Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-vfjqv Total loading time: 0 Render date: 2024-04-26T03:27:57.724Z Has data issue: false hasContentIssue false

Section 3 - Neurobiology

Published online by Cambridge University Press:  19 October 2021

Katherine D. Warburton
Affiliation:
University of California, Davis
Stephen M. Stahl
Affiliation:
University of California, San Diego
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2016

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Warburton, K. The new mission of forensic mental health systems: managing violence as a medial syndrome in an environment that balances treatment and safety. CNS Spectr. 2014 Oct; 19(5): 368373. DOI: 10.1017/S109285291400025X.Google Scholar
Stahl, SM, Morrissette, DA, Cummings, M, et al. The California Department of State Hospitals Violence Assessment and Treatment (CAL-VAT) guidelines. CNS Spectr. 2014; 19(5): 357365. DOI: 10.1017/S109285291400376.Google Scholar
Stahl, SM, Morrissette, DA, Citrome, L, et al. “Metaguidelines” for the management of patients with schizophrenia. CNS Spectr. 2013; 18(3): 150162.Google Scholar
Correll, CU. From receptor pharmacology to improved outcomes: individualising the selection, dosing, and switching of antipsychotics. Eur. Psychiatry. 2010; 25(Suppl. 2): S1221.Google Scholar
Davis, JM, Chen, N. Dose response and dose equivalence of antipsychotics. J. Clin. Psychopharmacol. 2004; 24(2): 192208.Google Scholar
Krakowski, MI, Kunz, M, Czobor, P, Volavka, J. Long-term high-dose neuroleptic treatment: who gets it and why? Hosp. Community Psychiatry. 1993; 44(7): 640644.Google ScholarPubMed
Comai, S, Tau, M, Gobbi, G. The psychopharmacology of aggressive behavior: a translational approach: part 1:neurobiology. J. Clin. Psychopharmacol. 2012; 32(1): 8394.Google Scholar
Comai, S, Tau, M, Pavlovic, Z, Gobbi, G. The psychopharmacology of aggressive behavior: a translational approach: part 2: clinical studies using atypical antipsychotics, anticonvulsants, and lithium. J. Clin. Psychopharmacol. 2012; 32(2): 237260.Google Scholar
Citrome, L, Volavka, J. The psychopharmacology of violence: making sensible decisions. CNS Spectr. 2014 Oct; 19(5): 411418. DOI: 10.1017/S109285291400054.Google Scholar
Volavka, J, Czobor, P, Citrome, L, Van Dorn, RA. Effectiveness of antipsychotic drugs against hostility with schizophrenia in the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) study. CNS Spectr. 2014; 0(5): 374381. DOI: 10.1017/S1092852913000849.CrossRefGoogle Scholar
Meyer, JM. A rational approach to employing high plasma levels of antipsychotics for violence associated with schizophrenia: case vignettes. CNS Spectr. 2014 Oct; 0(5): 432438. DOI: 10.1017/S10928514000236.Google Scholar
Morrissette, DA, Stahl, SM. Treating the violent patient with psychosis or impulsivity utilizing antipsychotic polypharmacy and high-dose monotherapy. CNS Spectr. 2014; 19(5). DOI: 10.1017/S1092852914000388.Google Scholar
Stahl, SM. Emerging guidelines for the use of antipsychotic polypharmacy. Rev. Psiquiatr. Salud. Ment. 2013; 6(3): 97100.Google Scholar
Stahl, SM. Antipsychotic polypharmacy: never say never, but never say always. Acta Psychiatr. Scand. 2012; 125(5): 349351.Google Scholar
Morrissette, DA, Stahl, SM. Should high dose or very long-term antipsychotic monotherapy be considered before antipsychotic polypharmacy? In Ritsner, MS, ed. Polypharmacy in Psychiatric Practice, Volume 1: Multiple Medication Use Strategies. Heidelberg: Springer; 2013: 107125.Google Scholar
Volavka, J, Citrome, L. Pathways to aggression in schizophrenia affect results of treatment. Schizophr. Bull. 2011; 37(5): 921929.CrossRefGoogle ScholarPubMed
Volavka, J, Citrome, L. Heterogeneity of violence in schizophrenia and implications for long-term treatment. Int. J. Clin. Pract. 2008; 62(8): 12371245.Google Scholar
Frogley, C, Taylor, D, Dickens, G, Picchioni, M. A systematic review of the evidence of clozapine’s antiaggressive effects. Int. J. Neuropsychopharmacol. 2012; 15(9): 13511371.Google Scholar
Stilwell, EN, Yates, SE, Brahm, NC. Violence among persons diagnosed with schizophrenia: how pharmacists can help. Res. Social Adm. Pharm. 2011; 7(4): 421429.Google Scholar
Swanson, JW, Swartz, MS, Van Dorn, RA, et al. Comparison of antipsychotic medication effects on reducing violence in people with schizophrenia. Br. J .Psychiatry. 2008; 193(1): 3743.Google Scholar
Volavka, J, Czobor, P, Nolan, K, et al. Overt aggression and psychotic symptoms in patients with schizophrenia treated with clozapine, olanzapine, risperidone, or haloperidol. J. Clin. Psychopharmacol. 2004; 24(2): 225228.Google Scholar
Topiwala, A, Fazel, S. The pharmacological management of violence in schizophrenia: a structured review. Expert Rev. Neurother. 2011; 11(1): 5363.Google Scholar
Bourget, D, Labelle, A. Managing pathologic aggression in people with psychotic disorders. J. Psychiatry Neurosci. 2012; 37(2): E34.Google Scholar
Citrome, L, Volavka, J. Pharmacological management of acute and persistent aggression in forensic psychiatry settings. CNS Drugs. 2011; 25(12): 10091021.Google Scholar
Roh, D, Chang, JG, Kim, CH, et al. Antipsychotic polypharmacy and high-dose prescription in schizophrenia: a 5-year comparison. Aust. N. Z. J. Psychiatry. 2014; 48(1): 5260.Google Scholar
Barnes, TR, Paton, C. Antipsychotic polypharmacy in schizophrenia: benefits and risks. CNS Drugs. 2011; 25(5): 383399.Google Scholar
Fleischhacker, WW, Uchida, H. Critical review of antipsychotic polypharmacy in the treatment of schizophrenia. Int. J. Neuropsychopharmacol. Jul; 17(7): 1083–1093.Google Scholar
Fujita, J, Nishida, A, Sakata, M, Noda, T, Ito, H. Excessive dosing and polypharmacy of antipsychotics caused by pro re nata in agitated patients with schizophrenia. Psychiatry Clin. Neurosci. 2013; 67(5): 345351.Google Scholar
Gallego, JA, Bonetti, J, Zhang, J, Kane, JM, Correll, CU. Prevalence and correlates of antipsychotic polypharmacy: a systematic review and meta-regression of global and regional trends from the 1970s to 2009. Schizophr. Res. 2012; 138(1): 1828.Google Scholar
Lochmann van Bennekom, MW, Gijsman, HJ, Zitman, FG. Antipsychotic polypharmacy in psychotic disorders: a critical review of neurobiology, efficacy, tolerability and cost effectiveness. J. Psychopharmacol. 2013; 27(4): 327336.Google Scholar
Langle, G, Steinert, T, Weiser, P, et al. Effects of polypharmacy on outcome in patients with schizophrenia in routine psychiatric treatment. Acta Psychiatr. Scand. 2012; 125(5): 372381.CrossRefGoogle ScholarPubMed
Essock, SM, Schooler, NR, Stroup, TS, et al. Effectiveness of switching from antipsychotic polypharmacy to monotherapy. Am. J. Psychiatry. 2011; 168(7): 702708.Google Scholar
Suzuki, T, Uchida, H, Tanaka, KF, et al. Revising polypharmacy to a single antipsychotic regimen for patients with chronic schizophrenia. Int. J. Neuropsychopharmacol. 2004; 7(2): 133142.CrossRefGoogle ScholarPubMed
Stahl, SM, Grady, MM. A critical review of atypical antipsychotic utilization: comparing monotherapy with polypharmacy and augmentation. Curr. Med .Chem. 2004; 11(3): 313327.Google Scholar
Aggarwal, NK, Sernyak, MJ, Rosenheck, RA. Prevalence of concomitant oral antipsychotic drug use among patients treated with long-acting, intramuscular, antipsychotic medications. J. Clin. Psychopharmacol. 2012; 32(3): 323328.Google Scholar
Stahl, SM. The last Diagnostic and Statistical Manual (DSM): replacing our symptom-based diagnoses with a brain circuit-based classification of mental illnesses. CNS Spectr. 2013; 18(2): 6568.Google Scholar
Singh, JP, Serper, M, Reinharth, J, Fazel, S. Structured assessment of violence risk in schizophrenia and other psychiatric disorders: a systematic review of the validity, reliability, and item content of 10 available instruments. Schizophr. Bull. 2011; 37(5): 899912.Google Scholar
Nolan, KA, Czobor, P, Roy, BB, et al. Characteristics of assaultive behavior among psychiatric inpatients. Psychiatr. Serv. 2003; 54(7): 10121016.Google Scholar
Abderhalden, C, Needham, I, Dassen, T, et al. Predicting inpatient violence using an extended version of the Brøset-Violence-Checklist: instrument development and clinical application. BMC Psychiatry. 2006; 6: 17.CrossRefGoogle ScholarPubMed
Quanbeck, CD, McDermott, BE, Lam, J, et al. Categorization of aggressive acts committed by chronically assaultive state hospital patients. Psychiatr. Serv. 2007; 58(4): 521528.Google Scholar
McDermott, BE, Holoyda, BJ. Assessment of aggression in inpatient settings. CNS Spectr. 2014 Oct; 0(5): 425431. DOI: 10.1017/S1092852914000224.Google Scholar
Monahan, J, Skeem, JL. The evolution of violence risk assessment. CNS Spectr. 2014 Oct; 0(5): 419424. DOI: 10.1017/S1092852914000145.Google Scholar
Stahl, SM, Morrissette, DA. Stahl’s Illustrated: Violence: Neural Circuits, Genetics and Treatment. Cambridge, UK: Cambridge University Press; 2014.Google Scholar
Wehring, HJ, Carpenter, WT. Violence and schizophrenia. Schizophr. Bull. 2011; 37(5): 877878.Google Scholar
Song, H, Min, SK. Aggressive behavior model in schizophrenic patients. Psychiatry Res. 2009; 167(1–2): 5865.Google Scholar
Serper, M, Beech, DR, Harvey, PD, Dill, C. Neuropsychological and symptom predictors of aggression on the psychiatric inpatient service. J. Clin. Exp. Neuropsychol. 2008; 30(6): 700709.Google Scholar
Stahl, SM. Stahl’s Essential Psychopharmacology. 4th edn. New York: Cambridge University Press; 2013.Google Scholar
