Nitrous oxide as a putative novel dual-mechanism treatment for bipolar depression: Proof-of-concept study design and methodology

Mikaela K Dimick, Danielle Omrin, Bradley J MacIntosh, Rachel H B Mitchell, Daniel Riegert, Anthony Levitt, Ayal Schaffer, Susan Belo, John Iazzetta, Garfield Detzler, Mabel Choi, Stephen Choi, Beverley A Orser, Benjamin I Goldstein, Mikaela K Dimick, Danielle Omrin, Bradley J MacIntosh, Rachel H B Mitchell, Daniel Riegert, Anthony Levitt, Ayal Schaffer, Susan Belo, John Iazzetta, Garfield Detzler, Mabel Choi, Stephen Choi, Beverley A Orser, Benjamin I Goldstein

Abstract

Introduction: Depressive symptoms predominate in the course of bipolar disorder (BD) and there is an urgent need to evaluate novel application of repurposed compounds that act on pre-specified treatment targets. Several lines of reasoning suggest that nitrous oxide (N2O) is an ideal medication to study as a potential treatment and as a strategy to identify the underlying pathophysiology of bipolar depression. N2O is a potent cerebral vasodilator and there is compelling evidence of reduced frontal cerebral blood flow (CBF; i.e. hypoperfusion) in depression. Therefore, N2O may increase CBF and thereby improve symptoms of depression. The goal of this randomized, double-blind trial is to study the effect of a single administration of N2O versus the active comparator midazolam on mood and CBF in adults with treatment-resistant bipolar depression.

Methods: Participants with BD-I/-II currently experiencing a major depressive episode will be randomized to one of two conditions (n = 20/group): 1) inhaled N2O plus intravenous saline, or 2) inhaled room air plus intravenous midazolam. Montgomery-Asberg Depression Rating Scale scores will serve as the primary endpoint. CBF will be measured via arterial spin labelling magnetic resonance imaging.

Conclusions: N2O is a potential novel treatment for bipolar depression, as it causes cerebral vasodilation. This proof-of-concept study will provide valuable information regarding the acute impact of N2O on mood and on CBF. If N2O proves to be efficacious in future larger-scale trials, its ubiquity, safety, low cost, and ease of use suggest that it has great potential to become a game-changing acute treatment for bipolar depression.

Keywords: Bipolar depression; Cerebral blood flow; Nitrous oxide; Novel therapeutics.

© 2020 Published by Elsevier Inc.

References

    1. Baldessarini R.J., Vieta E., Calabrese J.R. Bipolar depression: overview and commentary. Harv. Rev. Psychiatr. 2010;18:143–157.
    1. Sienaert P., Lambrichts L., Dols A. Evidence-based treatment strategies for treatment-resistant bipolar depression: a systematic review. Bipolar Disord. 2013;15:61–69.
    1. Frye M.A. Clinical practice. Bipolar disorder--a focus on depression. N. Engl. J. Med. 2011;364:51–59.
    1. Post R.M. The impact of bipolar depression. J. Clin. Psychiatr. 2005;66(Suppl 5):5–10.
    1. Judd L.L., Schettler P.J., Akiskal H.S. Long-term symptomatic status of bipolar I vs. bipolar II disorders. Int. J. Neuropsychopharmacol. 2003;6:127–137.
    1. Yatham L.N., Kennedy S.H., Parikh S.V. Canadian Network for Mood and Anxiety Treatments (CANMAT) and International Society for Bipolar Disorders (ISBD) collaborative update of CANMAT guidelines for the management of patients with bipolar disorder: update 2013. Bipolar Disord. 2013;15:1–44.
    1. Cipriani A., Barbui C., Salanti G. Comparative efficacy and acceptability of antimanic drugs in acute mania: a multiple-treatments meta-analysis. Lancet (London, England) 2011;378:1306–1315.
