochondrial respiration. These effects of BZD were much smaller in p38-knockdown mice. These results suggest that the excessive accumulation of p38 incerebellar Purkinje neurons contributes to motoric deficit associated with chronic BZD. They also suggest that Purkinje neuronal p38 mediates BZD-induced mitochondrial respiratory inhibition in cerebellum. Our findings may provide a new mechanistic insight into chronic BZD-induced motoric deficit.

Share and Cite:

Jung, M. and Metzger, D. (2013) Purkinje-neuron-specific down-regulation of p38 protects motoric function from the repeated use of benzodiazepine. Advances in Bioscience and Biotechnology, 4, 61-71. doi: 10.4236/abb.2013.46A009.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] Bogunovic, O.J. and Greenfield, S.F. (2004) Practical geriatrics: Use of benzodiazepines among elderly patients. Psychiatric Services, 55, 233-235. doi:10.1176/appi.ps.55.3.233
[2] Buffett-Jerrott, S.E. and Stewart, S.H. (2002) Cognitive and sedative effects of benzodiazepine use. Current Pharmaceutical Design, 8, 45-58. doi:10.2174/1381612023396654
[3] Leipzig, R.M., Cumming, R.G. and Tinetti, M.E. (1999) Drugs and falls in older people: A systematic review and meta-analysis: I. Psychotropic drugs. Journal of the American Geriatrics Society, 47, 30-39.
[4] Leipzig, R.M., Cumming, R.G. and Tinetti, M.E. (1999) Drugs and falls in older people: A systematic review and meta-analysis: II. Cardiac and analgesic drugs. Journal of the American Geriatrics Society, 47, 40-50.
[5] Wagner, A.K., Zhang, F., Soumerai, S.B., Walker, A.M., Gurwitz, J.H., Glynn, R.J. and Ross-Degnan, D. (2004) Benzodiazepine use and hip fractures in the elderly: Who is at greatest risk? Archives of Internal Medicine, 164, 1567-1572. doi:10.1001/archinte.164.14.1567
[6] Hemmelgarn, B., Suissa, S., Huang, A., Boivin, J.F. and Pinard, G. (1997) Benzodiazepine use and the risk of motor vehicle crash in the elderly. JAMA, 278, 27-31. doi:10.1001/jama.1997.03550010041037
[7] Cumming, R.G. and Le Couteur, D.G. (2003) Benzodiazepines and risk of hip fractures in older people: A review of the evidence. CNS Drugs, 17, 825-837. doi:10.2165/00023210-200317110-00004
[8] Dawson, J., Boyle, J., Stanley, N., Johnsen, S., Hindmarch, I. and Skene, D.J. (2008) Benzodiazepine-induced reduction in activity mirrors decrements in cognitive and psychomotor performance. Human Psychopharmacology, 23, 605-613. doi:10.1002/hup.961
[9] Gray, S.L., Penninx, B.W., Blough, D.K., Artz, M.B., Guralnik, J.M., Wallace, R.B., Buchner, D.M. and LaCroix, A.Z. (2003) Benzodiazepine use and physical performance in community-dwelling older women. Journal of the American Geriatrics Society, 51, 1563-1570. doi:10.1046/j.1532-5415.2003.51502.x
[10] Korpi, E.R., Koikkalainen, P., Vekovischeva, O.Y., Makela, R., Kleinz, R., Uusi-Oukari, M. and Wisden, W. (1999) Cerebellar granule-cell-specific GABAA recaptors attenuate benzodiazepine-induced ataxia: Evidence from alpha 6-subunit-deficient mice. European Journal of Neuroscience, 11, 233-240. doi:10.1046/j.1460-9568.1999.00421.x
[11] Stanley, J.L., Lincoln, R.J., Brown, T.A., McDonald, L.M., Dawson, G.R. and Reynolds, D.S. (2005) The mouse beam walking assay offers improved sensitivity over the mouse rotarod in determining motor coordination deficits induced by benzodiazepines. Journal of Psychopharmacology, 19, 221-227. doi:10.1177/0269881105051524
[12] Licata, S.C., Jensen, J.E., Penetar, D.M., Prescot, A.P., Lukas, S.E. and Renshaw, P.F. (2009) A therapeutic dose of zolpidem reduces thalamic GABA in healthy volunteers: A proton MRS study at 4 T. Psychopharmacology, 203, 819-829. doi:10.1007/s00213-008-1431-1
[13] Malcolm, R., Myrick, H., Roberts, J., Wang, W., Anton, R.F. and Ballenger, J.C. (2002) The effects of carbamazepine and lorazepam on single versus multiple previous alcohol withdrawals in an outpatient randomized trial. Journal of General Internal Medicine, 17, 349-355.