Siever, LJ. Neurobiology of aggression and violence. Am. J. Psychiatry. 2008; 165(4): 429442.Google Scholar
Nord, M, Farde, L. Antipsychotic occupancy of dopamine receptors in schizophrenia. CNS Neurosci. Ther. 2011; 17(2): 97103.Google Scholar
Mauri, MC, Volonteri, LS, Colasanti, A, et al. Clinical pharmacokinetics of atypical antipsychotics: a critical review of the relationship between plasma concentrations and clinical response. Clin. Pharmacokinet. 2007; 46(5): 359388.Google Scholar
Coccaro, EF, Sripada, CS, Yanowitch, RN, Phan, KL. Corticolimbic function in impulsive aggressive behavior. Biol. Psychiatry. 2011; 69(12): 11531159.Google Scholar
Coccaro, EF, McCloskey, MS, Fitzgerald, DA, Phan, KL. Amygdala and orbitofrontal reactivity to social threat in individuals with impulsive aggression. Biol. Psychiatry. 2007; 62(2): 168178.Google Scholar
Hoptman, MJ, Antonius, D. Neuroimaging correlates of aggression in schizophrenia: an update. Curr. Opin. Psychiatry. 2011; 24(2): 100106.Google Scholar
De Sanctis, P, Foxe, JJ, Czobor, P, et al. Early sensory-perceptual processing deficits for affectively valenced inputs are more pronounced in schizophrenia patients with a history of violence than in their non-violent peers. Soc. Cogn. Affect Neurosci. 2013; 8(6): 678687.CrossRefGoogle ScholarPubMed
Kumari, V, Barkataki, I, Goswami, S, et al. Dysfunctional, but not functional, impulsivity is associated with a history of seriously violent behaviour and reduced orbitofrontal and hippocampal volumes in schizophrenia. Psychiatry Res. 2009; 173(1): 3944.Google Scholar
Coccaro, EF. Intermittent explosive disorder as a disorder of impulsive aggression for DSM-5. Am. J. Psychiatry. 2012; 169(6): 577588.Google Scholar
Haller, J. The neurobiology of abnormal manifestations of aggression—a review of hypothalamic mechanisms in cats, rodents, and humans. Brain Res. Bull. 2013; 93: 97109.Google Scholar
Kumari, V, Aasen, I, Taylor, P, et al. Neural dysfunction and violence in schizophrenia: an fMRI investigation. Schizophr. Res. 2006; 84(1): 144164.Google Scholar
Krakowski, MI, Czobor, P, Nolan, KA. Atypical antipsychotics, neurocognitive deficits, and aggression in schizophrenic patients. J. Clin. Psychopharmacol. 2008; 28(5): 485493.Google Scholar
Krakowski, MI, Czobor, P. Executive function predicts response to antiaggression treatment in schizophrenia: a randomized controlled trial. J. Clin. Psychiatry. 2012; 73(1): 7480.Google Scholar
Elie, D, Poirier, M, Chianetta, J, et al. Cognitive effects of antipsychotic dosage and polypharmacy: a study with the BACS in patients with schizophrenia and schizoaffective disorder. J. Psychopharmacol. 2010; 24(7): 10371044.CrossRefGoogle ScholarPubMed
Singh, JP, Volavka, J, Czobor, P, Van Dorn, RA. A metaanalysis of the Val158Met COMT polymorphism and violent behavior in schizophrenia. PloS One. 2012; 7(8): e43423.Google Scholar
Pavlov, KA, Chistiakov, DA, Chekhonin, VP. Genetic determinants of aggression and impulsivity in humans. J. Appl. Genet. 2012; 53(1): 6182.Google Scholar
Rosell, DR, Thompson, JL, Slifstein, M, et al. Increased serotonin 2A receptor availability in the orbitofrontal cortex of physically aggressive personality disordered patients. Biol. Psychiatry. 2010; 67(12): 11541162.Google Scholar
Winstanley, CA, Theobald, DE, Dalley, JW, Glennon, JC, Robbins, TW. 5-HT2A and 5-HT2C receptor antagonists have opposing effects on a measure of impulsivity: interactions with global 5-HT depletion. Psychopharmacology (Berl). 2004; 176(3–4): 376385.Google Scholar

References

Tateno, A, Jorge, RE, Robinson, RG. Clinical correlates of aggressive behavior after traumatic brain injury. J. Neuropsychiatry Clin. Neurosci. 2003; 15(2): 155160.Google Scholar
Pardini, M, Krueger, F, Hodgkinson, C, et al. Prefrontal cortex lesions and MAO-A modulate aggression in penetrating traumatic brain injury. Neurology. 2011; 76(12): 10381045.Google Scholar
Comai, S, Tau, M, Gobbi, G. The psychopharmacology of aggressive behavior: a translational approach: part 1: neurobiology. J. Clin. Psychopharmacol. 2012; 32(1): 8394.Google Scholar
Goedhard, LE, Stolker, JJ, Heerdink, ER, et al. Pharmacotherapy for the treatment of aggressive behavior in general adult psychiatry: a systematic review. J. Clin. Psychiatry. 2006; 67(7): 10131024.Google Scholar
Tidey, JW, Miczek, KA. Effects of SKF 38393 and quinpirole on aggressive, motor and schedule-controlled behaviors in mice. Behav. Pharmacol. 1992; 3(6): 553565.Google Scholar
Rodríguez-Arias, M, Miňarro, J, Aguilar, MA, Pinazo, J, Simón, VM. Effects of risperidone and SCH 23390 on isolation-induced aggression in male mice. Eur. Neuropsychopharmacol. 1998; 8(2): 95103.Google Scholar
Aguilar, MA, Miňarro, J, Pérez-Iranzo, N, Simon, VM. Behavioral profile of raclopride in agonistic encounters between male mice. Pharmacol. Biochem. Behav. 1994; 47(3): 753756.Google Scholar
Miczek, KA, Tidey, JW. Amphetamines: aggressive and social behavior. In: Asghar, K, De Souza, E, eds. Pharmacology and Toxicology of Amphetamine and Related Designer Drugs. Washington, DC: National Institute on Drug Abuse; 1989: 68Y100.Google Scholar
Blader, JC, Pliszka, SR, Jensen, PS, Schooler, NR, Kafantaris, V. Stimulant-responsive and stimulant-refractory aggressive behavior among children with ADHD. Pediatrics. 2010; 126(4): 796806.Google Scholar
Huey, ED, Putnam, KT, Grafman, J. A systematic review of neurotransmitter deficits and treatments in frontotemporal dementia. Neurology. 2006; 66(1): 1722.Google Scholar
Huey, ED, Garcia, C, Wassermann, EM, Tierney, MC, Grafman, J. Stimulant treatment of frontotemporal dementia in 8 patients. J. Clin. Psychiatry. 2008; 69(12): 19811982.Google Scholar
Ko, JH, Monchi, O, Ptito, A, et al. Theta burst stimulation-induced inhibition of dorsolateral prefrontal cortex reveals hemispheric asymmetry in striatal dopamine release during a set-shifting task: a TMS-[(11)C]raclopride PET study. Eur. J. Neurosci. 2008; 28(10): 21472155.Google Scholar
Strafella, AP, Paus, T, Barrett, J, Dagher, A. Repetitive transcranial magnetic stimulation of the human prefrontal cortex induces dopamine release in the caudate nucleus. J. Neurosci. 2001; 21(15): RC157.CrossRefGoogle ScholarPubMed
Jaskiw, GE, Karoum, FK, Weinberger, DR. Persistent elevations in dopamine and its metabolites in the nucleus accumbens after mild subchronic stress in rats with ibotenic acid lesions of the medial prefrontal cortex. Brain Res. 1990; 534(1–2): 321323.Google Scholar
Seeman, P, Van Tol, HH. Dopamine receptor pharmacology. Trends Pharmacol. Sci. 1994; 15(7): 264270.Google Scholar
Volavka, J, Bilder, R, Nolan, K. Catecholamines and aggression: the role of COMT and MAO polymorphisms. Ann. N. Y. Acad. Sci. 2004; 1036: 393398.Google Scholar
Cohen, MX, Krohn-Grimberghe, A, Elger, CE, Weber, B. Dopamine gene predicts the brain’s response to dopaminergic drug. Eur. J. Neurosci. 2007; 26(12): 36523660.Google Scholar
Raymont, V, Greathouse, A, Reding, K, et al. Demographic, structural and genetic predictors of late cognitive decline after penetrating head injury. Brain. 2008; 131(Pt 2): 543558.Google Scholar
Cummings, JL, Mega, M, Gray, K, et al. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology. 1994; 44(12): 23082314.Google Scholar
Bremner, JD, Vermetten, E, Mazure, CM. Development and preliminary psychometric properties of an instrument for the measurement of childhood trauma: the Early Trauma Inventory. Depress. Anxiety. 2000; 12(1): 112.Google Scholar
Solomon, J, Raymont, V, Braun, A, Butman, JA, Grafman, J. User-friendly software for the analysis of brain lesions (ABLe). Comput. Methods Programs Biomed. 2007; 86(3): 245254.Google Scholar
Tzourio-Mazoyer, N, Landeau, B, Papathanassiou, D, et al. Automated anatomical labeling of activations in SPM using a macroscopic anatomical parcellation of the MNI MRI single-subject brain. Neuroimage. 2002; 15(1): 273289.Google Scholar
Koenigs, M, Huey, ED, Calamia, M, et al. Distinct regions of prefrontal cortex mediate resistance and vulnerability to depression. J. Neurosci. 2008; 28(47): 12,34112,348.Google Scholar
Hodgkinson, CA, Yuan, Q, Xu, K, et al. Addictions biology: haplotype-based analysis for 130 candidate genes on a single array. Alcohol Alcohol. 2008; 43(5): 505515.Google Scholar
Huang, W, Ma, JZ, Payne, TJ, et al. Significant association of DRD1 with nicotine dependence. Hum. Genet. 2008; 123(2): 133140.Google Scholar
Fukui, N, Suzuki, Y, Sugai, T, et al. Exploring functional polymorphisms in the dopamine receptor D2 gene using prolactin concentration in healthy subjects. Mol. Psychiatry. 2010; 16(4): 356358.Google Scholar
Hamidovic, A, Dlugos, A, Skol, A, Palmer, AA, de Wit, H. Evaluation of genetic variability in the dopamine receptor D2 in relation to behavioral inhibition and impulsivity/sensation seeking: an exploratory study with d-amphetamine in healthy participants. Exp, Clin. Psychopharmacol. 2009; 17(6): 374383.Google Scholar
Shih, JC, Chen, K, Ridd, MJ. Monoamine oxidase: from genes to behavior. Annu. Rev. Neurosci. 1999; 22(1): 197217.Google Scholar
Vijayraghavan, S, Wang, M, Birnbaum, SG, Williams, GV, Arnsten, AF. Inverted-U dopamine D1 receptor actions on prefrontal neurons engaged in working memory. Nat. Neurosci. 2007; 10(3): 376384.Google Scholar
Floresco, SB. Prefrontal dopamine and behavioral flexibility: shifting from an “inverted-U” toward a family of functions. Front. Neurosci. 2013; 7: 62.Google Scholar