    1. Sidor M.M., Macqueen G.M. Antidepressants for the acute treatment of bipolar depression: a systematic review and meta-analysis. J. Clin. Psychiatr. 2011;72:156–167. doi: 10.4088/JCP.09r05385gre. 2010/11/03.
    1. Trivedi M.H., Fava M., Wisniewski S.R. Medication augmentation after the failure of SSRIs for depression. N. Engl. J. Med. 2006;354:1243–1252. doi: 10.1056/NEJMoa052964.
    1. Sinyor M., Schaffer A., Levitt A. The sequenced treatment alternatives to relieve depression (STAR*D) trial: a review. Canadian J. Psyc.Revue Canadienne de Psychiatrie. 2010;55:126–135.
    1. Insel T.R. The NIMH experimental medicine initiative. World Psychiatr. : Off. J. World Psyc. Assoc. 2015;14:151–153. doi: 10.1002/wps.20227.
    1. Machado-Vieira R., Henter I.D., Zarate C.A., Jr. New targets for rapid antidepressant action. Prog. Neurobiol. 2017;152:21–37. doi: 10.1016/j.pneurobio.2015.12.001. 2016/01/03.
    1. Gyulai F.E., Firestone L.L., Mintun M.A. In vivo imaging of human limbic responses to nitrous oxide inhalation. Anesth. Analg. 1996;83:291–298.
    1. Dashdorj N., Corrie K., Napolitano A. Effects of subanesthetic dose of nitrous oxide on cerebral blood flow and metabolism: a multimodal magnetic resonance imaging study in healthy volunteers. Anesthesiology. 2013;118:577–586.
    1. Emmanouil D.E., Quock R.M. Advances in understanding the actions of nitrous oxide. Anesth. Prog. 2007;54:9–18.
    1. Montgomery S.A., Asberg M. A new depression scale designed to be sensitive to change. Br. J. Psychiatry. 1979;134:382–389. doi: 10.1192/bjp.134.4.382. 1979/04/01.
    1. Amrein R., Hetzel W., Bonetti E.P. Clinical pharmacology of dormicum (midazolam) and anexate (flumazenil) Resuscitation. 1988;16(Suppl) S5-27. 1988/01/01.
    1. Wilkinson S.T., Farmer C., Ballard E.D. Impact of midazolam vs. saline on effect size estimates in controlled trials of ketamine as a rapid-acting antidepressant. Neuropsychopharmacology. 2019;44:1233–1238. doi: 10.1038/s41386-019-0317-8. 2019/01/18.
    1. Nordt S.P., Clark R.F. Midazolam: a review of therapeutic uses and toxicity. J. Emerg. Med. 1997;15:357–365. 1997/05/01.
    1. Sinyor M., Williams M., Belo S. Ketamine augmentation for major depressive disorder and suicidal ideation: preliminary experience in an inpatient psychiatry setting. J. Affect. Disord. 2018;241:103–109. doi: 10.1016/j.jad.2018.07.073. 2018/08/15.
    1. Fava M., Freeman M.P., Flynn M. Double-blind, placebo-controlled, dose-ranging trial of intravenous ketamine as adjunctive therapy in treatment-resistant depression (TRD) Mol. Psychiatr. 2018 doi: 10.1038/s41380-018-0256-5. 2018/10/05.
    1. First M.B., Spitzer R.L., Gibbon M. American Psychiatric Pub; 1997. User's Guide for the Structured Clinical Interview for DSM-IV axis I Disorders SCID-I: Clinician Version.
    1. Young R.C., Biggs J.T., Ziegler V.E. A rating scale for mania: reliability, validity and sensitivity. Br. J. Psychiatry. 1978;133:429–435. doi: 10.1192/bjp.133.5.429. 1978/11/01.
    1. Flippo T.S., Holder W.D., Jr. Neurologic degeneration associated with nitrous oxide anesthesia in patients with vitamin B12 deficiency. Arch. Surg. 1993;128:1391–1395. 1993/12/01.