[14] Korpi, E.R., Mattila, M.J., Wisden, W. and Luddens, H. (1997) GABA(A)-receptor subtypes: Clinical efficacy and selectivity of benzodiazepine site ligands. Annals of Internal Medicine, 29, 275-282. doi:10.3109/07853899708999348
[15] Greenblatt, D.J., Harmatz, J.S., von Moltke, L.L., Wright, C.E. and Shader, R.I. (2004) Age and gender effects on the pharmacokinetics and pharmacodynamics of triazolam, a cytochrome P450 3A substrate. Clinical Pharmacology & Therapeutics, 76, 467-479. doi:10.1016/j.clpt.2004.07.009
[16] Rijcken, C.A., Knegtering, H., Bruggeman, R., Tobi, H. and de Jong-van den Berg, L.T. (2005) Sex differences in concomitant medication with benzodiazepines or antidepressants in first-break schizophrenic patients treated with antipsychotic medication. Psychiatry Research, 134, 143-150. doi:10.1016/j.psychres.2003.06.005
[17] Blazer, D., Hybels, C., Simonsick, E. and Hanlon, J.T. (2000) Sedative, hypnotic, and antianxiety medication use in an aging cohort over ten years: A racial compareson. Journal of the American Geriatrics Society, 48, 1073-1079.
[18] Gleason, P.P., Schulz, R., Smith, N.L., Newsom, J.T., Kroboth, P.D., Kroboth, F.J. and Psaty, B.M. (1998) Correlates and prevalence of benzodiazepine use in community-dwelling elderly. Journal of General Internal Medicine, 13, 243-250. doi:10.1046/j.1525-1497.1998.00074.x
[19] Hohmann, A.A. (1989) Gender bias in psychotropic drug prescribing in primary care. Medical Care, 27, 478-490. doi:10.1097/00005650-198905000-00004
[20] Mayer-Oakes, S.A., Kelman, G., Beers, M.H., De Jong, F., Matthias, R., Atchison, K.A., Lubben, J.E. and Schweitzer, S.O. (1993) Benzodiazepine use in older, community-dwelling southern Californians: Prevalence and clinical correlates. The Annals of Pharmacotherapy, 27, 416-421.
[21] Van der Waals, F.W., Mohrs, J. and Foets, M. (1993) Sex differences among recipients of benzodiazepines in Dutch general practice. BMJ, 307, 363-366. doi:10.1136/bmj.307.6900.363
[22] Verhaeghe, W., Mets, T. and Corne, L. (1996) Benzodiazepine use among elderly patients presenting at the emergency room. Archives of Gerontology and Geriatrics, 22, 55-62. doi:10.1016/0167-4943(95)00677-X
[23] Boyd, A.R., Shivshankar, P., Jiang, S., Berton, M.T. and Orihuela, C.J. (2012) Age-related defects in TLR2 signaling diminish the cytokine response by alveolar macrophages during murine pneumococcal pneumonia. Experimental Gerontology, 47, 507-518. doi:10.1016/j.exger.2012.04.004
[24] Horstmann, S., Kahle, P.J. and Borasio, G.D. (1998) Inhibitors of p38 mitogen-activated protein kinase promote neuronal survival in vitro. Journal of Neuroscience Research, 52, 483-490. doi:10.1002/(SICI)1097-4547(19980515)52:4<483::AID-JNR12>3.0.CO;2-4
[25] Skaper, S.D. and Walsh, F.S. (1998) Neurotrophic molecules: Strategies for designing effective therapeutic molecules in neurodegeneration. Molecular and Cellular Neuroscience, 12, 179-193. doi:10.1006/mcne.1998.0714
[26] Lee, J.C., Laydon, J.T., McDonnell, P.C., Gallagher, T.F., Kumar, S., Green, D., McNulty, D., Blumenthal, M.J., Heys, J.R., Landvatter, S.W., et al. (1994) A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature, 372, 739-746. doi:10.1038/372739a0
