1932

Abstract

Exposure to stressful stimuli activates kappa opioid receptor (KOR) signaling, a process known to produce aversion and dysphoria in humans and other species. This endogenous opioid system is dysregulated in stress-related disorders, specifically in major depressive disorder (MDD). These findings serve as the foundation for a growing interest in the therapeutic potential of KOR antagonists as novel antidepressants. In this review, data supporting the hypothesis of dysregulated KOR function in MDD are considered. The clinical data demonstrating the therapeutic efficacy and safety of selective and mixed opioid antagonists are then presented. Finally, the preclinical evidence illustrating the induction of behaviors relevant to the endophenotypes of MDD and KOR antagonist activity in stress-naïve and stress-exposed animals is evaluated. Overall, this review highlights the emergent literature supporting the pursuit of KOR antagonists as novel therapeutics for MDD and other stress-related disorders.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-010919-023317
2020-01-06
2024-04-23
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/60/1/annurev-pharmtox-010919-023317.html?itemId=/content/journals/10.1146/annurev-pharmtox-010919-023317&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    WHO (World Health Organ.) 2017. Depression and Other Common Mental Disorders: Global Health Estimates Geneva: WHO
  2. 2. 
    Greenberg PE, Fournier AA, Sisitsky T, Pike CT, Kessler RC 2015. The economic burden of adults with major depressive disorder in the United States (2005 and 2010). J. Clin. Psychiatry 76:155–62
    [Google Scholar]
  3. 3. 
    Fava M. 2003. Diagnosis and definition of treatment-resistant depression. Biol. Psychiatry 53:649–59
    [Google Scholar]
  4. 4. 
    McIntyre RS, Filteau MJ, Martin L, Patry S, Carvalho A et al. 2014. Treatment-resistant depression: definitions, review of the evidence, and algorithmic approach. J. Affect. Disord. 156:1–7
    [Google Scholar]
  5. 5. 
    Lutz PE, Kieffer BL. 2013. Opioid receptors: distinct roles in mood disorders. Trends Neurosci 36:195–206
    [Google Scholar]
  6. 6. 
    Van't Veer A, Carlezon WA Jr 2013. Role of kappa-opioid receptors in stress and anxiety-related behavior. Psychopharmacology 229:435–52
    [Google Scholar]
  7. 7. 
    Pietrzak RH, Naganawa M, Huang Y, Corsi-Travali S, Zheng MQ et al. 2014. Association of in vivo κ-opioid receptor availability and the transdiagnostic dimensional expression of trauma-related psychopathology. JAMA Psychiatry 71:1262–70
    [Google Scholar]
  8. 8. 
    Miller JM, Zanderigo F, Purushothaman PD, DeLorenzo C, Rubin-Falcone H et al. 2018. Kappa opioid receptor binding in major depression: a pilot study. Synapse 72:e22042
    [Google Scholar]
  9. 9. 
    Deo AJ, Huang YY, Hodgkinson CA, Xin Y, Oquendo MA et al. 2013. A large-scale candidate gene analysis of mood disorders: evidence of neurotrophic tyrosine kinase receptor and opioid receptor signaling dysfunction. Psychiatr. Genet. 23:47–55
    [Google Scholar]
  10. 10. 
    Anderson SA, Michaelides M, Zarnegar P, Ren Y, Fagergren P et al. 2013. Impaired periamygdaloid-cortex prodynorphin is characteristic of opiate addiction and depression. J. Clin. Investig. 123:5334–41
    [Google Scholar]
  11. 11. 
    Hurd YL. 2002. Subjects with major depression or bipolar disorder show reduction of prodynorphin mRNA expression in discrete nuclei of the amygdaloid complex. Mol. Psychiatry 7:75–81
    [Google Scholar]
  12. 12. 
    Peckys D, Hurd YL. 2001. Prodynorphin and κ opioid receptor mRNA expression in the cingulate and prefrontal cortices of subjects diagnosed with schizophrenia or affective disorders. Brain Res. Bull. 55:619–24
    [Google Scholar]
  13. 13. 
    Hurd YL, Herman MM, Hyde TM, Bigelow LB, Weinberger DR, Kleinman JE 1997. Prodynorphin mRNA expression is increased in the patch versus matrix compartment of the caudate nucleus in suicide subjects. Mol. Psychiatry 2:495–500
    [Google Scholar]
  14. 14. 
    Lutz PE, Almeida D, Belzeaux R, Yalcin I, Turecki G 2018. Epigenetic regulation of the kappa opioid receptor gene by an insertion–deletion in the promoter region. Eur. Neuropsychopharmacol. 28:334–40
    [Google Scholar]
  15. 15. 
    Lutz PE, Gross JA, Dhir SK, Maussion G, Yang J et al. 2018. Epigenetic regulation of the kappa opioid receptor by child abuse. Biol. Psychiatry 84:751–61
    [Google Scholar]
  16. 16. 
    Carroll FI, Carlezon WA Jr 2013. Development of kappa opioid receptor antagonists. J. Med. Chem. 56:2178–95
    [Google Scholar]
  17. 17. 
    Carlezon WA Jr, Krystal AD. 2016. Kappa-opioid antagonists for psychiatric disorders: from bench to clinical trials. Depress. Anxiety 33:895–906
    [Google Scholar]
  18. 18. 
    Urbano M, Guerrero M, Rosen H, Roberts E 2014. Antagonists of the kappa opioid receptor. Bioorg. Med. Chem. Lett. 24:2021–32
    [Google Scholar]
  19. 19. 
    Mague SD, Pliakas AM, Todtenkopf MS, Tomasiewicz HC, Zhang Y et al. 2003. Antidepressant-like effects of κ-opioid receptor antagonists in the forced swim test in rats. J. Pharmacol. Exp. Ther. 305:323–30
    [Google Scholar]
  20. 20. 
    Reed B, Butelman ER, Fry RS, Kimani R, Kreek MJ 2018. Repeated administration of opra kappa (LY2456302), a novel, short-acting, selective KOP-r antagonist, in persons with and without cocaine dependence. Neuropsychopharmacology 43:739–50
    [Google Scholar]
  21. 21. 