    1. BOC. Medical Gas Data Sheet (MGDS) Medical Nitrous Oxide.
    1. Hamilton M. A rating scale for depression. J. Neurol. Neurosurg. Psychiatr. 1960;23:56.
    1. Beck A.T., Ward C.H., Mendelson M. An inventory for measuring depression. Arch. Gen. Psychiatr. 1961;4:561–571.
    1. Overall J.E., Gorham D.R., Shawver J.R. Basic dimensions of change in the symptomatology of chronic schizophrenics. J. Abnorm. Soc. Psychol. 1961;63:597–602. 1961/11/01.
    1. Bremner J.D., Krystal J.H., Putnam F.W. Measurement of dissociative states with the clinician-administered dissociative states scale (CADSS) J. Trauma Stress. 1998;11:125–136. doi: 10.1023/a:1024465317902. 1998/02/28.
    1. Hamilton M. The assessment of anxiety states by rating. Br. J. Med. Psychol. 1959;32:50–55. 1959/01/01.
    1. Aitken R.C. Measurement of feelings using visual analogue scales. Proc. Roy. Soc. Med. 1969;62:989–993.
    1. Wisniewski S.R., Rush A.J., Balasubramani G.K. Self-rated global measure of the frequency, intensity, and burden of side effects. J. Psychiatr. Pract. 2006;12:71–79. 2006/05/27.
    1. Nagele P., Duma A., Kopec M. Nitrous oxide for treatment-resistant major depression: a proof-of-concept trial. Biol. Psychiatr. 2015;78:10–18. doi: 10.1016/j.biopsych.2014.11.016.
    1. Kuhlmann L., Foster B.L., Liley D.T. Modulation of functional EEG networks by the NMDA antagonist nitrous oxide. PloS One. 2013;8 doi: 10.1371/journal.pone.0056434. 2013/03/05.
    1. Murrough J.W., Iosifescu D.V., Chang L.C. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am. J. Psychiatr. 2013;170:1134–1142. doi: 10.1176/appi.ajp.2013.13030392. 2013/08/29.
    1. Hu Y.D., Xiang Y.T., Fang J.X. Single i.v. ketamine augmentation of newly initiated escitalopram for major depression: results from a randomized, placebo-controlled 4-week study. Psychol. Med. 2016;46:623–635. doi: 10.1017/s0033291715002159. 2015/10/20.
    1. Grunebaum M.F., Ellis S.P., Keilp J.G. Ketamine versus midazolam in bipolar depression with suicidal thoughts: a pilot midazolam-controlled randomized clinical trial. Bipolar Disord. 2017;19:176–183. doi: 10.1111/bdi.12487. 2017/04/30.
    1. Murrough J.W., Iosifescu D.V., Chang L.C. Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am. J. Psychiatr. 2013:1–9. AiA.
    1. Murrough J.W., Soleimani L., DeWilde K.E. Ketamine for rapid reduction of suicidal ideation: a randomized controlled trial. Psychol. Med. 2015;45:3571–3580. doi: 10.1017/s0033291715001506. 2015/08/13.
    1. Layzer R.B. Myeloneuropathy after prolonged exposure to nitrous oxide. Lancet (London, England) 1978;2:1227–1230. doi: 10.1016/s0140-6736(78)92101-3. 1978/12/09.
    1. Blanco G., Peters H.A. Myeloneuropathy and macrocytosis associated with nitrous oxide abuse. Arch. Neurol. 1983;40:416–418. 1983/07/01.
    1. Zacny J.P., Camarillo V.M., Sadeghi P. Effects of ethanol and nitrous oxide, alone and in combination, on mood, psychomotor performance and pain reports in healthy volunteers. Drug Alcohol Depend. 1998;52:115–123.
    1. Becker D.E., Rosenberg M. Nitrous oxide and the inhalation anesthetics. Anesth. Prog. 2008;55:124–132. doi: 10.2344/0003-3006-55.4.124.
    1. Vgontzas A.N., Kales A., Bixler E.O. Benzodiazepine side effects: role of pharmacokinetics and pharmacodynamics. Pharmacology. 1995;51:205–223. doi: 10.1159/000139363. 1995/10/01.