Cools, R, Frank, MJ, Gibbs, SE, et al. Striatal dopamine predicts outcome-specific reversal learning and its sensitivity to dopaminergic drug administration. J. Neurosci. 2009; 29(5): 15381543.Google Scholar
Knutson, B, Gibbs, SE. Linking nucleus accumbens dopamine and blood oxygenation. Psychopharmacology (Berl.). 2007; 191(3): 813822.Google Scholar
Scheres, A, Milham, MP, Knutson, B, Castellanos, FX. Ventral striatal hyporesponsiveness during reward anticipation in attention-deficit/hyperactivity disorder. Biol .Psychiatry. 2007; 61(5): 720724.Google Scholar
Couppis, MH, Kennedy, CH. The rewarding effect of aggression is reduced by nucleus accumbens dopamine receptor antagonism in mice. Psychopharmacology (Berl.). 2008; 197(3): 449456.Google Scholar
Zamboni, G, Huey, ED, Krueger, F, Nichelli, PF, Grafman, J. Apathy and disinhibition in frontotemporal dementia: insights into their neural correlates. Neurology. 2008; 71(10): 736742.Google Scholar
Takahata, R, Moghaddam, B. Target-specific glutamatergic regulation of dopamine neurons in the ventral tegmental area. J. Neurochem. 2000; 75(4): 17751778.Google Scholar
Diekhof, EK, Nerenberg, L, Falkai, P, et al. Impulsive personality and the ability to resist immediate reward: an fMRI study examining interindividual differences in the neural mechanisms underlying self-control. Hum. Brain Mapp. 2012; 33(12): 27682784.Google Scholar
Buckholtz, JW, Treadway, MT, Cowan, RL, et al. Mesolimbic dopamine reward system hypersensitivity in individuals with psychopathic traits. Nat.. Neurosci. 2010; 13(4): 419421.Google Scholar
Plichta, MM, Scheres, A. Ventral-striatal responsiveness during reward anticipation in ADHD and its relation to trait impulsivity in the healthy population: a meta-analytic review of the fMRI literature. Neurosci. Biobehav. Rev. 2014; 38: 125134.Google Scholar
Wong, TM. Brain injury and aggression: can we get some help? Neurology. 2011; 76(12): 10321033.Google Scholar
Fowler, SB, Hertzog, J, Wagner, BK. Pharmacological interventions for agitation in head-injured patients in the acute care setting. J. Neurosci. Nurs. 1995; 27(2): 119123.Google Scholar
Kline, AE, Massucci, JL, Zafonte, RD, et al. Differential effects of single versus multiple administrations of haloperidol and risperidone on functional outcome after experimental brain trauma. Crit. Care Med. 2007; 35(3): 919924.Google Scholar
Hoffman, AN, Cheng, JP, Zafonte, RD, Kline, AE. Administration of haloperidol and risperidone after neurobehavioral testing hinders the recovery of traumatic brain injury-induced deficits. Life Sci. 2008; 83(17–18): 602607.Google Scholar
de Almeida, J, Palacios, JM, Mengod, G. Distribution of 5-HT and DA receptors in primate prefrontal cortex: implications for pathophysiology and treatment. Prog. Brain Res. 2008; 172: 101115.Google Scholar
DeYoung, CG, Peterson, JB, Séguin, JR, et al. The dopamine D4 receptor gene and moderation of the association between externalizing behavior and IQ. Arch. Gen. Psychiatry. 2006; 63(12): 14101416.Google Scholar
Bakermans-Kranenburg, MJ, van Ijzendoorn, MH, Caspers, K, Philibert, R. DRD4 genotype moderates the impact of parental problems on unresolved loss or trauma. Attach. Hum. Dev. 2011; 13(3): 253269.Google Scholar

References

Nolan, KA, Czobor, P, Roy, BB, et al. Characteristics of assaultive behavior among psychiatric inpatients. Psychiatr. Serv. 2003; 54(7): 10121016.Google Scholar
Quanbeck, CD, McDermott, BE, Lam, J, et al. Categorization of aggressive acts committed by chronically assaultive state hospital patients. Psychiatr. Serv. 2007; 58(4): 521528.Google Scholar
Stahl, SM. Deconstructing violence as a medical syndrome: mapping psychotic, impulsive and predatory subtypes to malfunctioning brain circuits. CNS Spectr. 2014; 19(5): 357365.Google Scholar
Stahl, SM, Morrissette, DA. Stahl’s Illustrated: Violence: Neural Circuits, Genetics and Treatment. Cambridge, UK: Cambridge University Press; 2014.Google Scholar
Stahl, SM, Morrissette, DA, Cummings, M, et al. The California Department of State Hospitals Violence Assessment and Treatment (CAL-VAT) guidelines. CNS Spectr. 2014; 19(5): 449465.Google Scholar
Dardashti, L, O’Day, J, Barsom, M, Schwartz, E, Proctor, G. Illustrative cases to support Cal-VAT guidelines. CNS Spectr. 2015; 20(3): 311318. DOI: 10.1017/S1092852915000127.Google Scholar
Morrissette, DA, Stahl, SM. Treating the violent patient with psychosis or impulsivity utilizing antipsychotic polypharmacy and high-dose monotherapy. CNS Spectr. 2014; 19(5): 439448.Google Scholar
Coccaro, E, Fanning, J, Phan, KL, Lee, R. Serotonin and impulsive aggression. CNS Spectr. 2015; 20(3): 295302.Google Scholar
Rosell, D, Siever, L. The neurobiology of violence. CNS Spectr. 2015; 20(3): 126.Google Scholar
Grant, JE, Kim, SW. Brain circuitry of compulsivity and impulsivity. CNS Spectr. 2014; 19(1): 2127.Google Scholar
Fineberg, NA, Chamberlain, SR, Goudriaan, AE, et al. New developments in human neurocognition: clinical, genetic and brain imaging correlates of impulsivity and compulsivity. CNS Spectr. 2014; 19(1): 6989.Google Scholar
Berlin, GS, Hollander, E. Compulsivity, impulsivity, and the DSM 5 process. CNS Spectr. 2014; 19(1): 6268.Google Scholar
Stahl, SM, Grady, M. Stahl’s Illustrated: Substance Use and Impulsive Disorders. Cambridge, UK: Cambridge University Press; 2012.Google Scholar
Everitt, BJ, Robbins, TW. Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nat. Neurosci. 2005; 8(11): 14811489.Google Scholar
Everitt, BJ, Robbins, TW. From ventral to dorsal striatum: devolving views of their roles in drug addiction. Neurosci. Biobehav. Rev. 2013; 37(9 Pt A): 19461954.Google Scholar
Wyvell, CL, Berridge, KC. Intra-accumbens amphetamine increases the conditioned incentive salience of sucrose reward: enhancement of reward “wanting” without enhanced “liking” or response reinforcement. J. Neurosci. 2000; 20(21): 81228130.Google Scholar
Volkow, ND, Wang, GJ, Baler, RD. Reward, dopamine and the control of food intake: implications for obesity. Trends Cogn. Sci. 2011; 15(1): 3746.Google Scholar
Bartholomew, N, Morgan, R. Co-morbid mental illness and criminalness: implications for housing and treatment. CNS Spectr. 2015; 20(3): 110.Google Scholar
Felthous, A. The appropriateness of treating psychopathic disorders. CNS Spectr. 2015; 20(3): 182189.Google Scholar
Medalia, A, Opler, LA, Saperstein, AM. Integrating psychopharmacology and cognitive remediation to treat cognitive dysfunction in the psychotic disorders. CNS Spectr. 2014; 19(2): 115120.Google Scholar
Hooker, CI, Bruce, L, Fisher, M, et al. Neural activity during emotion recognition after combined cognitive plus social cognitive training in schizophrenia. Schizophr. Res. 2012; 139(1–3): 5359.Google Scholar
Stahl, SM. Stahl’s Essential Psychopharmacology. 4th edn. New York: Cambridge University Press; 2013.Google Scholar
McElroy, S, Hudson, JI, Mitchell, JE, et al. Efficacy and safety of lisdexamfetamine for treatment of adults with moderate to severe binge-eating disorder: a randomized clinical trial. JAMA Psychiatry. 2015; 72(3): 235246.Google Scholar

References

Compton, WM, Conway, KP, Stinson, FS, Colliver, JD, Grant, BF. Prevalence, correlates, and comorbidity of DSM-IV antisocial personality syndromes and alcohol and specific drug use disorders in the United States: results from the national epidemiologic survey on alcohol and related conditions. J. Clin. Psychiatry. 2005; 66(6): 677685.Google Scholar
Anderson, NE, Kiehl, KA. Psychopathy and aggression: when paralimbic dysfunction leads to violence. Curr. Top. Behav. Neurosci. 2014; 17: 369393.Google Scholar
Michaud, SG, Aynesworth, H. Ted Bundy: Conversations with a Killer. Irving, TX: Authorlink Press; 2000.Google Scholar
Macmillan, MB. An Odd Kind of Fame: Stories of Phineas Gage. Boston, MA: The MIT Press; 2002.Google Scholar
Harlow, JM. Passage of an iron rod through the head. Boston Medical and Surgical Journal. 1848; 39(20): 389393.Google Scholar
Bigelow, HJ. Dr. Harlow’s case of recovery from the passage of an iron bar through the head. Am. J. Med. Sci. 1850; 20(39): 1322.Google Scholar
Harlow, JM. Recovery from the Passage of an Iron Bar through the Head. Boston: David Clapp & Son; 1869.Google Scholar
Van Horn, JD, Irimia, A, Torgerson, CM, et al. Mapping connectivity damage in the case of Phineas Gage. PLoS ONE. 2012; 7(5): e37454.Google Scholar
Cleckley, HM. The Mask of Sanity: An Attempt to Clarify Some Issues about the So-Called Psychopathic Personality. 5th edn. Chicago: William A. Dolan; 1988.Google Scholar
Harpur, TJ, Hare, RD, Hakstain, AR. Two-factor conceptualization of psychopathy: construct validity and assessment implications. Psychol. Assess. 1989; 11(1): 617.Google Scholar
Cooke, DJ, Michie, C, Skeem, J. Understanding the structure of the psychopathy checklist–revised: an exploration of methodological confusion. Br. J. Psychiatry Suppl. 2007; 49: s39s50.Google Scholar
Raine, A. The Psychopathology of Crime: Criminal Behavior as a Clinical Disorder. 1st edn. San Diego, CA: Academic Press; 1997.Google Scholar
Raine, A. Autonomic nervous system factors underlying disinhibited, antisocial, and violent behavior: biosocial perspectives and treatment implications. Ann. N. Y. Acad. Sci. 1996; 794: 4659.Google Scholar
Thompson, DF, Ramos, CL, Willett, JK. Psychopathy: clinical features, developmental basis and therapeutic challenges. J. Clin. Pharm. Ther. 2014; 39(5): 485495.Google Scholar
Perry, BD, Pollard, R, Blakely, T, Baker, WL, Vigilante, D. Childhood trauma, the neurobiology of adaptation and “use-dependent” development of the brain: how “states” become “traits.” Infant Mental Health Journal. 1995; 16(4): 271291.Google Scholar