[27] Jiang, Y., Chen, C., Li, Z., Guo, W., Gegner, J.A., Lin, S. and Han, J. (1996) Characterization of the structure and function of a new mitogen-activated protein kinase (p38beta). The Journal of Biological Chemistry, 271, 17920-17926. doi:10.1074/jbc.271.30.17920
[28] Stein, B., Yang, M.X., Young, D.B., Janknecht, R., Hunter, T., Murray, B.W. and Barbosa, M.S. (1997) p38-2, a novel mitogen-activated protein kinase with distinct properties. The Journal of Biological Chemistry, 272, 19509-19517. doi:10.1074/jbc.272.31.19509
[29] Lechner, C., Zahalka, M.A., Giot, J.F., Moller, N.P. and Ullrich, A. (1996) ERK6, a mitogen-activated protein kinase involved in C2C12 myoblast differentiation. Proceedings of the National Academy of Sciences of the United States of America, 93, 4355-4359. doi:10.1073/pnas.93.9.4355
[30] Li, Z., Jiang, Y., Ulevitch, R.J. and Han, J. (1996) The primary structure of p38 gamma: A new member of p38 group of MAP kinases. Biochemical and Biophysical Research Communications, 228, 334-340. doi:10.1006/bbrc.1996.1662
[31] Jiang, Y., Gram, H., Zhao, M., New, L., Gu, J., Feng, L., Di Padova, F., Ulevitch, R.J. and Han, J. (1997) Characterization of the structure and function of the fourth member of p38 group mitogen-activated protein kinases, p38delta. The Journal of Biological Chemistry, 272, 30122-30128. doi:10.1074/jbc.272.48.30122
[32] Lee, S.H., Park, J., Che, Y., Han, P.L. and Lee, J.K. (2000) Constitutive activity and differential localization of p38alpha and p38beta MAPKs in adult mouse brain. Journal of Neuroscience Research, 60, 623-631. doi:10.1002/(SICI)1097-4547(20000601)60:5<623::AID-JNR7>3.0.CO;2-4
[33] Nonaka, Y., Miyajima, M., Ogino, I., Nakajima, M. and Arai, H. (2008) Analysis of neuronal cell death in the cerebral cortex of H-Tx rats with compensated hydrocephalus. Journal of Neurosurgery: Pediatrics, 1, 68-74. doi:10.3171/PED-08/01/068
[34] Xiong, W., Kojic, L.Z., Zhang, L., Prasad, S.S., Douglas, R., Wang, Y. and Cynader, M.S. (2006) Anisomycin activates p38 MAP kinase to induce LTD in mouse primary visual cortex. Brain Research, 1085, 68-76. doi:10.1016/j.brainres.2006.02.015
[35] Moriguchi, T., Toyoshima, F., Gotoh, Y., Iwamatsu, A., Irie, K., Mori, E., Kuroyanagi, N., Hagiwara, M., Matsumoto, K. and Nishida, E. (1996) Purification and identification of a major activator for p38 from osmotically shocked cells. Activation of mitogen-activated protein kinase kinase 6 by osmotic shock, tumor necrosis factor-alpha, and H2O2. The Journal of Biological Chemistry, 271, 26981-26988. doi:10.1074/jbc.271.43.26981
[36] Aydin, M.V., Sen, O., Kayaselcuk, F., Bolat, F., Tufan, K., Caner, H. and Altinors, N. (2005) Analysis and prevalence of inflammatory cells in subtypes of lumbar disc herniations under cyclooxygenase-2 inhibitor therapy. Journal of Neurology Research, 27, 609-612. doi:10.1179/016164105X49210
[37] Hensley, K., Floyd, R.A., Zheng, N.Y., Nael, R., Robinson, K.A., Nguyen, X., Pye, Q.N., Stewart, C.A., Geddes, J., Markesbery, W.R., Patel, E., Johnson, G.V. and Bing, G. (1999) p38 kinase is activated in the Alzheimer’s disease brain. Journal of Neurochemistry, 72, 2053-2058. doi:10.1046/j.1471-4159.1999.0722053.x
[38] Vereker, E., O’Donnell, E. and Lynch, M.A. (2000) The inhibitory effect of interleukin-1beta on long-term potentiation is coupled with increased activity of stress-activated protein kinases. The Journal of Neuroscience, 20, 6811-6819.