    1. Permoda-Osip A., Dorszewska J., Bartkowska-Sniatkowska A. Vitamin B12 level may be related to the efficacy of single ketamine infusion in bipolar depression. Pharmacopsychiatry. 2013 Sep;46(6):227–228.
    1. Caton P.W., Tousman S.A., Quock R.M. Involvement of nitric oxide in nitrous oxide anxiolysis in the elevated plus-maze. Pharmacol. Biochem. Behav. 1994;48:689–692.
    1. Li S., Dai Y., Quock R.M. Antisense knockdown of neuronal nitric oxide synthase antagonizes nitrous oxide-induced behavior. Brain Res. 2003;968:167–170.
    1. Li S., Quock R.M. Effects of a nitric oxide donor on behavior and interaction with nitrous oxide in the mouse light/dark exploration test. Eur. J. Pharmacol. 2002;447:75–78.
    1. Li S., Ohgami Y., Dai Y. Antagonism of nitrous oxide-induced anxiolytic-like behavior in the mouse light/dark exploration procedure by pharmacologic disruption of endogenous nitric oxide function. Psychopharmacology. 2003;166:366–372.
    1. Gergerlioglu H.S., Savas H.A., Bulbul F. Changes in nitric oxide level and superoxide dismutase activity during antimanic treatment. Prog. Neuro- Psychopharm. Biol. Psychiatr. 2007;31:697–702.
    1. Andreazza A.C., Kauer-Sant'anna M., Frey B.N. Oxidative stress markers in bipolar disorder: a meta-analysis. J. Affect. Disord. 2008;111:135–144.
    1. Thompson J.M., Neave N., Moss M.C. Cognitive properties of sedation agents: comparison of the effects of nitrous oxide and midazolam on memory and mood. Br. Dent. J. 1999;187:557–562.
    1. Sambataro F., Wolf R.C. Embarking on antidepressant response prediction using brain perfusion estimation. EClininical. Med. 2019;10:4–5. doi: 10.1016/j.eclinm.2019.04.008. 2019/06/14.
    1. Kessler R.C., van Loo H.M., Wardenaar K.J. Using patient self-reports to study heterogeneity of treatment effects in major depressive disorder. Epidemiol. Psychiatr. Sci. 2017;26:22–36. doi: 10.1017/s2045796016000020. 2016/01/27.
    1. Simon G.E., Perlis R.H. Personalized medicine for depression: can we match patients with treatments? Am. J. Psychiatr. 2010;167:1445–1455. doi: 10.1176/appi.ajp.2010.09111680. 2010/09/17.
    1. Breitenstein B., Scheuer S., Holsboer F. Are there meaningful biomarkers of treatment response for depression? Drug Discov. Today. 2014;19:539–561. doi: 10.1016/j.drudis.2014.02.002. 2014/02/25.
    1. Ranft A., Kurz J., Becker K. Nitrous oxide (N2O) pre- and postsynaptically attenuates NMDA receptor-mediated neurotransmission in the amygdala. Neuropharmacology. 2007;52:716–723.
    1. Zarate C.A., Jr., Machado-Vieira R. Potential pathways involved in the rapid antidepressant effects of nitrous oxide. Biol. Psychiatr. 2015;78:2–4. doi: 10.1016/j.biopsych.2015.04.007.
    1. Zorumski C.F., Nagele P., Mennerick S. Treatment-resistant major depression: rationale for NMDA receptors as targets and nitrous oxide as therapy. Front. Psychiatr. 2015;6 doi: 10.3389/fpsyt.2015.00172. 172-172.
    1. Deutsch G., Samra S.K. Effects of nitrous oxide on global and regional cortical blood flow. Stroke. 1990;21:1293–1298. doi: 10.1161/01.str.21.9.1293.
    1. Ates-Alagoz Z., Adejare A. NMDA receptor antagonists for treatment of depression. Pharmaceuticals. 2013;6:480–499. doi: 10.3390/ph6040480. 2013/11/28.