Byrd, AL, Manuck, SB. MAOA, childhood maltreatment, and antisocial behavior: meta-analysis of a gene-environment interaction. Biol. Psychiatry. 2014; 75(1): 917.Google Scholar
Haberstick, BC, Lessem, JM, Hewitt, JK, et al. MAOA genotype, childhood maltreatment, and their interaction in the etiology of adult antisocial behaviors. Biol. Psychiatry. 2014; 75(1): 2530.Google Scholar
Moul, C, Dobson-Stone, C, Brennan, J, Hawes, D, Dadds, M. An exploration of the serotonin system in antisocial boys with high levels of callous-unemotional traits. PLoS ONE. 2013; 8(2): e56619.Google Scholar
Kretschmer, T, Vitaro, F, Barker, ED. The association between peer and own aggression is moderated by the BDNF Val-Met polymorphism. J. Res. Adolesc. 2014; 24(1): 177185.Google Scholar
Smearman, EL, Winiarski, DA, Brennan, PA, Najman, J, Johnson, KC. Social stress and the oxytocin receptor gene interact to predict antisocial behavior in an at-risk cohort. Dev. Psychopathol. 2015; 27(1): 309318. DOI: 10.1017/S0954579414000649.Google Scholar
Frick, PJ. Extending the construct of psychopathy to youth: implications for understanding, diagnosing, and treating antisocial children and adolescents. Can. J. Psychiatry. 2009; 54(12): 803812.Google Scholar
Trebuchon, A, Bartolomei, F, McGonigal, A, Laguitton, V, Chauvel, P. Reversible antisocial behavior in ventromedial prefrontal lobe epilepsy. Epilepsy Behav. 2013; 29(2): 367373.Google Scholar
Venables, NC, Patrick, CJ. Reconciling discrepant findings for P3 brain response in criminal psychopathy through reference to the concept of externalizing proneness. Psychophysiology. 2014; 51(5): 427436.Google Scholar
Drislane, LE, Vaidyanathan, U, Patrick, CJ. Reduced cortical call to arms differentiates psychopathy from antisocial personality disorder. Psychol. Medicine. 2013; 43(4): 825835.Google Scholar
Yoder, KJ, Decety, J. Spatiotemporal neural dynamics of moral judgment: a high-density ERP study. Neuropsychologia. 2014; 60: 3945.Google Scholar
Hyde, LW, Byrd, AL, Votruba-Drzal, E, Hariri, AR, Manuck, SB. Amygdala reactivity and negative emotionality: divergent correlates of antisocial personality and psychopathy traits in a community sample. J. Abnorm. Psychol. 2014; 123(1): 214224.Google Scholar
Decety, J, Skelly, L, Yoder, KJ, Kiehl, KA. Neural processing of dynamic emotional facial expressions in psychopaths. Soc. Neurosci. 2014; 9(1): 3649.Google Scholar
Ermer, E, Cope, LM, Nyalakanti, PK, Calhoun, VD, Kiehl, KA. Aberrant paralimbic gray matter in criminal psychopathy. J. Abnorm. Psychol. 2012; 121(3): 649658.Google Scholar
Shenhav, A, Greene, JD. Integrative moral judgment: dissociating the roles of the amygdala and ventromedial prefrontal cortex. J. Neurosci. 2014; 34(13): 47414749.Google Scholar
Motzkin, JC, Newman, JP, Kiehl, KA, Koenigs, M. Reduced prefrontal connectivity in psychopathy. J. Neurosci. 2011; 31(48): 17,34817,357.Google Scholar
Contreras-Rodríguez, O, Pujol, J, Batalla, I, et al. Functional connectivity bias in the prefrontal cortex of psychopaths. Biol. Psychiatry. 2015; 78(9): 647655. DOI: 10.1016/j.biopsych.2014.03.007.Google Scholar
Li, W, Mai, X, Liu, C. The default mode network and social understanding of others: what do brain connectivity studies tell us? Front. Hum. Neurosci. 2014; 8: 74.Google Scholar
Raine, A, Lencz, T, Taylor, K, et al. Corpus callosum abnormalities in psychopathic antisocial individuals. Arch. Gen. Psychiatry. 2003; 60(11): 11341142.Google Scholar
Sundram, F, Deeley, Q, Sarkar, S, et al. White matter microstructural abnormalities in the frontal lobe of adults with antisocial personality disorder. Cortex. 2012; 48(2): 216229.Google Scholar
Gregory, S, Ffytche, D, Simmons, A, et al. The antisocial brain: psychopathy matters: a structural MRI investigation of antisocial male violent offenders. JAMA Psychiatry. 2012; 69(9): 962972.Google Scholar
Haker, H, Schimansky, J, Rossler, W. [Sociophysiology: basic processes of empathy]. Neuropsychiatrie. 2010; 24(3): 151160.Google Scholar
Cai, X, Padoa-Schioppa, C. Neuronal encoding of subjective value in dorsal and ventral anterior cingulate cortex. J. Neurosci. 2012; 32(11): 37913808.Google Scholar
Buckholtz, JW, Treadway, MT, Cowan, RL, et al. Mesolimbic dopamine reward system hypersensitivity in individuals with psychopathic traits. Nat. Neurosci. 2010; 13(4): 419421.Google Scholar
Hare, RD, Neumann, CS. Psychopathy as a clinical and empirical construct. Ann. Rev. Clin. Psychol. 2008; 4: 217246.Google Scholar
Coid, JW. Personality disorders in prisoners and their motivation for dangerous and disruptive behaviour. Crim. Behav. Ment. Health. 2002; 12(3): 209226.Google Scholar
Sobral, J, Luengo, A, Gómez-Fraguela, JA, Romero, E, Villar, P. [Personality, gender and violent criminality in prison inmates]. Psicothema. 2007; 19(2): 269275.Google Scholar
Endrass, J, Urbaniok, F, Gerth, J, Rossegger, A. [Prison violence: prevalence, manifestation and risk factors]. Praxis (Bern 1994). 2009; 98(22): 12791283.Google Scholar
Kennedy, HG. Therapeutic uses of security: mapping forensic mental health services by stratifying risk. Advances in Psychiatric Treatment. 2002; 8(6): 433443.Google Scholar
Meffert, H, Gazzola, V, den Boer, JA, Bartels, AA, Keysers, C. Reduced spontaneous but relatively normal deliberate vicarious representations in psychopathy. Brain. 2013; 136(8): 25502562.Google Scholar
Bonta, J, Andrews, DA. Risk-Need-Responsivity Model for Offender Assessment and Rehabilitation 2007–06. Ottawa: Her Majesty the Queen in Right of Canada; 2007.Google Scholar
Gitlin, MJ. Pharmacotherapy of personality disorders: conceptual framework and clinical strategies. J. Clin. Psychopharmacol. 1993; 13(5): 343353.Google Scholar
Citrome, L, Volavka, J. Pharmacological management of acute and persistent aggression in forensic psychiatry settings. CNS Drugs. 2011; 25(12): 10091021.Google Scholar
Ripoll, LH, Triebwasser, J, Siever, LJ. Evidence-based pharmacotherapy for personality disorders. Int. J. Neuropsychopharmacol. 2011; 14(9): 12571288.Google Scholar
Tupin, JP, Smith, DB, Clanon, TL, et al. The long-term use of lithium in aggressive prisoners. Compr. Psychiatry. 1973; 14(4): 311317.Google Scholar
Dunlop, BW, DeFife, JA, Marx, L, et al. The effects of sertraline on psychopathic traits. Int. Clin. Psychopharmacol. 2011; 26(6): 329337.Google Scholar
Brown, D, Larkin, F, Sengupta, S, et al. Clozapine: an effective treatment for seriously violent and psychopathic men with antisocial personality disorder in a UK high-security hospital. CNS Spectr. 2014; 19(5): 391402.Google Scholar
Bari, A, Niu, T, Langevin, JP, Fried, I. Limbic neuromodulation: implications for addiction, posttraumatic stress disorder, and memory. Neurosurg. Clin. N. Am. 2014; 25(1): 137145.Google Scholar

References

Hall, JE, Simon, TR, Lee, RD, Mercy, JA. Implications of direct protective factors for public health research and prevention strategies to reduce youth violence. Am. J. Prev. Med. 2012; 43(2 Suppl 1): S76S83.Google Scholar
Modi, MN, Palmer, S, Armstrong, A. The role of Violence Against Women Act in addressing intimate partner violence: a public health issue. J. Womens Health (Larchmt). 2014; 23(3): 253259.Google Scholar
Ramsay, SE, Bartley, A, Rodger, AJ. Determinants of assault-related violence in the community: potential for public health interventions in hospitals. Emerg. Med. J. 2014; 31: 986989.Google Scholar
Whitaker, S. Preventing violent conflict: a revised mandate for the public health professional? J. Public Health Policy. 2013; 34(1): 4654.Google Scholar
Bushman, BJ, Anderson, CA. Is it time to pull the plug on the hostile versus instrumental aggression dichotomy? Psychol. Rev. 2001; 108(1): 273279.Google Scholar
Baker, LA, Raine, A, Liu, J, Jacobson, KC. Differential genetic and environmental influences on reactive and proactive aggression in children. J. Abnorm. Child Psychol. 2008; 36(8): 12651278.Google Scholar
Bezdjian, S, Tuvblad, C, Raine, A, Baker, LA. The genetic and environmental covariation among psychopathic personality traits, and reactive and proactive aggression in childhood. Child Dev. 2011; 82(4): 12671281.Google Scholar
Cima, M, Raine, A, Meesters, C, Popma, A. Validation of the Dutch Reactive Proactive Questionnaire (RPQ): differential correlates of reactive and proactive aggression from childhood to adulthood. Aggress. Behav. 2013; 39(2): 99113.Google Scholar
Fossati, A, Raine, A, Borroni, S, et al. A cross-cultural study of the psychometric properties of the Reactive-Proactive Aggression Questionnaire among Italian nonclinical adolescents. Psychol. Assess. 2009; 21(1): 131135.Google Scholar
Fung, AL, Raine, A, Gao, Y. Cross-cultural generalizability of the Reactive-Proactive Aggression Questionnaire (RPQ). J. Pers. Assess. 2009; 91(5): 473479.Google Scholar
Raine, A, Dodge, K, Loeber, R, et al. The Reactive-Proactive Aggression Questionnaire: differential correlates of reactive and proactive aggression in adolescent boys. Aggress .Behav. 2006; 32(2): 159171.Google Scholar
Gardner, KJ, Archer, J, Jackson, S. Does maladaptive coping mediate the relationship between borderline personality traits and reactive and proactive aggression? Aggress. Behav. 2012; 38(5): 403413.Google Scholar
Lobbestael, J, Cima, M, Arntz, A. The relationship between adult reactive and proactive aggression, hostile interpretation bias, and antisocial personality disorder. J. Pers. Disord. 2013; 27(1): 5366.Google Scholar