    Naganawa M, Dickinson GL, Zheng MQ, Henry S, Vandenhende F et al. 2016. Receptor occupancy of the κ-opioid antagonist LY2456302 measured with positron emission tomography and the novel radiotracer 11C-LY2795050. J. Pharmacol. Exp. Ther. 356:260–66
    [Google Scholar]
  22. 22. 
    Rorick-Kehn LM, Witkin JM, Statnick MA, Eberle EL, McKinzie JH et al. 2014. LY2456302 is a novel, potent, orally-bioavailable small molecule kappa-selective antagonist with activity in animal models predictive of efficacy in mood and addictive disorders. Neuropharmacology 77:131–44
    [Google Scholar]
  23. 23. 
    Lowe SL, Wong CJ, Witcher J, Gonzales CR, Dickinson GL et al. 2014. Safety, tolerability, and pharmacokinetic evaluation of single- and multiple-ascending doses of a novel kappa opioid receptor antagonist LY2456302 and drug interaction with ethanol in healthy subjects. J. Clin. Pharmacol. 54:968–78
    [Google Scholar]
  24. 24. 
    Krystal AD, Pizzagalli DA, Mathew SJ, Sanacora G, Keefe R et al. 2018. The first implementation of the NIMH FAST-FAIL approach to psychiatric drug development. Nat. Rev. Drug Discov. 18:82–84
    [Google Scholar]
  25. 25. 
    Toljan K, Vrooman B. 2018. Low-dose naltrexone (LDN)—review of therapeutic utilization. Med. Sci. 6:82
    [Google Scholar]
  26. 26. 
    Farren CK, O'Malley S, Grebski G, Maniar S, Porter M, Kreek MJ 1999. Variable dose naltrexone-induced hypothalamic-pituitary-adrenal stimulation in abstinent alcoholics: a preliminary study. Alcohol Clin. Exp. Res. 23:502–8
    [Google Scholar]
  27. 27. 
    Volavka J, Cho D, Mallya A, Bauman J 1979. Naloxone increases ACTH and cortisol levels in man. N. Engl. J. Med. 300:1056–57
    [Google Scholar]
  28. 28. 
    Mischoulon D, Hylek L, Yeung AS, Clain AJ, Baer L et al. 2017. Randomized, proof-of-concept trial of low dose naltrexone for patients with breakthrough symptoms of major depressive disorder on antidepressants. J. Affect. Disord. 208:6–14
    [Google Scholar]
  29. 29. 
    Williams NR, Heifets BD, Blasey C, Sudheimer K, Pannu J et al. 2018. Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. Am. J. Psychiatry 175:1205–15
    [Google Scholar]
  30. 30. 
    Raynor K, Kong H, Chen Y, Yasuda K, Yu L et al. 1994. Pharmacological characterization of the cloned kappa-, delta-, and mu-opioid receptors. Mol. Pharmacol. 45:330–34
    [Google Scholar]
  31. 31. 
    Gupta A, Devi LA, Gomes I 2011. Potentiation of μ-opioid receptor-mediated signaling by ketamine. J. Neurochem. 119:294–302
    [Google Scholar]
  32. 32. 
    Nemeth CL, Paine TA, Rittiner JE, Beguin C, Carroll FI et al. 2010. Role of kappa-opioid receptors in the effects of salvinorin A and ketamine on attention in rats. Psychopharmacology 210:263–74
    [Google Scholar]
  33. 33. 
    Ritter AJ. 2002. Naltrexone in the treatment of heroin dependence: relationship with depression and risk of overdose. Aust. N. Z. J. Psychiatry 36:224–28
    [Google Scholar]
  34. 34. 
    Krupitsky E, Zvartau E, Blokhina E, Verbitskaya E, Wahlgren V et al. 2016. Anhedonia, depression, anxiety, and craving in opiate dependent patients stabilized on oral naltrexone or an extended release naltrexone implant. Am. J. Drug Alcohol Abuse 42:614–20
    [Google Scholar]
  35. 35. 
    Latif ZE, Saltyte Benth J, Solli KK, Opheim A, Kunoe N et al. 2019. Anxiety, depression, and insomnia among adults with opioid dependence treated with extended-release naltrexone versus buprenorphine-naloxone: a randomized clinical trial and follow-up study. JAMA Psychiatry 76:2127–34
    [Google Scholar]
  36. 36. 
    Mysels DJ, Cheng WY, Nunes EV, Sullivan MA 2011. The association between naltrexone treatment and symptoms of depression in opioid-dependent patients. Am. J. Drug Alcohol Abuse 37:22–26
    [Google Scholar]
  37. 37. 
    Zaaijer ER, van Dijk L, de Bruin K, Goudriaan AE, Lammers LA et al. 2015. Effect of extended-release naltrexone on striatal dopamine transporter availability, depression and anhedonia in heroin-dependent patients. Psychopharmacology 232:2597–607
    [Google Scholar]
  38. 38. 
    Brown ES, Beard L, Dobbs L, Rush AJ 2006. Naltrexone in patients with bipolar disorder and alcohol dependence. Depress. Anxiety 23:492–95
    [Google Scholar]
  39. 39. 
    Gerra G, Fantoma A, Zaimovic A 2006. Naltrexone and buprenorphine combination in the treatment of opioid dependence. J. Psychopharmacol. 20:806–14
    [Google Scholar]
  40. 40. 
    Petrakis IL, Ralevski E, Desai N, Trevisan L, Gueorguieva R et al. 2012. Noradrenergic versus serotonergic antidepressant with or without naltrexone for veterans with PTSD and comorbid alcohol dependence. Neuropsychopharmacology 37:996–1004
    [Google Scholar]
  41. 41. 
    Cowan A. 2007. Buprenorphine: the basic pharmacology revisited. J. Addict. Med. 1:68–72
    [Google Scholar]
  42. 42. 
    Kosten TR, Morgan C, Kosten TA 1990. Depressive symptoms during buprenorphine treatment of opioid abusers. J. Subst. Abuse Treat. 7:51–54
    [Google Scholar]
  43. 43. 
    Streck JM, Ochalek TA, Badger GJ, Sigmon SC 2018. Interim buprenorphine treatment during delays to comprehensive treatment: changes in psychiatric symptoms. Exp. Clin. Psychopharmacol. 26:403–9
    [Google Scholar]
  44. 44. 