    1. Machado-Vieira R., Salvadore G., Diazgranados N. Ketamine and the next generation of antidepressants with a rapid onset of action. Pharmacol. Ther. 2009;123:143–150. doi: 10.1016/j.pharmthera.2009.02.010. 2009/04/29.
    1. Kaufman M.B. Pharmaceutical approval update. P T. 2019;44:251–254.
    1. Krystal J.H., Sanacora G., Duman R.S. Rapid-acting glutamatergic antidepressants: the path to ketamine and beyond. Biol. Psychiatr. 2013;73:1133–1141.
    1. Zarate C.A., Jr., Singh J.B., Carlson P.J. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch. Gen. Psychiatr. 2006;63:856–864.
    1. Zarate C.A., Jr., Brutsche N.E., Ibrahim L. Replication of ketamine's antidepressant efficacy in bipolar depression: a randomized controlled add-on trial. Biol. Psychiatr. 2012;71:939–946.
    1. Goldstein B.I., Schaffer A., Wang S. Excessive and premature new-onset cardiovascular disease among adults with bipolar disorder in the US NESARC cohort. J. Clin. Psychiatr. 2015;76:163–169. doi: 10.4088/JCP.14m09300. 2015/03/06.
    1. Goldstein B.I., Carnethon M.R., Matthews K.A. Major depressive disorder and bipolar disorder predispose youth to accelerated atherosclerosis and early cardiovascular disease: a scientific statement from the American heart association. Circulation. 2015;132:965–986. doi: 10.1161/CIR.0000000000000229.
    1. Crump C., Sundquist K., Winkleby M.A. Comorbidities and mortality in bipolar disorder: a Swedish national cohort studycomorbidities and mortality in bipolar disordercomorbidities and mortality in bipolar disorder. JAMA Psychiatry. 2013;70:931–939.
    1. Goldstein B.I., Fagiolini A., Houck P. Cardiovascular disease and hypertension among adults with bipolar I disorder in the United States. Bipolar Disord. 2009;11:657–662.
    1. McDonald W.M., Tupler L.A., Marsteller F.A. Hyperintense lesions on magnetic resonance images in bipolar disorder. Biol. Psychiatr. 1999;45:965–971.
    1. Mahon K., Burdick K.E., Szeszko P.R. A role for white matter abnormalities in the pathophysiology of bipolar disorder. Neurosci. Biobehav. Rev. 2010;34:533–554.
    1. Rybakowski J.K., Wykretowicz A., Heymann-Szlachcinska A. Impairment of endothelial function in unipolar and bipolar depression. Biol. Psychiatr. 2006;60:889–891.
    1. Orosz A, Jann K, Federspiel A, et al. Reduced cerebral blood flow within the default-mode network and within total gray matter in major depression. Brain Connect.; 2:. 303-310.
    1. Ishizaki J., Yamamoto H., Takahashi T. Changes in regional cerebral blood flow following antidepressant treatment in late-life depression. Int. J. Geriatr. Psychiatr. 2008;23:805–811.
    1. Takano H., Motohashi N., Uema T. Changes in regional cerebral blood flow during acute electroconvulsive therapy in patients with depression: positron emission tomographic study. Br. J. Psychiatry. 2007;190:63–68.
    1. Bench C.J., Frackowiak R.S., Dolan R.J. Changes in regional cerebral blood flow on recovery from depression. Psychol. Med. 1995;25:247–261.
    1. Bench C.J., Friston K.J., Brown R.G. Regional cerebral blood flow in depression measured by positron emission tomography: the relationship with clinical dimensions. Psychol. Med. 1993;23:579–590.
    1. Berman K.F., Doran A.R., Pickar D. Is the mechanism of prefrontal hypofunction in depression the same as in schizophrenia? Regional cerebral blood flow during cognitive activation. Br. J. Psychiatry. 1993;162:183–192.