[39] Jung, M.E., Ju, X., Metzger, D.B. and Simpkins, J.W. (2011) Ethanol withdrawal hastens the aging of cytochrome c oxidase. Neurobiology Aging, 33, 618.e21-32.
[40] Kulisz, A., Chen, N., Chandel, N.S., Shao, Z. and Schumacker, P.T. (2002) Mitochondrial ROS initiate phosphorylation of p38 MAP kinase during hypoxia in cardiomyocytes. American Journal of Physiology. Lung Cellular and Molecular Physiology, 282, L1324-L1329.
[41] Kumar, C.N., Andrade, C. and Murthy, P. (2009) A randomized, double-blind comparison of lorazepam and chlordiazepoxide in patients with uncomplicated alcohol withdrawal. Journal of Studies on Alcohol and Drugs, 70, 467-474.
[42] Greenblatt, D.J., Von Moltke, L.L., Ehrenberg, B.L., Harmatz, J.S., Corbett, K.E., Wallace, D.W. and Shader, R.I. (2000) Kinetics and dynamics of lorazepam during and after continuous intravenous infusion. Critical Care Medicine, 28, 2750-2757. doi:10.1097/00003246-200008000-00011
[43] Papini, O., da Cunha, S.P., da Silva Mathes Ado, C., Bertucci, C., Moises, E.C., de Barros Duarte, L., de Carvalho Cavalli, R. and Lanchote, V.L. (2006) Kinetic disposition of lorazepam with focus on the glucuronidation capacity, transplacental transfer in parturients and racemization in biological samples. Journal of Pharmaceutical and Biomedical Analysis, 40, 397-403. doi:10.1016/j.jpba.2005.07.021
[44] Bourin, M., Hascoet, M., Mansouri, B., Colombel, M.C. and Bradwejn, J. (1992) Comparison of behavioral effects after single and repeated administrations of four benzodiazepines in three mice behavioral models. Journal of Psychiatry and Neuroscience, 17, 72-77.
[45] Naassila, M., Legrand, E., d’Alche-Biree, F. and Daoust, M. (1998) Cyamemazine decreases ethanol intake in rats and convulsions during ethanol withdrawal syndrome in mice. Psychopharmacology, 140, 421-428. doi:10.1007/s002130050785
[46] Wieland, S., Belluzzi, J., Hawkinson, J.E., Hogenkamp, D., Upasani, R., Stein, L., Wood, P.L., Gee, K.W. and Lan, N.C. (1997) Anxiolytic and anticonvulsant activity of a synthetic neuroactive steroid Co 3-0593. Psychopharmacology, 134, 46-54. doi:10.1007/s002130050424
[47] Rewal, M., Jung, M.E. and Simpkins, J.W. (2004) Role of the GABA-A system in estrogen-induced protection against brain lipid peroxidation in ethanol-withdrawn rats. Alcoholism: Clinical and Experimental Research, 28, 1907-1915. doi:10.1097/01.ALC.0000148100.78628.E7
[48] Guo, G. and Bhat, N.R. (2007) p38alpha MAP kinase mediates hypoxia-induced motor neuron cell death: A potential target of minocycline’s neuroprotective action. Neurochemical Research, 32, 2160-2166. doi:10.1007/s11064-007-9408-8
[49] Sudo, T., Yagasaki, Y., Hama, H., Watanabe, N. and Osada, H. (2002) Exip, a new alternative splicing variant of p38 alpha, can induce an earlier onset of apoptosis in HeLa cells. Biochemical and Biophysical Research Communications, 291, 838-843. doi:10.1006/bbrc.2002.6529
[50] Guan, J., Luo, Y. and Denker, B.M. (2005) Purkinje cell protein-2 (Pcp2) stimulates differentiation in PC12 cells by Gbetagamma-mediated activation of Ras and p38 MAPK. Biochemical Journal, 392, 389-397. doi:10.1042/BJ20042102
[51] Ju, X., Wen, Y., Metzger, D. and Jung, M. (2013) The role of p38 in mitochondrial respiration in male and female mice. Neuroscience Letters, 544, 152-156. http://authors.elsevier.com/sd/article/S0304394013003534