    Ahmadi J, Jahromi MS, Ehsaei Z 2018. The effectiveness of different singly administered high doses of buprenorphine in reducing suicidal ideation in acutely depressed people with co-morbid opiate dependence: a randomized, double-blind, clinical trial. Trials 19:462
    [Google Scholar]
  45. 45. 
    Ahmadi J, Sefidfard Jahromi M 2018. Ultrarapid influence of buprenorphine on major depression in opioid-dependent patients: a double blind, randomized clinical trial. Subst. Use Misuse 53:286–89
    [Google Scholar]
  46. 46. 
    Emrich HM, Vogt P, Herz A 1982. Possible antidepressive effects of opioids: action of buprenorphine. Ann. N. Y. Acad. Sci. 398:108–12
    [Google Scholar]
  47. 47. 
    Bodkin JA, Zornberg GL, Lukas SE, Cole JO 1995. Buprenorphine treatment of refractory depression. J. Clin. Psychopharmacol. 15:49–57
    [Google Scholar]
  48. 48. 
    Ehrich E, Turncliff R, Du Y, Leigh-Pemberton R, Fernandez E et al. 2015. Evaluation of opioid modulation in major depressive disorder. Neuropsychopharmacology 40:1448–55
    [Google Scholar]
  49. 49. 
    Fava M, Memisoglu A, Thase ME, Bodkin JA, Trivedi MH et al. 2016. Opioid modulation with buprenorphine/samidorphan as adjunctive treatment for inadequate response to antidepressants: a randomized double-blind placebo-controlled trial. Am. J. Psychiatry 173:499–508
    [Google Scholar]
  50. 50. 
    Karp JF, Butters MA, Begley AE, Miller MD, Lenze EJ et al. 2014. Safety, tolerability, and clinical effect of low-dose buprenorphine for treatment-resistant depression in midlife and older adults. J. Clin. Psychiatry 75:e785–93
    [Google Scholar]
  51. 51. 
    Nyhuis PW, Gastpar M, Scherbaum N 2008. Opiate treatment in depression refractory to antidepressants and electroconvulsive therapy. J. Clin. Psychopharmacol. 28:593–95
    [Google Scholar]
  52. 52. 
    Yovell Y, Bar G, Mashiah M, Baruch Y, Briskman I et al. 2016. Ultra-low-dose buprenorphine as a time-limited treatment for severe suicidal ideation: a randomized controlled trial. Am. J. Psychiatry 173:491–98
    [Google Scholar]
  53. 53. 
    Norelli LJ, Smith HS, Sher L, Blackwood TA 2013. Buprenorphine in the treatment of non-suicidal self-injury: a case series and discussion of the literature. Int. J. Adolesc. Med. Health 25:323–30
    [Google Scholar]
  54. 54. 
    Bershad AK, Jaffe JH, Childs E, de Wit H 2015. Opioid partial agonist buprenorphine dampens responses to psychosocial stress in humans. Psychoneuroendocrinology 52:281–88
    [Google Scholar]
  55. 55. 
    Bershad AK, Seiden JA, de Wit H 2016. Effects of buprenorphine on responses to social stimuli in healthy adults. Psychoneuroendocrinology 63:43–49
    [Google Scholar]
  56. 56. 
    Bershad AK, Miller MA, Norman GJ, de Wit H 2018. Effects of opioid- and non-opioid analgesics on responses to psychosocial stress in humans. Horm. Behav. 102:41–47
    [Google Scholar]
  57. 57. 
    Bershad AK, Ruiz NA, de Wit H 2018. Effects of buprenorphine on responses to emotional stimuli in individuals with a range of mood symptomatology. Int. J. Neuropsychopharmacol. 21:120–27
    [Google Scholar]
  58. 58. 
    Syal S, Ipser J, Terburg D, Solms M, Panksepp J et al. 2015. Improved memory for reward cues following acute buprenorphine administration in humans. Psychoneuroendocrinology 53:10–15
    [Google Scholar]
  59. 59. 
    Ipser JC, Terburg D, Syal S, Phillips N, Solms M et al. 2013. Reduced fear-recognition sensitivity following acute buprenorphine administration in healthy volunteers. Psychoneuroendocrinology 38:166–70
    [Google Scholar]
  60. 60. 
    Fava M, Thase ME, Trivedi MH, Ehrich E, Martin WF et al. 2018. Opioid system modulation with buprenorphine/samidorphan combination for major depressive disorder: two randomized controlled studies. Mol. Psychiatry. https://doi.org/10.1038/s41380-018-0284-1
    [Crossref] [Google Scholar]
  61. 61. 
    Peckham AM, De La Cruz A, Dufresne RL 2018. Kappa opioid receptor antagonism: Are opioids the answer for treatment resistant depression. ? Ment. Health Clin. 8:175–83
    [Google Scholar]
  62. 62. 
    Bruchas MR, Land BB, Chavkin C 2010. The dynorphin/kappa opioid system as a modulator of stress-induced and pro-addictive behaviors. Brain Res 1314:44–55
    [Google Scholar]
  63. 63. 
    Knoll AT, Carlezon WA Jr 2010. Dynorphin, stress, and depression. Brain Res 1314:56–73
    [Google Scholar]
  64. 64. 
    Lalanne L, Ayranci G, Kieffer BL, Lutz PE 2014. The kappa opioid receptor: from addiction to depression, and back. Front. Psychiatry 5:170
    [Google Scholar]
  65. 65. 
    Maqueda AE, Valle M, Addy PH, Antonijoan RM, Puntes M et al. 2015. Salvinorin-A induces intense dissociative effects, blocking external sensory perception and modulating interoception and sense of body ownership in humans. Int. J. Neuropsychopharmacol. 18:pyv065
    [Google Scholar]
  66. 66. 
    Pfeiffer A, Brantl V, Herz A, Emrich HM 1986. Psychotomimesis mediated by kappa opiate receptors. Science 233:774–76
    [Google Scholar]
  67. 67. 
    Ranganathan M, Schnakenberg A, Skosnik PD, Cohen BM, Pittman B et al. 2012. Dose-related behavioral, subjective, endocrine, and psychophysiological effects of the κ opioid agonist salvinorin A in humans. Biol. Psychiatry 72:871–79
    [Google Scholar]
  68. 68. 