    1. Almeida J.R.C., Mourao-Miranda J., Aizenstein H.J. Pattern recognition analysis of anterior cingulate cortex blood flow to classify depression polarity. Br. J. Psychiatr. 2013 doi: 10.1192/bjp.bp.112.122838.
    1. Martinowich K., Jimenez D.V., Zarate C.A. Rapid antidepressant effects: moving right along. Mol. Psychiatr. 2013;18:856–863.
    1. Kraus C., Rabl U., Vanicek T. Administration of ketamine for unipolar and bipolar depression. Int. J. Psychiatr. Clin. Pract. 2017;21:2–12. doi: 10.1080/13651501.2016.1254802.
    1. Diazgranados N., Ibrahim L., Brutsche N.E. A randomized add-on trial of an n-methyl-d-aspartate antagonist in treatment-resistant bipolar depression. Arch. Gen. Psychiatr. 2010;67:793–802.
    1. Salvadore G., van der Veen J.W., Zhang Y. An investigation of amino-acid neurotransmitters as potential predictors of clinical improvement to ketamine in depression. Int. J. Neuropsychopharmacol. 2012;15:1063–1072. 2012.
    1. Salvadore G., Cornwell B.R., Colon-Rosario V. Increased anterior cingulate cortical activity in response to fearful faces: a neurophysiological biomarker that predicts rapid antidepressant response to ketamine. Biol. Psychiatr. 2009;65:289–295.
    1. Salvadore G., Cornwell B.R., Sambataro F. Anterior cingulate desynchronization and functional connectivity with the amygdala during a working memory task predict rapid antidepressant response to ketamine. Neuropsychopharmacology. 2010;35:1415–1422.
    1. Zarate C.A., Jr., Brutsche N., Laje G. Relationship of ketamine's plasma metabolites with response, diagnosis, and side effects in major depression. Biol. Psychiatr. 2012;72:331–338.
    1. Luckenbaugh D.A., Ibrahim L., Brutsche N. Family history of alcohol dependence and antidepressant response to an N-methyl-D-aspartate antagonist in bipolar depression. Bipolar Disord. 2012;14:880–887.
    1. Laje G., Lally N., Mathews D. Brain-derived neurotrophic factor Val66Met polymorphism and antidepressant efficacy of ketamine in depressed patients. Biol. Psychiatr. 2012;72:e27–28.
    1. Zarate C.A., Jr., Mathews D.C., Furey M.L. Human biomarkers of rapid antidepressant effects. Biol. Psychiatr. 2013;73:1142–1155.
    1. Carlson P.J., Diazgranados N., Nugent A.C. Neural correlates of rapid antidepressant response to ketamine in treatment-resistant unipolar depression: a preliminary positron emission tomography study. Biol. Psychiatr. 2013;73:1213–1221.
    1. Hintikka J., Tolmunen T., Tanskanen A. High vitamin B12 level and good treatment outcome may be associated in major depressive disorder. BMC Psychiatr. 2003;3 doi: 10.1186/1471-244x-3-17. 17. 2003/12/03.
    1. Singh J.B., Fedgchin M., Daly E.J. A double-blind, randomized, placebo-controlled, dose-frequency study of intravenous ketamine in patients with treatment-resistant depression. Am. J. Psychiatr. 2016;173:816–826. doi: 10.1176/appi.ajp.2016.16010037. 2016/04/09.
    1. Shiroma P.R., Johns B., Kuskowski M. Augmentation of response and remission to serial intravenous subanesthetic ketamine in treatment resistant depression. J. Affect. Disord. 2014;155:123–129. doi: 10.1016/j.jad.2013.10.036. 2013/11/26.
    1. Murrough J.W., Perez A.M., Pillemer S. Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol. Psychiatr. 2013;74:250–256. doi: 10.1016/j.biopsych.2012.06.022. 2012/07/31.
    1. Perico C.A.-M., Skaf C.R., Yamada A. Relationship between regional cerebral blood flow and separate symptom clusters of major depression: a single photon emission computed tomography study using statistical parametric mapping. Neurosci. Lett. 2005;384:265–270.