[52] Bruchas, M.R., Schindler, A.G., Shankar, H., Messinger, D.I., Miyatake, M., Land, B.B., Lemos, J.C., Hagan, C.E., Neumaier, J.F., Quintana, A., Palmiter, R.D. and Chavkin, C. (2011) Selective p38alpha MAPK deletion in serotonergic neurons produces stress resilience in models of depression and addiction. Neuron, 71, 498-511. doi:10.1016/j.neuron.2011.06.011
[53] Park, G.B., Kim, Y.S., Lee, H.K., Song, H., Kim, S., Cho, D.H. and Hur, D.Y. (2011) Reactive oxygen species and p38 MAPK regulate Bax translocation and calcium redistribution in salubrinal-induced apoptosis of EBV-transformed B cells. Cancer Letters, 313, 235-248. doi:10.1016/j.canlet.2011.09.011
[54] Mausset, A.L., De Seze, R., Montpeyroux, F. and Privat, A. (2001) Effects of radiofrequency exposure on the GABAergic system in the rat cerebellum: Clues from semi-quantitative immunohistochemistry. Brain Research, 912, 33-46. doi:10.1016/S0006-8993(01)02599-9
[55] Jung, M.E., Yang, S.H., Brun-Zinkernagel, A.M. and Simpkins, J.W. (2002) Estradiol protects against cerebellar damage and motor deficit in ethanol-withdrawn rats. Alcohol, 26, 83-93. doi:10.1016/S0741-8329(01)00199-9
[56] Watanabe, M. (2008) Molecular mechanisms governing competitive synaptic wiring in cerebellar Purkinje cells. The Tohoku Journal of Experimental Medicine, 214, 175190. doi:10.1620/tjem.214.175
[57] Sankar, R. (2011) GABA(A) receptor physiology and its relationship to the mechanism of action of the 1,5-benzodiazepine clobazam. CNS Drugs, 26, 229-244. doi:10.2165/11599020-000000000-00000
[58] Verster, J.C., Volkerts, E.R. and Verbaten, M.N. (2002) Effects of alprazolam on driving ability, memory functioning and psychomotor performance: A randomized, placebo-controlled study. Neuropsychopharmacology, 27, 260-269. doi:10.1016/S0893-133X(02)00310-X
[59] Ashton, H. (1994) The treatment of benzodiazepine dependence. Addiction, 89, 1535-1541. doi:10.1111/j.1360-0443.1994.tb03755.x
[60] Savic, M.M., Huang, S., Furtmuller, R., Clayton, T., Huck, S., Obradovic, D.I., Ugresic, N.D., Sieghart, W., Bokonjic, D.R. and Cook, J.M. (2008) Are GABAA receptors containing alpha5 subunits contributing to the sedative properties of benzodiazepine site agonists? Neuropsychopharmacology, 33, 332-339. doi:10.1038/sj.npp.1301403
[61] Lalonde, R. and Strazielle, C. (2003) Motor coordination, exploration, and spatial learning in a natural mouse mutation (nervous) with Purkinje cell degeneration. Behavior Genetics, 33, 59-66. doi:10.1023/A:1021003600900
[62] Rewal, M., Jung, M., Wen, Y., Brun-Zinkernagel, A. and Simpkins, J. (2003) Role of the GABAA system in behavioral, motoric, and cerebellar protection by estrogen during ethanol withdrawal. Alcohol, 31, 49-61. doi:10.1016/j.alcohol.2003.07.005
[63] Rakotomamonjy, J., Levenes, C., Baulieu, E.E., Schumacher, M. and Ghoumari, A.M. (2011) Novel protective effect of mifepristone on detrimental GABAA receptor activity to immature Purkinje neurons. The FASEB Journal, 25, 3999-4010. doi:10.1096/fj.11-183384
[64] Garrard, P., Martin, N.H., Giunti, P. and Cipolotti, L. (2008) Cognitive and social cognitive functioning in spinocerebellar ataxia: A preliminary characterization. Journal of Neurology, 255, 398-405. doi:10.1007/s00415-008-0680-6