Dodge, KA, Lochman, JE, Harnish, JD, Bates, JE, Pettit, GS. Reactive and proactive aggression in school children and psychiatrically impaired chronically assaultive youth. J. Abnorm. Psychol. 1997; 106(1): 3751.Google Scholar
Kolla, NJ, Malcolm, C, Attard, S, et al. Childhood maltreatment and aggressive behaviour in violent offenders with psychopathy. Can. J. Psychiatry. 2013; 58(8): 487494.Google Scholar
Arsenio, WF, Adams, E, Gold, J. Social information processing, moral reasoning, and emotion attributions: relations with adolescents’ reactive and proactive aggression. Child Dev. 2009; 80(6): 17391755.Google Scholar
Crick, NR, Dodge, KA. Social information-processing mechanisms in reactive and proactive aggression. Child Dev. 1996; 67(3): 9931002.Google Scholar
Dodge, KA, Coie, JD. Social-information-processing factors in reactive and proactive aggression in children’s peer groups. J. Pers. Soc. Psychol. 1987; 53(6): 11461158.Google Scholar
Hubbard, JA, Dodge, KA, Cillessen, AH, Coie, JD, Schwartz, D. The dyadic nature of social information processing in boys’ reactive and proactive aggression. J. Pers. Soc. Psychol. 2001; 80(2): 268280.Google Scholar
Smithmyer, CM, Hubbard, JA, Simons, RF. Proactive and reactive aggression in delinquent adolescents: relations to aggression outcome expectancies. J. Clin. Child Psychol. 2000; 29(1): 8693.Google Scholar
Walters, GD. Measuring proactive and reactive criminal thinking with the PICTS: correlations with outcome expectancies and hostile attribution biases. J. Interpers. Violence. 2007; 22(4): 371385.Google Scholar
Brugman, S, Lobbestael, J, Arntz, A, et al. Identifying cognitive predictors of reactive and proactive aggression. Aggress. Behav. 2014; 41: 5164. DOI: 10.1002/AB.21573.Google Scholar
Coccaro, EF, Kavoussi, RJ, Berman, ME, Lish, JD. Intermittent explosive disorder–revised: development, reliability, and validity of research criteria. Compr. Psychiatry. 1998; 39(6): 368376.Google Scholar
McCloskey, MS, Berman, ME, Noblett, KL, Coccaro, EF. Intermittent explosive disorder-integrated research diagnostic criteria: convergent and discriminant validity. J. Psychiatr. Res. 2006; 40(3): 231242.Google Scholar
Coccaro, EF. Intermittent explosive disorder: development of integrated research criteria for Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Compr. Psychiatry. 2011; 52(2): 119125.Google Scholar
Coccaro, EF, Lee, R, Kavoussi, RJ. Aggression, suicidality, and intermittent explosive disorder: serotonergic correlates in personality disorder and healthy control subjects. Neuropsychopharmacology. 2010; 35(2): 435444.Google Scholar
Salzman, CD, Fusi, S. Emotion, cognition, and mental state representation in amygdala and prefrontal cortex. Annu. Rev. Neurosci. 2010; 33: 173202.Google Scholar
Fernando, AB, Murray, JE, Milton, AL. The amygdala: securing pleasure and avoiding pain. Front. Behav. Neurosci. 2013; 7: 190.Google Scholar
Sah, P, Faber, ES, Lopez De, AM, Power, J. The amygdaloid complex: anatomy and physiology. Physiol. Rev. 2003; 83(3): 803834.Google Scholar
John, YJ, Bullock, D, Zikopoulos, B, Barbas, H. Anatomy and computational modeling of networks underlying cognitive-emotional interaction. Front. Hum. Neurosci. 2013; 7: 101.Google Scholar
Lee, S, Kim, SJ, Kwon, OB, Lee, JH, Kim, JH. Inhibitory networks of the amygdala for emotional memory. Front. Neural. Circuits. 2013; 7: 129.Google Scholar
Bzdok, D, Laird, AR, Zilles, K, Fox, PT, Eickhoff, SB. An investigation of the structural, connectional, and functional subspecialization in the human amygdala. Hum. Brain Mapp. 2013; 34(12): 32473266.Google Scholar
Matthies, S, Rusch, N, Weber, M, et al. Small amygdala-high aggression? The role of the amygdala in modulating aggression in healthy subjects. World J. Biol. Psychiatry. 2012; 13(1): 7581.Google Scholar
Pardini, DA, Raine, A, Erickson, K, Loeber, R. Lower amygdala volume in men is associated with childhood aggression, early psychopathic traits, and future violence. Biol. Psychiatry. 2014; 75(1): 7380.Google Scholar
Gopal, A, Clark, E, Allgair, A, et al. Dorsal/ventral parcellation of the amygdala: relevance to impulsivity and aggression. Psychiatry Res. 2013; 211(1): 2430.Google Scholar
Bobes, MA, Ostrosky, F, Diaz, K, et al. Linkage of functional and structural anomalies in the left amygdala of reactive-aggressive men. Soc. Cogn. Affect. Neurosci. 2013; 8(8): 928936.Google Scholar
Dyck, M, Loughead, J, Kellermann, T, et al. Cognitive versus automatic mechanisms of mood induction differentially activate left and right amygdala. Neuroimage. 2011; 54(3): 25032513.Google Scholar
New, AS, Hazlett, EA, Newmark, RE, et al. Laboratory induced aggression: a positron emission tomography study of aggressive individuals with borderline personality disorder. Biol. Psychiatry. 2009; 66(12): 11071114.Google Scholar
Coccaro, EF, McCloskey, MS, Fitzgerald, DA, Phan, KL. Amygdala and orbitofrontal reactivity to social threat in individuals with impulsive aggression. Biol. Psychiatry. 2007; 62(2): 168178.Google Scholar
Lozier, LM, Cardinale, EM, Vanmeter, JW, Marsh, AA. Mediation of the relationship between callous-unemotional traits and proactive aggression by amygdala response to fear among children with conduct problems. JAMA Psychiatry. 2014; 71(6): 627636.Google Scholar
Walton, ME, Croxson, PL, Behrens, TE, Kennerley, SW, Rushworth, MF. Adaptive decision making and value in the anterior cingulate cortex. Neuroimage. 2007; 36(Suppl 2): T142T154.Google Scholar
Rudebeck, PH, Murray, EA. The orbitofrontal oracle: cortical mechanisms for the prediction and evaluation of specific behavioral outcomes. Neuron. 2014; 84(6): 11431156.Google Scholar
Gansler, DA, McLaughlin, NC, Iguchi, L, et al. A multivariate approach to aggression and the orbital frontal cortex in psychiatric patients. Psychiatry Res. 2009; 171(3): 145154.Google Scholar
Antonucci, AS, Gansler, DA, Tan, S, et al. Orbitofrontal correlates of aggression and impulsivity in psychiatric patients. Psychiatry Res. 2006; 147(2–3): 213220.Google Scholar
Boes, AD, Tranel, D, Anderson, SW, Nopoulos, P. Right anterior cingulate: a neuroanatomical correlate of aggression and defiance in boys. Behav. Neurosci. 2008; 122(3): 677684.Google Scholar
Ducharme, S, Hudziak, JJ, Botteron, KN, et al. Right anterior cingulate cortical thickness and bilateral striatal volume correlate with child behavior checklist aggressive behavior scores in healthy children. Biol .Psychiatry. 2011; 70(3): 283290.Google Scholar
Soloff, PH, Meltzer, CC, Greer, PJ, Constantine, D, Kelly, TM. A fenfluramine-activated FDG-PET study of borderline personality disorder. Biol. Psychiatry. 2000; 47(6): 540547.Google Scholar
New, AS, Hazlett, EA, Buchsbaum, MS, et al. Blunted prefrontal cortical 18fluorodeoxyglucose positron emission tomography response to meta-chlorophenylpiperazine in impulsive aggression. Arch. Gen. Psychiatry. 2002; 59(7): 621629.Google Scholar
Ghashghaei, HT, Hilgetag, CC, Barbas, H. Sequence of information processing for emotions based on the anatomic dialogue between prefrontal cortex and amygdala. Neuroimage. 2007; 34(3): 905923.Google Scholar
Timbie, C, Barbas, H. Specialized pathways from the primate amygdala to posterior orbitofrontal cortex. J. Neurosci. 2014; 34(24): 81068118.Google Scholar
Ghashghaei, HT, Barbas, H. Pathways for emotion: interactions of prefrontal and anterior temporal pathways in the amygdala of the rhesus monkey. Neuroscience. 2002; 115(4): 12611279.Google Scholar
Roy, AK, Shehzad, Z, Margulies, DS, et al. Functional connectivity of the human amygdala using resting state fMRI. Neuroimage. 2009; 45(2): 614626.Google Scholar
Hoptman, MJ, D’Angelo, D, Catalano, D, et al. Amygdalofrontal functional disconnectivity and aggression in schizophrenia. Schizophr. Bull. 2010; 36(5): 10201028.Google Scholar
Fulwiler, CE, King, JA, Zhang, N. Amygdala-orbitofrontal restingstate functional connectivity is associated with trait anger. Neuroreport. 2012; 23(10): 606610.Google Scholar
Beyer, F, Munte, TF, Wiechert, J, Heldmann, M, Kramer, UM. Trait aggressiveness is not related to structural connectivity between orbitofrontal cortex and amygdala. PLoS One. 2014; 9(6): e101105.Google Scholar
New, AS, Hazlett, EA, Buchsbaum, MS, et al. Amygdala-prefrontal disconnection in borderline personality disorder. Neuropsychopharmacology. 2007; 32(7): 16291640.Google Scholar
Hornboll, B, Macoveanu, J, Rowe, J, et al. Acute serotonin 2A receptor blocking alters the processing of fearful faces in the orbitofrontal cortex and amygdala. J. Psychopharmacol. 2013; 27(10): 903914.Google Scholar
Rosell, DR, Thompson, JL, Slifstein, M, et al. Increased serotonin 2A receptor availability in the orbitofrontal cortex of physically aggressive personality disordered patients. Biol. Psychiatry. 2010; 67(12): 11541162.Google Scholar
Passamonti, L, Crockett, MJ, Apergis-Schoute, AM, et al. Effects of acute tryptophan depletion on prefrontal-amygdala connectivity while viewing facial signals of aggression. Biol. Psychiatry. 2012; 71(1): 3643.Google Scholar
Pezawas, L, Meyer-Lindenberg, A, Drabant, EM, et al. 5-HTTLPR polymorphism impacts human cingulate-amygdala interactions: a genetic susceptibility mechanism for depression. Nat. Neurosci. 2005; 8(6): 828834.Google Scholar
Heinz, A, Braus, DF, Smolka, MN, et al. Amygdala-prefrontal coupling depends on a genetic variation of the serotonin transporter. Nat. Neurosci. 2005; 8(1): 2021.Google Scholar