    Butelman ER, Kreek MJ. 2015. Salvinorin A, a kappa-opioid receptor agonist hallucinogen: pharmacology and potential template for novel pharmacotherapeutic agents in neuropsychiatric disorders. Front. Pharmacol. 6:190
    [Google Scholar]
  69. 69. 
    von Voigtlander PF, Lewis RA 1982. U50,488, a selective kappa opioid agonist: comparison to other reputed kappa agonists. Prog. Neuropsychopharmacol. Biol. Psychiatry 6:467–70
    [Google Scholar]
  70. 70. 
    Anderson RI, Morales M, Spear LP, Varlinskaya EI 2014. Pharmacological activation of kappa opioid receptors: aversive effects in adolescent and adult male rats. Psychopharmacology 231:1687–93
    [Google Scholar]
  71. 71. 
    Land BB, Bruchas MR, Schattauer SS, Giardino WJ, Aita M et al. 2009. Activation of the kappa opioid receptor in the dorsal raphe nucleus mediates the aversive effects of stress and reinstates drug seeking. PNAS 106:19168–73
    [Google Scholar]
  72. 72. 
    Chefer VI, Backman CM, Gigante ED, Shippenberg TS 2013. Kappa opioid receptors on dopaminergic neurons are necessary for kappa-mediated place aversion. Neuropsychopharmacology 38:2623–31
    [Google Scholar]
  73. 73. 
    Tejeda HA, Counotte DS, Oh E, Ramamoorthy S, Schultz-Kuszak KN et al. 2013. Prefrontal cortical kappa-opioid receptor modulation of local neurotransmission and conditioned place aversion. Neuropsychopharmacology 38:1770–79
    [Google Scholar]
  74. 74. 
    Shippenberg TS, Herz A. 1987. Place preference conditioning reveals the involvement of D1-dopamine receptors in the motivational properties of μ- and κ-opioid agonists. Brain Res 436:169–72
    [Google Scholar]
  75. 75. 
    Valenza M, Butelman ER, Kreek MJ 2017. Effects of the novel relatively short-acting kappa opioid receptor antagonist LY2444296 in behaviors observed after chronic extended-access cocaine self-administration in rats. Psychopharmacology 234:2219–31
    [Google Scholar]
  76. 76. 
    Bechara A, van der Kooy D 1987. Kappa receptors mediate the peripheral aversive effects of opiates. Pharmacol. Biochem. Behav. 28:227–33
    [Google Scholar]
  77. 77. 
    Bruchas MR, Land BB, Aita M, Xu M, Barot SK et al. 2007. Stress-induced p38 mitogen-activated protein kinase activation mediates κ-opioid-dependent dysphoria. J. Neurosci. 27:11614–23
    [Google Scholar]
  78. 78. 
    Ehrich JM, Messinger DI, Knakal CR, Kuhar JR, Schattauer SS et al. 2015. Kappa opioid receptor-induced aversion requires p38 MAPK activation in VTA dopamine neurons. J. Neurosci. 35:12917–31
    [Google Scholar]
  79. 79. 
    Grimwood S, Lu Y, Schmidt AW, Vanase-Frawley MA, Sawant-Basak A et al. 2011. Pharmacological characterization of 2-methyl-N-((2′-(pyrrolidin-1-ylsulfonyl)biphenyl-4-yl)methyl)propan-1-amine (PF-04455242), a high-affinity antagonist selective for κ-opioid receptors. J. Pharmacol. Exp. Ther. 339:555–66
    [Google Scholar]
  80. 80. 
    Kivell BM, Paton KF, Kumar N, Morani AS, Culverhouse A et al. 2018. Kappa opioid receptor agonist Mesyl Sal B attenuates behavioral sensitization to cocaine with fewer aversive side-effects than Salvinorin A in rodents. Molecules 23:2602
    [Google Scholar]
  81. 81. 
    Laman-Maharg AR, Copeland T, Sanchez EO, Campi KL, Trainor BC 2017. The long-term effects of stress and kappa opioid receptor activation on conditioned place aversion in male and female California mice. Behav. Brain Res. 332:299–307
    [Google Scholar]
  82. 82. 
    Land BB, Bruchas MR, Lemos JC, Xu M, Melief EJ, Chavkin C 2008. The dysphoric component of stress is encoded by activation of the dynorphin κ-opioid system. J. Neurosci. 28:407–14
    [Google Scholar]
  83. 83. 
    Logrip ML, Janak PH, Ron D 2009. Blockade of ethanol reward by the kappa opioid receptor agonist U50,488H. Alcohol 43:359–65
    [Google Scholar]
  84. 84. 
    Robles CF, McMackin MZ, Campi KL, Doig IE, Takahashi EY et al. 2014. Effects of kappa opioid receptors on conditioned place aversion and social interaction in males and females. Behav. Brain Res. 262:84–93
    [Google Scholar]
  85. 85. 
    Simonin F, Valverde O, Smadja C, Slowe S, Kitchen I et al. 1998. Disruption of the κ-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective κ-agonist U-50,488H and attenuates morphine withdrawal. EMBO J 17:886–97
    [Google Scholar]
  86. 86. 
    Wiley MD, Poveromo LB, Antapasis J, Herrera CM, Bolanos Guzman CA 2009. κ-Opioid system regulates the long-lasting behavioral adaptations induced by early-life exposure to methylphenidate. Neuropsychopharmacology 34:1339–50
    [Google Scholar]
  87. 87. 
    Braida D, Limonta V, Capurro V, Fadda P, Rubino T et al. 2008. Involvement of κ-opioid and endocannabinoid system on Salvinorin A–induced reward. Biol. Psychiatry 63:286–92
    [Google Scholar]
  88. 88. 
    Sufka KJ, Loria MJ, Lewellyn K, Zjawiony JK, Ali Z et al. 2014. The effect of Salvia divinorum and Mitragyna speciosa extracts, fraction and major constituents on place aversion and place preference in rats. J. Ethnopharmacol. 151:361–64
    [Google Scholar]
  89. 89. 
    Zhang Y, Butelman ER, Schlussman SD, Ho A, Kreek MJ 2005. Effects of the plant-derived hallucinogen salvinorin A on basal dopamine levels in the caudate putamen and in a conditioned place aversion assay in mice: agonist actions at kappa opioid receptors. Psychopharmacology 179:551–58
    [Google Scholar]
  90. 90. 