[65] Lalonde, R. and Strazielle, C. (2003) The effects of cerebellar damage on maze learning in animals. Cerebellum, 2, 300-309. doi:10.1080/14734220310017456
[66] Madamanchi, N.R. and Runge, M.S. (2007) Mitochondrial dysfunction in atherosclerosis. Circulation Research, 100, 460-473. doi:10.1161/01.RES.0000258450.44413.96
[67] Sarnowska, A., Beresewicz, M., Zablocka, B. and Domanska-Janik, K. (2009) Diazepam neuroprotection in excitotoxic and oxidative stress involves a mitochondrial mechanism additional to the GABAAR and hypothermic effects. Neurochemistry International, 55, 164-173. doi:10.1016/j.neuint.2009.01.024
[68] Davis, L.F., Gatz, E.E. and Jones, J.R. (1971) Effects of chlordiazepoxide and diazepam on respiration and oxidative phosphorylation in rat brain mitochondria. Biochemical Pharmacology, 20, 1883-1887. doi:10.1016/0006-2952(71)90387-X
[69] Roald, O.K., Steen, P.A., Milde, J.H. and Michenfelder, J.D. (1986) Reversal of the cerebral effects of diazepam in the dog by the benzodiazepine antagonist Ro15-1788. Acta Anaesthesiologica Scandinavica, 30, 341-345. doi:10.1111/j.1399-6576.1986.tb02427.x
[70] Hirsch, J.D., Beyer, C.F., Malkowitz, L., Beer, B. and Blume, A.J. (1989) Mitochondrial benzodiazepine receptors mediate inhibition of mitochondrial respiratory control. Molecular Pharmacology, 35, 157-163.
[71] Chelli, B., Falleni, A., Salvetti, F., Gremigni, V., Lucacchini, A. and Martini, C. (2001) Peripheral-type benzodiazepine receptor ligands: Mitochondrial permeability transition induction in rat cardiac tissue. Biochemical Pharmacology, 61, 695-705. doi:10.1016/S0006-2952(00)00588-8
[72] Fan, M., Rhee, J., St-Pierre, J., Handschin, C., Puigserver, P., Lin, J., Jaeger, S., Erdjument-Bromage, H., Tempst, P. and Spiegelman, B.M. (2004) Suppression of mitochondrial respiration through recruitment of p160 myb binding protein to PGC-1alpha: Modulation by p38 MAPK. Genes & Development, 18, 278-289. doi:10.1101/gad.1152204
[73] Appaix, F., Guerrero, K., Rampal, D., Izikki, M., Kaambre, T., Sikk, P., Brdiczka, D., Riva-Lavieille, C., Olivares, J., Longuet, M., Antonsson, B. and Saks, V.A. (2002) Bax and heart mitochondria: Uncoupling and inhibition of respiration without permeability transition. Biochimica et Biophysica Acta, 1556, 155-167. doi:10.1016/S0005-2728(02)00358-4
[74] Simpkins, J.W., Wang, J., Wang, X., Perez, E., Prokai, L. and Dykens, J.A. (2005) Mitochondria play a central role in estrogen-induced neuroprotection. Current Drug Targets-CNS & Neurological Disorders, 4, 69-83. doi:10.2174/1568007053005073
[75] Blouin, R.T., Conard, P.F., Perreault, S. and Gross, J.B. (1993) The effect of flumazenil on midazolam-induced depression of the ventilatory response to hypoxia during isohypercarbia. Anesthesiology, 78, 635-641. doi:10.1097/00000542-199304000-00004
[76] Megarbane, B., Lesguillons, N., Galliot-Guilley, M., Borron, S.W., Trout, H., Decleves, X., Risede, P., Monier, C., Boschi, G. and Baud, F.J. (2005) Cerebral and plasma kinetics of a high dose of midazolam and correlations with its respiratory effects in rats. Toxicology Letters, 159, 22-31. doi:10.1016/j.toxlet.2005.04.003

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.