    1. Fountoulakis K.N., Iacovides A., Gerasimou G. The relationship of regional cerebral blood flow with subtypes of major depression. Prog. Neuro- Psychopharm. Biol. Psychiatr. 2004;28:537–546.
    1. Davies J., Lloyd K.R., Jones I.K. Changes in regional cerebral blood flow with venlafaxine in the treatment of major depression. Am. J. Psychiatr. 2003;160:374–376.
    1. Rubin E., Sackeim H.A., Prohovnik I. Regional cerebral blood flow in mood disorders: IV. Comparison of mania and depression. Psychiatr. Res. 1995;61:1–10.
    1. Dolan R.J., Bench C.J., Brown R.G. Neuropsychological dysfunction in depression: the relationship to regional cerebral blood flow. Psychol. Med. 1994;24:849–857.
    1. Bonne O., Krausz Y., Gorfine M. Cerebral hypoperfusion in medication resistant, depressed patients assessed by Tc99m HMPAO SPECT. J. Affect. Disord. 1996;41:163–171.
    1. Kito S, Hasegawa T and Koga Y. Cerebral blood flow in the ventromedial prefrontal cortex correlates with treatment response to low-frequency right prefrontal repetitive transcranial magnetic stimulation in the treatment of depression. Psychiatr. Clin. Neurosci.; 66:. 138-145.
    1. Kosel M, Brockmann H, Frick C, et al. Chronic vagus nerve stimulation for treatment-resistant depression increases regional cerebral blood flow in the dorsolateral prefrontal cortex. Psychiatr. Res.; 191:. 153-159.
    1. Kito S., Fujita K., Koga Y. Changes in regional cerebral blood flow after repetitive transcranial magnetic stimulation of the left dorsolateral prefrontal cortex in treatment-resistant depression. J. Neuropsychiatry Clin. Neurosci. 2008;20:74–80.
    1. Segawa K., Azuma H., Sato K. Regional cerebral blood flow changes in depression after electroconvulsive therapy. Psychiatr. Res. 2006;147:135–143.
    1. Joe A.Y., Tielmann T., Bucerius J. Response-dependent differences in regional cerebral blood flow changes with citalopram in treatment of major depression. J. Nucl. Med. 2006;47:1319–1325.
    1. Silfverskiold P., Risberg J. Regional cerebral blood flow in depression and mania. Arch. Gen. Psychiatr. 1989;46:253–259.
    1. Kruger S., Seminowicz D., Goldapple K. State and trait influences on mood regulation in bipolar disorder: blood flow differences with an acute mood challenge. Biol. Psychiatr. 2003;54:1274–1283.
    1. Benabarre A., Vieta E., Martin F. Clinical value of 99mTc-HMPAO SPECT in depressed bipolar I patients. Psychiatr. Res. 2004;132:285–289.
    1. Toma S., MacIntosh B.J., Swardfager W. Cerebral blood flow in bipolar disorder: a systematic review. J. Affect. Disord. 2018;241:505–513. doi: 10.1016/j.jad.2018.08.040. 2018/08/28.
    1. Adams A.K. The delayed arrival: from Davy (1800) to Morton (1846) J. R. Soc. Med. 1996;89:96–100. 1996/02/01.
    1. Jevtovic-Todorovic V., Todorovic S.M., Mennerick S. Nitrous oxide (laughing gas) is an NMDA antagonist, neuroprotectant and neurotoxin. Nat. Med. 1998;4:460–463.
    1. Walsh K., Das R.K., Kamboj S.K. The subjective response to nitrous oxide is a potential pharmaco-endophenotype for alcohol use disorder: a preliminary study with heavy drinkers. Int. J. Neuropsychopharmacol. 2016;20:346–350. doi: 10.1093/ijnp/pyw063.