    Shippenberg TS, Bals-Kubik R, Herz A 1993. Examination of the neurochemical substrates mediating the motivational effects of opioids: role of the mesolimbic dopamine system and D-1 versus D-2 dopamine receptors. J. Pharmacol. Exp. Ther. 265:53–59
    [Google Scholar]
  91. 91. 
    Zan GY, Wang Q, Wang YJ, Chen JC, Wu X et al. 2016. p38 mitogen-activated protein kinase activation in amygdala mediates κ opioid receptor agonist U50,488H-induced conditioned place aversion. Neuroscience 320:122–28
    [Google Scholar]
  92. 92. 
    Zannas AS, Kim JH, West AE 2017. Regulation and function of MeCP2 Ser421 phosphorylation in U50488-induced conditioned place aversion in mice. Psychopharmacology 234:913–23
    [Google Scholar]
  93. 93. 
    Bals-Kubik R, Ableitner A, Herz A, Shippenberg TS 1993. Neuroanatomical sites mediating the motivational effects of opioids as mapped by the conditioned place preference paradigm in rats. J. Pharmacol. Exp. Ther. 264:489–95
    [Google Scholar]
  94. 94. 
    Sante AB, Nobre MJ, Brandao ML 2000. Place aversion induced by blockade of μ or activation of κ opioid receptors in the dorsal periaqueductal gray matter. Behav. Pharmacol. 11:583–89
    [Google Scholar]
  95. 95. 
    Todtenkopf MS, Marcus JF, Portoghese PS, Carlezon WA Jr 2004. Effects of κ-opioid receptor ligands on intracranial self-stimulation in rats. Psychopharmacology 172:463–70
    [Google Scholar]
  96. 96. 
    Conway SM, Puttick D, Russell S, Potter D, Roitman MF, Chartoff EH 2019. Females are less sensitive than males to the motivational- and dopamine-suppressing effects of kappa opioid receptor activation. Neuropharmacology 146:231–41
    [Google Scholar]
  97. 97. 
    Dinieri JA, Nemeth CL, Parsegian A, Carle T, Gurevich VV et al. 2009. Altered sensitivity to rewarding and aversive drugs in mice with inducible disruption of cAMP response element–binding protein function within the nucleus accumbens. J. Neurosci. 29:1855–59
    [Google Scholar]
  98. 98. 
    Russell SE, Rachlin AB, Smith KL, Muschamp J, Berry L et al. 2014. Sex differences in sensitivity to the depressive-like effects of the kappa opioid receptor agonist U-50488 in rats. Biol. Psychiatry 76:213–22
    [Google Scholar]
  99. 99. 
    Beguin C, Potter DN, Dinieri JA, Munro TA, Richards MR et al. 2008. N-methylacetamide analog of salvinorin A: a highly potent and selective κ-opioid receptor agonist with oral efficacy. J. Pharmacol. Exp. Ther. 324:188–95
    [Google Scholar]
  100. 100. 
    Carlezon WA Jr, Beguin C, DiNieri JA, Baumann MH, Richards MR et al. 2006. Depressive-like effects of the κ-opioid receptor agonist salvinorin A on behavior and neurochemistry in rats. J. Pharmacol. Exp. Ther. 316:440–47
    [Google Scholar]
  101. 101. 
    Chartoff EH, Ebner SR, Sparrow A, Potter D, Baker PM et al. 2016. Relative timing between kappa opioid receptor activation and cocaine determines the impact on reward and dopamine release. Neuropsychopharmacology 41:989–1002
    [Google Scholar]
  102. 102. 
    Ebner SR, Roitman MF, Potter DN, Rachlin AB, Chartoff EH 2010. Depressive-like effects of the kappa opioid receptor agonist salvinorin A are associated with decreased phasic dopamine release in the nucleus accumbens. Psychopharmacology 210:241–52
    [Google Scholar]
  103. 103. 
    Negus SS, O'Connell R, Morrissey E, Cheng K, Rice KC 2012. Effects of peripherally restricted kappa opioid receptor agonists on pain-related stimulation and depression of behavior in rats. J. Pharmacol. Exp. Ther. 340:501–9
    [Google Scholar]
  104. 104. 
    Leitl MD, Onvani S, Bowers MS, Cheng K, Rice KC et al. 2014. Pain-related depression of the mesolimbic dopamine system in rats: expression, blockade by analgesics, and role of endogenous κ-opioids. Neuropsychopharmacology 39:614–24
    [Google Scholar]
  105. 105. 
    Muschamp JW, Van't Veer A, Parsegian A, Gallo MS, Chen M et al. 2011. Activation of CREB in the nucleus accumbens shell produces anhedonia and resistance to extinction of fear in rats. J. Neurosci. 31:3095–103
    [Google Scholar]
  106. 106. 
    Dogra S, Kumar A, Umrao D, Sahasrabuddhe AA, Yadav PN 2016. Chronic kappa opioid receptor activation modulates NR2B: implication in treatment resistant depression. Sci. Rep. 6:33401
    [Google Scholar]
  107. 107. 
    Henderson-Redmond A, Czachowski C. 2014. Effects of systemic opioid receptor ligands on ethanol- and sucrose seeking and drinking in alcohol-preferring (P) and Long Evans rats. Psychopharmacology 231:4309–21
    [Google Scholar]
  108. 108. 
    Haj-Mirzaian A, Nikbakhsh R, Ramezanzadeh K, Rezaee M, Amini-Khoei H et al. 2019. Involvement of opioid system in behavioral despair induced by social isolation stress in mice. Biomed. Pharmacother. 109:938–44
    [Google Scholar]
  109. 109. 
    Casal-Dominguez JJ, Clark M, Traynor JR, Husbands SM, Bailey SJ 2013. In vivo and in vitro characterization of naltrindole-derived ligands at the κ-opioid receptor. J. Psychopharmacol. 27:192–202
    [Google Scholar]
  110. 110. 
    Laman-Maharg A, Williams AV, Zufelt MD, Minie VA, Ramos-Maciel S et al. 2018. Sex differences in the effects of a kappa opioid receptor antagonist in the forced swim test. Front. Pharmacol. 9:93
    [Google Scholar]
  111. 111. 
    Williams AV, Laman-Maharg A, Armstrong CV, Ramos-Maciel S, Minie VA, Trainor BC 2018. Acute inhibition of kappa opioid receptors before stress blocks depression-like behaviors in California mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 86:166–74
    [Google Scholar]
  112. 112. 