Haber, SN. The primate basal ganglia: parallel and integrative networks. J. Chem. Neuroanat. 2003; 26(4): 317330.Google Scholar
Crockett, MJ, Apergis-Schoute, A, Herrmann, B, et al. Serotonin modulates striatal responses to fairness and retaliation in humans. J. Neurosci. 2013; 33(8): 35053513.Google Scholar
van de Giessen, E, Rosell, DR, Thompson, JL, et al. Serotonin transporter availability in impulsive aggressive personality disordered patients: a PET study with [(11)C]DASB. J. Psychiatr. Res. 2014; 58: 147154.Google Scholar
Malick, JB, Barnett, A. The role of serotonergic pathways in isolation-induced aggression in mice. Pharmacol. Biochem. Behav. 1976; 5(1): 5561.Google Scholar
Brown, GL, Goodwin, FK, Ballenger, JC, Goyer, PF, Major, LF. Aggression in humans correlates with cerebrospinal fluid amine metabolites. Psychiatry Res. 1979; 1(2): 131139.Google Scholar
Brown, GL, Ebert, MH, Goyer, PF, et al. Aggression, suicide, and serotonin: relationships to CSF amine metabolites. Am. J. Psychiatry. 1982; 139(6): 741746.Google Scholar
Brown, CS, Kent, TA, Bryant, SG, et al. Blood platelet uptake of serotonin in episodic aggression. Psychiatry Res. 1989; 27(1): 512.Google Scholar
Stoff, DM, Pollock, L, Vitiello, B, Behar, D, Bridger, WH. Reduction of (3H)-imipramine binding sites on platelets of conduct-disordered children. Neuropsychopharmacology. 1987; 1(1): 5562.Google Scholar
Coccaro, EF, Kavoussi, RJ, Hauger, RL. Physiological responses to d-fenfluramine and ipsapirone challenge correlate with indices of aggression in males with personality disorder. Int. Clin. Psychopharmacol. 1995; 10(3): 177179.Google Scholar
Coccaro, EF, Berman, ME, Kavoussi, RJ, Hauger, RL. Relationship of prolactin response to d-fenfluramine to behavioral and questionnaire assessments of aggression in personality-disordered men. Biol. Psychiatry. 1996; 40(3): 157164.Google Scholar
Coccaro, EF, Kavoussi, RJ, Cooper, TB, Hauger, RL. Central serotonin activity and aggression: inverse relationship with prolactin response to d-fenfluramine, but not CSF 5-HIAA concentration, in human subjects. Am. J. Psychiatry. 1997; 154(10): 14301435.Google Scholar
Coccaro, EF, Astill, JL, Herbert, JL, Schut, AG. Fluoxetine treatment of impulsive aggression in DSM-III-R personality disorder patients. J. Clin. Psychopharmacol. 1990; 10(5): 373375.Google Scholar
Coccaro, EF, Kavoussi, RJ. Fluoxetine and impulsive aggressive behavior in personality-disordered subjects. Arch. Gen. Psychiatry. 1997; 54(12): 10811088.Google Scholar
Kruesi, MJ, Rapoport, JL, Hamburger, S, et al. Cerebrospinal fluid monoamine metabolites, aggression, and impulsivity in disruptive behavior disorders of children and adolescents. Arch. Gen. Psychiatry. 1990; 47(5): 419426.Google Scholar
Coccaro, EF, Lee, R. Cerebrospinal fluid 5-hydroxyindolacetic acid and homovanillic acid: reciprocal relationships with impulsive aggression in human subjects. J. Neural. Transm. 2010; 117(2): 241248.Google Scholar
Coccaro, EF, Kavoussi, RJ, Hauger, RL, Cooper, TB, Ferris, CF. Cerebrospinal fluid vasopressin levels: correlates with aggression and serotonin function in personality-disordered subjects. Arch. Gen. Psychiatry. 1998; 55(8): 708714.Google Scholar
Goveas, JS, Csernansky, JG, Coccaro, EF. Platelet serotonin content correlates inversely with life history of aggression in personality-disordered subjects. Psychiatry Res. 2004; 126(1): 2332.Google Scholar
Coccaro, EF, Kavoussi, RJ, Sheline, YI, Berman, ME, Csernansky, JG. Impulsive aggression in personality disorder correlates with platelet 5-HT2A receptor binding. Neuropsychopharmacology. 1997; 16(3): 211216.Google Scholar
Marseille, R, Lee, R, Coccaro, EF. Inter-relationship between different platelet measures of 5-HT and their relationship to aggression in human subjects. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2012; 36(2): 277281.Google Scholar
Modai, I, Gibel, A, Rauchverger, B, et al. Paroxetine binding in aggressive schizophrenic patients. Psychiatry Res. 2000; 94(1): 7781.Google Scholar
Sarne, Y, Mandel, J, Goncalves, MH, et al. Imipramine binding to blood platelets and aggressive behavior in offenders, schizophrenics and normal volunteers. Neuropsychobiology. 1995; 31(3): 120124.Google Scholar
Coccaro, EF, Kavoussi, RJ, Hauger, RL. Serotonin function and antiaggressive response to fluoxetine: a pilot study. Biol. Psychiatry. 1997; 42(7): 546552.Google Scholar
Carpenter, LL, Anderson, GM, Pelton, GH, et al. Tryptophan depletion during continuous CSF sampling in healthy human subjects. Neuropsychopharmacology. 1998; 19(1): 2635.Google Scholar
Moreno, FA, McGavin, C, Malan, TP, et al. Tryptophan depletion selectively reduces CSF 5-HT metabolites in healthy young men: results from single lumbar puncture sampling technique. Int. J. Neuropsychopharmacol. 2000; 3(4): 277283.Google Scholar
Williams, WA, Shoaf, SE, Hommer, D, Rawlings, R, Linnoila, M. Effects of acute tryptophan depletion on plasma and cerebrospinal fluid tryptophan and 5-hydroxyindoleacetic acid in normal volunteers. J. Neurochem. 1999; 72(4): 16411647.Google Scholar
Bjork, JM, Dougherty, DM, Moeller, FG, Cherek, DR, Swann, AC. The effects of tryptophan depletion and loading on laboratory aggression in men: time course and a food-restricted control. Psychopharmacology (Berl). 1999; 142(1): 2430.Google Scholar
Bjork, JM, Dougherty, DM, Moeller, FG, Swann, AC. Differential behavioral effects of plasma tryptophan depletion and loading in aggressive and nonaggressive men. Neuropsychopharmacology. 2000; 22(4): 357369.Google Scholar
Kramer, UM, Riba, J, Richter, S, Munte, TF. An fMRI study on the role of serotonin in reactive aggression. PLoS One. 2011; 6(11): e27668.Google Scholar
Kotting, WF, Bubenzer, S, Helmbold, K, et al. Effects of tryptophan depletion on reactive aggression and aggressive decision-making in young people with ADHD. Acta Psychiatr. Scand. 2013; 128(2): 114123.Google Scholar
Stadler, C, Zepf, FD, Demisch, L, et al. Influence of rapid tryptophan depletion on laboratory-provoked aggression in children with ADHD. Neuropsychobiology. 2007; 56(2–3): 104110.Google Scholar
Zimmermann, M, Grabemann, M, Mette, C, et al. The effects of acute tryptophan depletion on reactive aggression in adults with attention-deficit/hyperactivity disorder (ADHD) and healthy controls. PLoS One. 2012; 7(3): e32023.Google Scholar
Fanning, JR, Berman, ME, Guillot, CR, Marsic, A, McCloskey, MS. Serotonin (5-HT) augmentation reduces provoked aggression associated with primary psychopathy traits. J. Pers. Disord. 2014; 28(3): 449461.Google Scholar
Berman, ME, McCloskey, MS, Fanning, JR, Schumacher, JA, Coccaro, EF. Serotonin augmentation reduces response to attack in aggressive individuals. Psychol. Sci. 2009; 20(6): 714720.Google Scholar
Rubia, K, Lee, F, Cleare, AJ, et al. Tryptophan depletion reduces right inferior prefrontal activation during response inhibition in fast, event-related fMRI. Psychopharmacology (Berl). 2005; 179(4): 791803.Google Scholar
Lee, RJ, Gill, A, Chen, B, McCloskey, M, Coccaro, EF. Modulation of central serotonin affects emotional information processing in impulsive aggressive personality disorder. J. Clin. Psychopharmacol. 2012; 32(3): 329335.Google Scholar
Grady, CL, Siebner, HR, Hornboll, B, et al. Acute pharmacologically induced shifts in serotonin availability abolish emotion-selective responses to negative face emotions in distinct brain networks. Eur. Neuropsychopharmacol. 2013; 23(5): 368378.Google Scholar
Zhang, X, Beaulieu, JM, Sotnikova, TD, Gainetdinov, RR, Caron, MG. Tryptophan hydroxylase-2 controls brain serotonin synthesis. Science. 2004; 305(5681): 217.Google Scholar
Osipova, DV, Kulikov, AV, Popova, NK. C1473G polymorphism in mouse tph2 gene is linked to tryptophan hydroxylase-2 activity in the brain, intermale aggression, and depressive-like behavior in the forced swim test. J. Neurosci. Res. 2009; 87(5): 11681174.Google Scholar
Takahashi, A, Shiroishi, T, Koide, T. Genetic mapping of escalated aggression in wild-derived mouse strain MSM/Ms: association with serotonin-related genes. Front. Neurosci. 2014; 8: 156.Google Scholar
Mosienko, V, Bert, B, Beis, D, et al. Exaggerated aggression and decreased anxiety in mice deficient in brain serotonin. Transl. Psychiatry. 2012; 2: e122.Google Scholar
Chen, GL, Novak, MA, Meyer, JS, et al. The effect of rearing experience and TPH2 genotype on HPA axis function and aggression in rhesus monkeys: a retrospective analysis. Horm. Behav. 2010; 57(2): 184191.Google Scholar
Yang, J, Lee, MS, Lee, SH, et al. Association between tryptophan hydroxylase 2 polymorphism and anger-related personality traits among young Korean women. Neuropsychobiology. 2010; 62(3): 158163.Google Scholar
Yoon, HK, Lee, HJ, Kim, L, Lee, MS, Ham, BJ. Impact of tryptophan hydroxylase 2 G-703T polymorphism on anger-related personality traits and orbitofrontal cortex. Behav. Brain Res. 2012; 231(1): 105110.Google Scholar
Gutknecht, L, Jacob, C, Strobel, A, et al. Tryptophan hydroxylase-2 gene variation influences personality traits and disorders related to emotional dysregulation. Int. J. Neuropsychopharmacol. 2007; 10(3): 309320.Google Scholar
Inoue, H, Yamasue, H, Tochigi, M, et al. Effect of tryptophan hydroxylase-2 gene variants on amygdalar and hippocampal volumes. Brain Res. 2010; 1331: 5157.Google Scholar
Perez-Rodriguez, MM, Weinstein, S, New, AS, et al. Tryptophan-hydroxylase 2 haplotype association with borderline personality disorder and aggression in a sample of patients with personality disorders and healthy controls. J. Psychiatr. Res. 2010; 44(15): 10751081.Google Scholar