    1. Kolbitsch C., Lorenz I.H., Hörmann C. Sevoflurane and nitrous oxide increase regional cerebral blood flow (rCBF) and regional cerebral blood volume (rCBV) in a drug-specific manner in human volunteers. Magn. Reson. Imag. 2001;19:1253–1260.
    1. Reinstrup P., Ryding E., Algotsson L. Effects of nitrous oxide on human regional cerebral blood flow and isolated pial arteries. Anesthesiology. 1994;81:396–402.
    1. Abi Zeid Daou M., Boyd B.D., Donahue M.J. Anterior-posterior gradient differences in lobar and cingulate cortex cerebral blood flow in late-life depression. J. Psychiatr. Res. 2018;97:1–7. doi: 10.1016/j.jpsychires.2017.11.005. 2017/11/21.
    1. Kolbitsch C., Lorenz I.H., Hormann C. Sevoflurane and nitrous oxide increase regional cerebral blood flow (rCBF) and regional cerebral blood volume (rCBV) in a drug-specific manner in human volunteers. Magn. Reson. Imaging. 2001;19:1253–1260. doi: 10.1016/s0730-725x(01)00465-9. 2002/01/24.
    1. Deutsch G., Samra S.K. Effects of nitrous oxide on global and regional cortical blood flow. Stroke. 1990;21:1293–1298.
    1. Lavoie K.L., Pelletier R., Arsenault A. Association between clinical depression and endothelial function measured by forearm hyperemic reactivity. Psychosom. Med. 2010;72:20–26.
    1. Broadley A.J.M., Korszun A., Abdelaal E. Metyrapone improves endothelial dysfunction in patients with treated depression. J. Am. Coll. Cardiol. 2006;48:170–175.
    1. de Wet J., Lichtigfeld F.J., Gillman M.A. Psychotropic analgesic nitrous oxide (PAN) for hyperactivity. Aust. N. Z. J. Psychiatr. 2001;35:543–544.
    1. Gillman M.A., Lichtigfeld F.J., Young T.N. Psychotropic analgesic nitrous oxide for alcoholic withdrawal states. Cochrane Database Syst. Rev. 2007;18(2) doi: 10.1002/14651858.CD005190.pub2.
    1. Daynes G., Gillman M.A. Psychotropic analgesic nitrous oxide prevents craving after withdrawal for alcohol, cannabis and tobacco. Int. J. Neurosci. 1994;76:13–16.
    1. File S.E., Easton P., Skelly A.M. Amnesia for dental procedures and mood change following treatment with nitrous oxide or midazolam. Int. Clin. Psychopharmacol. 1991;6:169–178.
    1. Zacny J.P., Hurst R.J., Graham L. Preoperative dental anxiety and mood changes during nitrous oxide inhalation. JADA (J. Am. Dent. Assoc.) 2002;133:82–88.
    1. Nagele P., Zorumski C.F., Conway C. Exploring nitrous oxide as treatment of mood disorders: basic concepts. J. Clin. Psychopharmacol. 2018;38:144–148. doi: 10.1097/JCP.0000000000000837.
    1. Abraini J.H., David H.N., Lemaire M. Potentially neuroprotective and therapeutic properties of nitrous oxide and xenon. Ann. N. Y. Acad. Sci. 2005;1053:289–300. doi: 10.1196/annals.1344.025. 2005/09/24.
    1. Logan R.W., McClung C.A. Animal models of bipolar mania: the past, present and future. Neuroscience. 2016;321:163–188. doi: 10.1016/j.neuroscience.2015.08.041. 08/24.
    1. Das R.K., Tamman A., Nikolova V. Nitrous oxide speeds the reduction of distressing intrusive memories in an experimental model of psychological trauma. Psychol. Med. 2016;46:1749–1759. doi: 10.1017/S003329171600026X.
    1. Alkire M.T., Hayama H., Reist C. American Society of Anesthesiologists Annual Meeting San Francisco. 2013. Emotional memory and emotional responsiveness with the NMDA antagonists ketamine or nitrous oxide: an fMRI study.

Source: PubMed

3
Iratkozz fel