    Zhang LS, Wang J, Chen JC, Tao YM, Wang YH et al. 2015. Novel κ-opioid receptor agonist MB-1C-OH produces potent analgesia with less depression and sedation. Acta Pharmacol. Sin. 36:565–71
    [Google Scholar]
  113. 113. 
    Hang A, Wang YJ, He L, Liu JG 2015. The role of the dynorphin/κ opioid receptor system in anxiety. Acta Pharmacol. Sin. 36:783–90
    [Google Scholar]
  114. 114. 
    Narita M, Kaneko C, Miyoshi K, Nagumo Y, Kuzumaki N et al. 2006. Chronic pain induces anxiety with concomitant changes in opioidergic function in the amygdala. Neuropsychopharmacology 31:739–50
    [Google Scholar]
  115. 115. 
    Bruchas MR, Land BB, Lemos JC, Chavkin C 2009. CRF1-R activation of the dynorphin/kappa opioid system in the mouse basolateral amygdala mediates anxiety-like behavior. PLOS ONE 4:e8528
    [Google Scholar]
  116. 116. 
    Gillett K, Harshberger E, Valdez GR 2013. Protracted withdrawal from ethanol and enhanced responsiveness stress: regulation via the dynorphin/kappa opioid receptor system. Alcohol 47:359–65
    [Google Scholar]
  117. 117. 
    Smith JS, Schindler AG, Martinelli E, Gustin RM, Bruchas MR, Chavkin C 2012. Stress-induced activation of the dynorphin/kappa-opioid receptor system in the amygdala potentiates nicotine conditioned place preference. J. Neurosci. 32:1488–95
    [Google Scholar]
  118. 118. 
    Valdez GR, Harshberger E. 2012. Kappa opioid regulation of anxiety-like behavior during acute ethanol withdrawal. Pharmacol. Biochem. Behav. 102:44–47
    [Google Scholar]
  119. 119. 
    Wang YJ, Hang A, Lu YC, Long Y, Zan GY et al. 2016. κ Opioid receptor activation in different brain regions differentially modulates anxiety-related behaviors in mice. Neuropharmacology 110:92–101
    [Google Scholar]
  120. 120. 
    Varlinskaya EI, Spear LP, Diaz MR 2018. Stress alters social behavior and sensitivity to pharmacological activation of kappa opioid receptors in an age-specific manner in Sprague Dawley rats. Neurobiol. Stress 9:124–32
    [Google Scholar]
  121. 121. 
    Vanderschuren LJ, Niesink RJ, Spruijt BM, Van Ree JM 1995. μ- and κ-opioid receptor-mediated opioid effects on social play in juvenile rats. Eur. J. Pharmacol. 276:257–66
    [Google Scholar]
  122. 122. 
    Wright EC, Parks TV, Alexander JO, Supra R, Trainor BC 2018. Activation of kappa opioid receptors in the dorsal raphe have sex dependent effects on social behavior in California mice. Behav. Brain Res. 351:83–92
    [Google Scholar]
  123. 123. 
    Abraham AD, Fontaine HM, Song AJ, Andrews MM, Baird MA et al. 2018. κ-Opioid receptor activation in dopamine neurons disrupts behavioral inhibition. Neuropsychopharmacology 43:362–72
    [Google Scholar]
  124. 124. 
    Almatroudi A, Husbands SM, Bailey CP, Bailey SJ 2015. Combined administration of buprenorphine and naltrexone produces antidepressant-like effects in mice. J. Psychopharmacol. 29:812–21
    [Google Scholar]
  125. 125. 
    Almatroudi A, Ostovar M, Bailey CP, Husbands SM, Bailey SJ 2018. Antidepressant-like effects of BU10119, a novel buprenorphine analogue with mixed κ/μ receptor antagonist properties, in mice. Br. J. Pharmacol. 175:2869–80
    [Google Scholar]
  126. 126. 
    Beardsley PM, Howard JL, Shelton KL, Carroll FI 2005. Differential effects of the novel kappa opioid receptor antagonist, JDTic, on reinstatement of cocaine-seeking induced by footshock stressors versus cocaine primes and its antidepressant-like effects in rats. Psychopharmacology 183:118–26
    [Google Scholar]
  127. 127. 
    Browne CA, van Nest DS, Lucki I 2015. Antidepressant-like effects of buprenorphine in rats are strain dependent. Behav. Brain Res. 278:385–92
    [Google Scholar]
  128. 128. 
    Browne CA, Falcon E, Robinson SA, Berton O, Lucki I 2018. Reversal of stress-induced social interaction deficits by buprenorphine. Int. J. Neuropsychopharmacol. 21:164–74
    [Google Scholar]
  129. 129. 
    Bruning CA, Martini F, Soares SM, Sampaio TB, Gai BM et al. 2015. m-Trifluoromethyl-diphenyl diselenide, a multi-target selenium compound, prevented mechanical allodynia and depressive-like behavior in a mouse comorbid pain and depression model. Prog. Neuropsychopharmacol. Biol. Psychiatry 63:35–46
    [Google Scholar]
  130. 130. 
    Bruning CA, Martini F, Soares SM, Savegnago L, Sampaio TB, Nogueira CW 2015. Depressive-like behavior induced by tumor necrosis factor-α is attenuated by m-trifluoromethyl-diphenyl diselenide in mice. J. Psychiatr. Res. 66–67:75–83
    [Google Scholar]
  131. 131. 
    Bruning CA, Souza AC, Gai BM, Zeni G, Nogueira CW 2011. Antidepressant-like effect of m-trifluoromethyl-diphenyl diselenide in the mouse forced swimming test involves opioid and serotonergic systems. Eur. J. Pharmacol. 658:145–49
    [Google Scholar]
  132. 132. 
    Burke NN, Ferdousi M, Deaver DR, Finn DP, Roche M, Kelly JP 2019. Locomotor and anti-immobility effects of buprenorphine in combination with the opioid receptor modulator samidorphan in rats. Neuropharmacology 146:327–36
    [Google Scholar]
  133. 133. 
    Falcon E, Maier K, Robinson SA, Hill-Smith TE, Lucki I 2015. Effects of buprenorphine on behavioral tests for antidepressant and anxiolytic drugs in mice. Psychopharmacology 232:907–15
    [Google Scholar]
  134. 134. 