Brown, SM, Peet, E, Manuck, SB, et al. A regulatory variant of the human tryptophan hydroxylase-2 gene biases amygdala reactivity. Mol. Psychiatry. 2005; 10(9): 884888.Google Scholar
Booij, L, Turecki, G, Leyton, M, et al. Tryptophan hydroxylase(2) gene polymorphisms predict brain serotonin synthesis in the orbitofrontal cortex in humans. Mol. Psychiatry. 2012; 17(8): 809817.Google Scholar
Heiming, RS, Monning, A, Jansen, F, et al. To attack, or not to attack? The role of serotonin transporter genotype in the display of maternal aggression. Behav. Brain Res. 2013; 242: 135141.Google Scholar
Holmes, A, Murphy, DL, Crawley, JN. Reduced aggression in mice lacking the serotonin transporter. Psychopharmacology (Berl.). 2002; 161(2): 160167.Google Scholar
Yu, Q, Teixeira, CM, Mahadevia, D, et al. Dopamine and serotonin signaling during two sensitive developmental periods differentially impact adult aggressive and affective behaviors in mice. Mol. Psychiatry. 2014; 19(6): 688698.Google Scholar
Kiryanova, V, Dyck, RH. Increased aggression, improved spatial memory, and reduced anxiety-like behaviour in adult male mice exposed to fluoxetine early in life. Dev. Neurosci. 2014; 36(5): 396408.Google Scholar
Heils, A, Teufel, A, Petri, S, et al. Allelic variation of human serotonin transporter gene expression. J. Neurochem. 1996; 66(6): 26212624.Google Scholar
May, ME, Lightfoot, DA, Srour, A, Kowalchuk, RK, Kennedy, CH. Association between serotonin transporter polymorphisms and problem behavior in adult males with intellectual disabilities. Brain Res. 2010; 1357: 97103.Google Scholar
Hallikainen, T, Saito, T, Lachman, HM, et al. Association between low activity serotonin transporter promoter genotype and early onset alcoholism with habitual impulsive violent behavior. Mol. Psychiatry. 1999; 4(4): 385388.Google Scholar
Haberstick, BC, Smolen, A, Hewitt, JK. Family-based association test of the 5HTTLPR and aggressive behavior in a general population sample of children. Biol. Psychiatry. 2006; 59(9): 836843.Google Scholar
Beitchman, JH, Baldassarra, L, Mik, H, et al. Serotonin transporter polymorphisms and persistent, pervasive childhood aggression. Am. J. Psychiatry. 2006; 163(6): 11031105.Google Scholar
Verona, E, Joiner, TE, Johnson, F, Bender, TW. Gender specific gene-environment interactions on laboratory-assessed aggression. Biol. Psychol. 2006; 71(1): 3341.Google Scholar
Conway, CC, Keenan-Miller, D, Hammen, C, et al. Coaction of stress and serotonin transporter genotype in predicting aggression at the transition to adulthood. J. Clin. Child Adolesc. Psychol. 2012; 41(1): 5363.Google Scholar
Reif, A, Rosler, M, Freitag, CM, et al. Nature and nurture predispose to violent behavior: serotonergic genes and adverse childhood environment. Neuropsychopharmacology. 2007; 32(11): 23752383.Google Scholar
Aluja, A, Garcia, LF, Blanch, A, De, LD, Fibla, J. Impulsive-disinhibited personality and serotonin transporter gene polymorphisms: association study in an inmate’s sample. J. Psychiatr. Res. 2009; 43(10): 906914.Google Scholar
Payer, DE, Nurmi, EL, Wilson, SA, McCracken, JT, London, ED. Effects of methamphetamine abuse and serotonin transporter gene variants on aggression and emotion-processing neurocircuitry. Transl. Psychiatry. 2012; 2: e80.Google Scholar
Philibert, R, Madan, A, Andersen, A, et al. Serotonin transporter mRNA levels are associated with the methylation of an upstream CpG island. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2007; 144B(1): 101105.Google Scholar
Koenen, KC, Uddin, M, Chang, SC, et al. SLC6A4 methylation modifies the effect of the number of traumatic events on risk for posttraumatic stress disorder. Depress. Anxiety. 2011; 28(8): 639647.Google Scholar
Zhao, J, Goldberg, J, Bremner, JD, Vaccarino, V. Association between promoter methylation of serotonin transporter gene and depressive symptoms: a monozygotic twin study. Psychosom. Med. 2013; 75(6): 523529.Google Scholar
Dannlowski, U, Kugel, H, Redlich, R, et al. Serotonin transporter gene methylation is associated with hippocampal gray matter volume. Hum. Brain Mapp. 2014; 35(11): 53565367.Google Scholar
Nikolova, YS, Koenen, KC, Galea, S, et al. Beyond genotype: serotonin transporter epigenetic modification predicts human brain function. Nat. Neurosci. 2014; 17(9): 11531155.Google Scholar
Wang, D, Szyf, M, Benkelfat, C, et al. Peripheral SLC6A4 DNA methylation is associated with in vivo measures of human brain serotonin synthesis and childhood physical aggression. PLoS One. 2012; 7(6): e39501.Google Scholar
Frankle, WG, Lombardo, I, New, AS, et al. Brain serotonin transporter distribution in subjects with impulsive aggressivity: a positron emission study with [11C]McN 5652. Am. J. Psychiatry. 2005; 162(5): 915923.Google Scholar
Bortolato, M, Chen, K, Shih, JC. Monoamine oxidase inactivation: from pathophysiology to therapeutics. Adv. Drug Deliv. Rev. 2008; 60(13–14): 15271533.Google Scholar
Brunner, HG, Nelen, M, Breakefield, XO, Ropers, HH, van Oost, BA. Abnormal behavior associated with a point mutation in the structural gene for monoamine oxidase A. Science. 1993; 262(5133): 578580.Google Scholar
Sabol, SZ, Hu, S, Hamer, D. A functional polymorphism in the monoamine oxidase A gene promoter. Hum. Genet. 1998; 103(3): 273279.Google Scholar
Kuepper, Y, Grant, P, Wielpuetz, C, Hennig, J. MAOA-uVNTR genotype predicts interindividual differences in experimental aggressiveness as a function of the degree of provocation. Behav. Brain Res. 2013; 247: 7378.Google Scholar
Manuck, SB, Flory, JD, Ferrell, RE, Mann, JJ, Muldoon, MF. A regulatory polymorphism of the monoamine oxidase-A gene may be associated with variability in aggression, impulsivity, and central nervous system serotonergic responsivity. Psychiatry Res. 2000; 95(1): 923.Google Scholar
Stetler, DA, Davis, C, Leavitt, K, et al. Association of low-activity MAOA allelic variants with violent crime in incarcerated offenders. J. Psychiatr. Res. 2014; 58: 6975.Google Scholar
Byrd, AL, Manuck, SB. MAOA, childhood maltreatment, and antisocial behavior: meta-analysis of a gene-environment interaction. Biol. Psychiatry. 2014; 75(1): 917.Google Scholar
Karere, GM, Kinnally, EL, Sanchez, JN, et al. What is an “adverse” environment? Interactions of rearing experiences and MAOA genotype in rhesus monkeys. Biol. Psychiatry. 2009; 65(9): 770777.Google Scholar
Newman, TK, Syagailo, YV, Barr, CS, et al. Monoamine oxidase A gene promoter variation and rearing experience influences aggressive behavior in rhesus monkeys. Biol. Psychiatry. 2005; 57(2): 167172.Google Scholar
Alia-Klein, N, Goldstein, RZ, Kriplani, A, et al. Brain monoamine oxidase A activity predicts trait aggression. J. Neurosci. 2008; 28(19): 50995104.Google Scholar
Cases, O, Seif, I, Grimsby, J, et al. Aggressive behavior and altered amounts of brain serotonin and norepinephrine in mice lacking MAOA. Science. 1995; 268(5218): 17631766.Google Scholar
Scott, AL, Bortolato, M, Chen, K, Shih, JC. Novel monoamine oxidase A knock out mice with human-like spontaneous mutation. Neuroreport. 2008; 19(7): 739743.Google Scholar
Cases, O, Vitalis, T, Seif, I, et al. Lack of barrels in the somatosensory cortex of monoamine oxidase A-deficient mice: role of a serotonin excess during the critical period. Neuron. 1996; 16(2): 297307.Google Scholar
Bortolato, M, Godar, SC, Tambaro, S, et al. Early postnatal inhibition of serotonin synthesis results in long-term reductions of perseverative behaviors, but not aggression, in MAO A-deficient mice. Neuropharmacology. 2013; 75: 223232.Google Scholar
Garcia-Garcia, AL, Newman-Tancredi, A, Leonardo, ED. 5-HT(1A) [corrected] receptors in mood and anxiety: recent insights into autoreceptor versus heteroreceptor function. Psychopharmacology (Berl.). 2014; 231(4): 623636.Google Scholar
Polter, AM, Li, X. 5-HT1A receptor-regulated signal transduction pathways in brain. Cell Signal. 2010; 22(10): 14061412.Google Scholar
Miczek, KA, Hussain, S, Faccidomo, S. Alcohol-heightened aggression in mice: attenuation by 5-HT1A receptor agonists. Psychopharmacology (Berl.). 1998; 139(1–2): 160168.Google Scholar
Pruus, K, Skrebuhhova-Malmros, T, Rudissaar, R, Matto, V, Allikmets, L. 5-HT1A receptor agonists buspirone and gepirone attenuate apomorphine-induced aggressive behaviour in adult male Wistar rats. J. Physiol. Pharmacol. 2000; 51(4 Pt 2): 833846.Google Scholar
Centenaro, LA, Vieira, K, Zimmermann, N, et al. Social instigation and aggressive behavior in mice: role of 5-HT1A and 5-HT1B receptors in the prefrontal cortex. Psychopharmacology (Berl.). 2008; 201(2): 237248.Google Scholar
da Veiga, CP, Miczek, KA, Lucion, AB, de Almeida, RM. Social instigation and aggression in postpartum female rats: role of 5-Ht1A and 5-Ht1B receptors in the dorsal raphe nucleus and prefrontal cortex. Psychopharmacology (Berl.). 2011; 213(2–3): 475487.Google Scholar
de Boer, SF, Lesourd, M, Mocaer, E, Koolhaas, JM. Somatodendritic 5-HT(1A) autoreceptors mediate the anti-aggressive actions of 5-HT(1A) receptor agonists in rats: an ethopharmacological study with S-15535, alnespirone, and WAY-100635. Neuropsychopharmacology. 2000; 23(1): 2033.Google Scholar
Stein, DJ, Miczek, KA, Lucion, AB, de Almeida, RM. Aggression-reducing effects of F15599, a novel selective 5-HT1A receptor agonist, after microinjection into the ventral orbital prefrontal cortex, but not in infralimbic cortex in male mice. Psychopharmacology (Berl.). 2013; 230(3): 375387.Google Scholar