    Falcon E, Browne CA, Leon RM, Fleites VC, Sweeney R et al. 2016. Antidepressant-like effects of buprenorphine are mediated by kappa opioid receptors. Neuropsychopharmacology 41:2344–51
    [Google Scholar]
  135. 135. 
    Huang P, Yakovleva T, Aldrich JV, Tunis J, Parry C, Liu-Chen LY 2016. Two short-acting kappa opioid receptor antagonists (zyklophin and LY2444296) exhibited different behavioral effects from the long-acting antagonist norbinaltorphimine in mouse anxiety tests. Neurosci. Lett. 615:15–20
    [Google Scholar]
  136. 136. 
    Huang P, Tunis J, Parry C, Tallarida R, Liu-Chen LY 2016. Synergistic antidepressant-like effects between a kappa opioid antagonist (LY2444296) and a delta opioid agonist (ADL5859) in the mouse forced swim test. Eur. J. Pharmacol. 781:53–59
    [Google Scholar]
  137. 137. 
    Reed B, Fang N, Mayer-Blackwell B, Chen S, Yuferov V et al. 2012. Chromatin alterations in response to forced swimming underlie increased prodynorphin transcription. Neuroscience 220:109–18
    [Google Scholar]
  138. 138. 
    Rosa SG, Pesarico AP, Tagliapietra CF, da Luz SC, Nogueira CW 2017. Opioid system contribution to the antidepressant-like action of m-trifluoromethyl-diphenyl diselenide in mice: a compound devoid of tolerance and withdrawal syndrome. J. Psychopharmacol. 31:1250–62
    [Google Scholar]
  139. 139. 
    Rosa SG, Pesarico AP, Martini F, Nogueira CW 2018. m-Trifluoromethyl-diphenyl diselenide regulates prefrontal cortical MOR and KOR protein levels and abolishes the phenotype induced by repeated forced swim stress in mice. Mol. Neurobiol. 55:8991–9000
    [Google Scholar]
  140. 140. 
    Smith KL, Cunningham JI, Eyerman DJ, Dean RL 3rd, Deaver DR, Sanchez C 2019. Opioid system modulators buprenorphine and samidorphan alter behavior and extracellular neurotransmitter concentrations in the Wistar Kyoto rat. Neuropharmacology 146:316–26
    [Google Scholar]
  141. 141. 
    Wang J, Song Q, Xu A, Bao Y, Xu Y, Zhu Q 2017. Design, synthesis and biological evaluation of aminobenzyloxyarylamide derivatives as selective κ opioid receptor antagonists. Eur. J. Med. Chem. 130:15–25
    [Google Scholar]
  142. 142. 
    Pliakas AM, Carlson RR, Neve RL, Konradi C, Nestler EJ, Carlezon WA Jr 2001. Altered responsiveness to cocaine and increased immobility in the forced swim test associated with elevated cAMP response element-binding protein expression in nucleus accumbens. J. Neurosci. 21:7397–403
    [Google Scholar]
  143. 143. 
    Zhang H, Shi Y, Woods JH, Watson SJ, Ko M 2007. Central κ-opioid receptor-mediated antidepressant-like effects of nor-Binaltrophimine: behavioral and BDNF mRNA expression studies. Eur. J. Pharmacol. 570:89–96
    [Google Scholar]
  144. 144. 
    Carr GV, Bangasser DA, Bethea T, Young M, Valentino RJ, Lucki I 2010. Antidepressant-like effects of κ-opioid receptor antagonists in Wistar Kyoto rats. Neuropsychopharmacology 35:752–63
    [Google Scholar]
  145. 145. 
    Kaster MP, Budni J, Santos AR, Rodrigues AL 2007. Pharmacological evidence for the involvement of the opioid system in the antidepressant-like effect of adenosine in the mouse forced swimming test. Eur. J. Pharmacol. 576:91–98
    [Google Scholar]
  146. 146. 
    Zomkowski AD, Santos AR, Rodrigues AL 2005. Evidence for the involvement of the opioid system in the agmatine antidepressant-like effect in the forced swimming test. Neurosci. Lett. 381:279–83
    [Google Scholar]
  147. 147. 
    Browne CA, Erickson RL, Blendy JA, Lucki I 2017. Genetic variation in the behavioral effects of buprenorphine in female mice derived from a murine model of the OPRM1 A118G polymorphism. Neuropharmacology 117:401–7
    [Google Scholar]
  148. 148. 
    Carr GV, Lucki I. 2010. Comparison of the kappa-opioid receptor antagonist DIPPA in tests of anxiety-like behavior between Wistar Kyoto and Sprague Dawley rats. Psychopharmacology 210:295–302
    [Google Scholar]
  149. 149. 
    Knoll AT, Meloni EG, Thomas JB, Carroll FI, Carlezon WA Jr 2007. Anxiolytic-like effects of κ-opioid receptor antagonists in models of unlearned and learned fear in rats. J. Pharmacol. Exp. Ther. 323:838–45
    [Google Scholar]
  150. 150. 
    Robinson SA, Erickson RL, Browne CA, Lucki I 2017. A role for the mu opioid receptor in the antidepressant effects of buprenorphine. Behav. Brain Res. 319:96–103
    [Google Scholar]
  151. 151. 
    Tejeda HA, Hanks AN, Scott L, Mejias-Aponte C, Hughes ZA, O'Donnell P 2015. Prefrontal cortical kappa opioid receptors attenuate responses to amygdala inputs. Neuropsychopharmacology 40:2856–64
    [Google Scholar]
  152. 152. 
    Wittmann W, Schunk E, Rosskothen I, Gaburro S, Singewald N et al. 2009. Prodynorphin-derived peptides are critical modulators of anxiety and regulate neurochemistry and corticosterone. Neuropsychopharmacology 34:775–85
    [Google Scholar]
  153. 153. 
    Donahue RJ, Landino SM, Golden SA, Carroll FI, Russo SJ, Carlezon WA Jr 2015. Effects of acute and chronic social defeat stress are differentially mediated by the dynorphin/kappa-opioid receptor system. Behav. Pharmacol. 26:654–63
    [Google Scholar]
  154. 154. 
    Negus SS, Morrissey EM, Rosenberg M, Cheng K, Rice KC 2010. Effects of kappa opioids in an assay of pain-depressed intracranial self-stimulation in rats. Psychopharmacology 210:149–59
    [Google Scholar]
  155. 155. 
    Segat HJ, Martini F, Barcelos RC, Bruning CA, Nogueira CW, Burger ME 2016. m-Trifluoromethyl-diphenyldiselenide as a pharmacological tool to treat preference symptoms related to AMPH-induced dependence in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry 66:1–7
    [Google Scholar]
  156. 156. 
    Cordery SF, Taverner A, Ridzwan IE, Guy RH, Delgado-Charro MB et al. 2014. A non-rewarding, non-aversive buprenorphine/naltrexone combination attenuates drug-primed reinstatement to cocaine and morphine in rats in a conditioned place preference paradigm. Addict. Biol. 19:575–86
    [Google Scholar]
  157. 157. 
    Van't Veer A, Yano JM, Carroll FI, Cohen BM, Carlezon WA Jr 2012. Corticotropin-releasing factor (CRF)-induced disruption of attention in rats is blocked by the κ-opioid receptor antagonist JDTic. Neuropsychopharmacology 37:2809–16
    [Google Scholar]
  158. 158. 
    Beard C, Donahue RJ, Dillon DG, Van't Veer A, Webber C et al. 2015. Abnormal error processing in depressive states: a translational examination in humans and rats. Transl. Psychiatry 5:e564
    [Google Scholar]
  159. 159. 
    Donahue RJ, Venkataraman A, Carroll FI, Meloni EG, Carlezon WA Jr 2016. Pituitary adenylate cyclase-activating polypeptide disrupts motivation, social interaction, and attention in male Sprague Dawley rats. Biol. Psychiatry 80:955–64
    [Google Scholar]
  160. 160. 
    Braida D, Donzelli A, Martucci R, Capurro V, Sala M 2011. Learning and memory impairment induced by salvinorin A, the principal ingredient of Salvia divinorum, in Wistar rats. Int. J. Toxicol. 30:650–61
    [Google Scholar]
  161. 161. 
    Knoll AT, Muschamp JW, Sillivan SE, Ferguson D, Dietz DM et al. 2011. Kappa opioid receptor signaling in the basolateral amygdala regulates conditioned fear and anxiety in rats. Biol. Psychiatry 70:425–33
    [Google Scholar]
  162. 162. 
    Newton SS, Thome J, Wallace TL, Shirayama Y, Schlesinger L et al. 2002. Inhibition of cAMP response element-binding protein or dynorphin in the nucleus accumbens produces an antidepressant-like effect. J. Neurosci. 22:10883–90
    [Google Scholar]
  163. 163. 
    Rogala B, Li Y, Li S, Chen X, Kirouac GJ 2012. Effects of a post-shock injection of the kappa opioid receptor antagonist norbinaltorphimine (norBNI) on fear and anxiety in rats. PLOS ONE 7:e49669
    [Google Scholar]
  164. 164. 
    Shirayama Y, Ishida H, Iwata M, Hazama GI, Kawahara R, Duman RS 2004. Stress increases dynorphin immunoreactivity in limbic brain regions and dynorphin antagonism produces antidepressant-like effects. J. Neurochem. 90:1258–68
    [Google Scholar]
  165. 165. 
    Carey AN, Lyons AM, Shay CF, Dunton O, McLaughlin JP 2009. Endogenous κ opioid activation mediates stress-induced deficits in learning and memory. J. Neurosci. 29:4293–300
    [Google Scholar]
  166. 166. 
    McLaughlin JP, Marton-Popovici M, Chavkin C 2003. κ Opioid receptor antagonism and prodynorphin gene disruption block stress-induced behavioral responses. J. Neurosci. 23:5674–83
    [Google Scholar]
  167. 167. 
    McLaughlin JP, Land BB, Li S, Pintar JE, Chavkin C 2006. Prior activation of kappa opioid receptors by U50,488 mimics repeated forced swim stress to potentiate cocaine place preference conditioning. Neuropsychopharmacology 31:787–94
    [Google Scholar]
  168. 168. 
    McLaughlin JP, Li S, Valdez J, Chavkin TA, Chavkin C 2006. Social defeat stress-induced behavioral responses are mediated by the endogenous kappa opioid system. Neuropsychopharmacology 31:1241–48
    [Google Scholar]
  169. 169. 
    Wells AM, Ridener E, Bourbonais CA, Kim W, Pantazopoulos H et al. 2017. Effects of chronic social defeat stress on sleep and circadian rhythms are mitigated by kappa-opioid receptor antagonism. J. Neurosci. 37:7656–68
    [Google Scholar]
  170. 170. 
    Rosa SG, Pesarico AP, Nogueira CW 2018. m-Trifluoromethyl-diphenyl diselenide promotes resilience to social avoidance induced by social defeat stress in mice: contribution of opioid receptors and MAPKs. Prog. Neuropsychopharmacol. Biol. Psychiatry 82:123–35
    [Google Scholar]
  171. 171. 
    Peters MF, Zacco A, Gordon J, Maciag CM, Litwin LC et al. 2011. Identification of short-acting κ-opioid receptor antagonists with anxiolytic-like activity. Eur. J. Pharmacol. 661:27–34
    [Google Scholar]
  172. 172. 
    Al-Hasani R, Bruchas MR. 2011. Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology 115:1363–81
    [Google Scholar]
  173. 173. 
    Vergura R, Balboni G, Spagnolo B, Gavioli E, Lambert DG et al. 2008. Anxiolytic- and antidepressant-like activities of H-Dmt-Tic-NH-CH(CH2-COOH)-Bid (UFP-512), a novel selective delta opioid receptor agonist. Peptides 29:93–103
    [Google Scholar]
  174. 174. 
    Richards EM, Mathews DC, Luckenbaugh DA, Ionescu DF, Machado-Vieira R et al. 2016. A randomized, placebo-controlled pilot trial of the delta opioid receptor agonist AZD2327 in anxious depression. Psychopharmacology 233:1119–30
    [Google Scholar]
  175. 175. 
    Bailey SJ, Husbands SM. 2018. Targeting opioid receptor signaling in depression: Do we need selective κ opioid receptor antagonists?. Neuronal Signal 2:NS20170145
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-010919-023317
Loading
/content/journals/10.1146/annurev-pharmtox-010919-023317
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error