Naumenko, VS, Kozhemyakina, RV, Plyusnina, IF, Kulikov, AV, Popova, NK. Serotonin 5-HT1A receptor in infancy-onset aggression: comparison with genetically defined aggression in adult rats. Behav. Brain Res. 2013; 243: 97101.Google Scholar
Popova, NK, Naumenko, VS, Plyusnina, IZ, Kulikov, AV. Reduction in 5-HT1A receptor density, 5-HT1A mRNA expression, and functional correlates for 5-HT1A receptors in genetically defined aggressive rats. J. Neurosci. Res. 2005; 80(2): 286292.Google Scholar
Popova, NK, Naumenko, VS, Plyusnina, IZ. Involvement of brain serotonin 5-HT1A receptors in genetic predisposition to aggressive behavior. Neurosci. Behav. Physiol. 2007; 37(6): 631635.Google Scholar
van der Vegt, BJ, de Boer, SF, Buwalda, B, et al. Enhanced sensitivity of postsynaptic serotonin-1A receptors in rats and mice with high trait aggression. Physiol. Behav. 2001; 74(1–2): 205211.Google Scholar
Audero, E, Mlinar, B, Baccini, G, et al. Suppression of serotonin neuron firing increases aggression in mice. J. Neurosci. 2013; 33(20): 86788688.Google Scholar
Almeida, M, Lee, R, Coccaro, EF. Cortisol responses to ipsapirone challenge correlate with aggression, while basal cortisol levels correlate with impulsivity, in personality disorder and healthy volunteer subjects. J. Psychiatr. Res. 2010; 44(14): 874880.Google Scholar
Coccaro, EF, Gabriel, S, Siever, LJ. Buspirone challenge: preliminary evidence for a role for central 5-HT1a receptor function in impulsive aggressive behavior in humans. Psychopharmacol. Bull. 1990; 26(3): 393405.Google Scholar
Benko, A, Lazary, J, Molnar, E, et al. Significant association between the C(-1019)G functional polymorphism of the HTR1A gene and impulsivity. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2010; 153B(2): 592599.Google Scholar
Joyce, PR, Stephenson, J, Kennedy, M, Mulder, RT, McHugh, PC. The presence of both serotonin 1A receptor (HTR1A) and dopamine transporter (DAT1) gene variants increase the risk of borderline personality disorder. Front. Genet. 2014; 4: 313.Google Scholar
Witte, AV, Floel, A, Stein, P, et al. Aggression is related to frontal serotonin-1A receptor distribution as revealed by PET in healthy subjects. Hum. Brain Mapp. 2009; 30(8): 25582570.Google Scholar
Parsey, RV, Oquendo, MA, Simpson, NR, et al. Effects of sex, age, and aggressive traits in man on brain serotonin 5-HT1A receptor binding potential measured by PET using [C-11]WAY-100635. Brain Res. 2002; 954(2): 173182.Google Scholar
Sari, Y. Serotonin1B receptors: from protein to physiological function and behavior. Neurosci. Biobehav. Rev. 2004; 28(6): 565582.Google Scholar
Bannai, M, Fish, EW, Faccidomo, S, Miczek, KA. Anti-aggressive effects of agonists at 5-HT1B receptors in the dorsal raphe nucleus of mice. Psychopharmacology (Berl.). 2007; 193(2): 295304.Google Scholar
de Almeida, RM, Miczek, KA. Aggression escalated by social instigation or by discontinuation of reinforcement (“frustration”) in mice: inhibition by anpirtoline: a 5-HT1B receptor agonist. Neuropsychopharmacology. 2002; 27(2): 171181.Google Scholar
de Almeida, RM, Rosa, MM, Santos, DM, et al. 5-HT(1B) receptors, ventral orbitofrontal cortex, and aggressive behavior in mice. Psychopharmacology (Berl.). 2006; 185(4): 441450.Google Scholar
Faccidomo, S, Bannai, M, Miczek, KA. Escalated aggression after alcohol drinking in male mice: dorsal raphe and prefrontal cortex serotonin and 5-HT(1B) receptors. Neuropsychopharmacology. 2008; 33(12): 28882899.Google Scholar
Faccidomo, S, Quadros, IM, Takahashi, A, Fish, EW, Miczek, KA. Infralimbic and dorsal raphe microinjection of the 5-HT(1B) receptor agonist CP-93,129: attenuation of aggressive behavior in CFW male mice. Psychopharmacology (Berl.). 2012; 222(1): 117128.Google Scholar
Fish, EW, Faccidomo, S, Miczek, KA. Aggression heightened by alcohol or social instigation in mice: reduction by the 5-HT(1B) receptor agonist CP-94,253. Psychopharmacology (Berl.). 1999; 146(4): 391399.Google Scholar
Fish, EW, McKenzie-Quirk, SD, Bannai, M, Miczek, KA. 5-HT(1B) receptor inhibition of alcohol-heightened aggression in mice: comparison to drinking and running. Psychopharmacology (Berl.). 2008; 197(1): 145156.Google Scholar
Veiga, CP, Miczek, KA, Lucion, AB, Almeida, RM. Effect of 5-HT1B receptor agonists injected into the prefrontal cortex on maternal aggression in rats. Braz. J. Med. Biol. Res. 2007; 40(6): 825830.Google Scholar
Ramboz, S, Saudou, F, Amara, DA, et al. 5-HT1B receptor knock out—behavioral consequences. Behav. Brain Res. 1996; 73(1–2): 305312.Google Scholar
Bouwknecht, JA, Hijzen, TH, van der Gugten, J, et al. Absence of 5-HT(1B) receptors is associated with impaired impulse control in male 5-HT(1B) knockout mice. Biol. Psychiatry. 2001; 49(7): 557568.Google Scholar
Conner, TS, Jensen, KP, Tennen, H, et al. Functional polymorphisms in the serotonin 1B receptor gene (HTR1B) predict self-reported anger and hostility among young men. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2010; 153B(1): 6778.Google Scholar
Hakulinen, C, Jokela, M, Hintsanen, M, et al. Serotonin receptor 1B genotype and hostility, anger and aggressive behavior through the lifespan: the Young Finns study. J. Behav. Med. 2013; 36(6): 583590.Google Scholar
Jensen, KP, Covault, J, Conner, TS, et al. A common polymorphism in serotonin receptor 1B mRNA moderates regulation by miR-96 and associates with aggressive human behaviors. Mol. Psychiatry. 2009; 14(4): 381389.Google Scholar
Zouk, H, McGirr, A, Lebel, V, et al. The effect of genetic variation of the serotonin 1B receptor gene on impulsive aggressive behavior and suicide. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2007; 144B(8): 9961002.Google Scholar
Potenza, MN, Walderhaug, E, Henry, S, et al. Serotonin 1B receptor imaging in pathological gambling. World J. Biol. Psychiatry. 2013; 14(2): 139145.Google Scholar
Hu, J, Henry, S, Gallezot, JD, et al. Serotonin 1B receptor imaging in alcohol dependence. Biol. Psychiatry. 2010; 67(9): 800803.Google Scholar
Murrough, JW, Henry, S, Hu, J, et al. Reduced ventral striatal/ ventral pallidal serotonin1B receptor binding potential in major depressive disorder. Psychopharmacology (Berl.). 2011; 213(2–3): 547553.Google Scholar
Higgins, GA, Enderlin, M, Haman, M, Fletcher, PJ. The 5-HT2A receptor antagonist M100,907 attenuates motor and ‘impulsive-type’ behaviours produced by NMDA receptor antagonism. Psychopharmacology (Berl.). 2003; 170(3): 309319.Google Scholar
Sakaue, M, Ago, Y, Sowa, C, et al. Modulation by 5-hT2A receptors of aggressive behavior in isolated mice. Jpn J. Pharmacol. 2002; 89(1): 8992.Google Scholar
Winstanley, CA, Theobald, DE, Dalley, JW, Glennon, JC, Robbins, TW. 5-HT2A and 5-HT2C receptor antagonists have opposing effects on a measure of impulsivity: interactions with global 5-HT depletion. Psychopharmacology (Berl.). 2004; 176(3–4): 376385.Google Scholar
Giegling, I, Hartmann, AM, Moller, HJ, Rujescu, D. Anger- and aggression-related traits are associated with polymorphisms in the 5-HT-2A gene. J. Affect. Disord. 2006; 96(1–2): 7581.Google Scholar
Bruce, KR, Steiger, H, Joober, R, et al. Association of the promoter polymorphism -1438G/A of the 5-HT2A receptor gene with behavioral impulsiveness and serotonin function in women with bulimia nervosa. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2005; 137B(1): 4044.Google Scholar
Jakubczyk, A, Wrzosek, M, Lukaszkiewicz, J, et al. The CC genotype in HTR2A T102C polymorphism is associated with behavioral impulsivity in alcohol-dependent patients. J. Psychiatr. Res. 2012; 46(1): 4449.Google Scholar
Jakubczyk, A, Klimkiewicz, A, Kopera, M, et al. The CC genotype in the T102C HTR2A polymorphism predicts relapse in individuals after alcohol treatment. J. Psychiatr. Res. 2013; 47(4): 527533.Google Scholar
Preuss, UW, Koller, G, Bondy, B, Bahlmann, M, Soyka, M. Impulsive traits and 5-HT2A receptor promoter polymorphism in alcohol dependents: possible association but no influence of personality disorders. Neuropsychobiology. 2001; 43(3): 186191.Google Scholar
Tsuang, HC, Chen, WJ, Lin, SH, et al. Impaired impulse control is associated with a 5-HT2A receptor polymorphism in schizophrenia. Psychiatry Res. 2013; 208(2): 105110.Google Scholar
Bjork, JM, Moeller, FG, Dougherty, DM, et al. Serotonin 2a receptor T102C polymorphism and impaired impulse control. Am. J. Med. Genet. 2002; 114(3): 336339.Google Scholar
Oquendo, MA, Russo, SA, Underwood, MD, et al. Higher postmortem prefrontal 5-HT2A receptor binding correlates with lifetime aggression in suicide. Biol. Psychiatry. 2006; 59(3): 235243.Google Scholar
Dwivedi, Y, Mondal, AC, Payappagoudar, GV, Rizavi, HS. Differential regulation of serotonin (5HT)2A receptor mRNA and protein levels after single and repeated stress in rat brain: role in learned helplessness behavior. Neuropharmacology. 2005; 48(2): 204214.Google Scholar
Soloff, PH, Chiappetta, L, Mason, NS, Becker, C, Price, JC. Effects of serotonin-2A receptor binding and gender on personality traits and suicidal behavior in borderline personality disorder. Psychiatry Res. 2014; 222(3): 140148.Google Scholar
Meyer, JH, Wilson, AA, Rusjan, P, et al. Serotonin2A receptor binding potential in people with aggressive and violent behaviour. J. Psychiatry Neurosci. 2008; 33(6): 499508.Google Scholar
Rylands, AJ, Hinz, R, Jones, M, et al. Pre- and postsynaptic serotonergic differences in males with extreme levels of impulsive aggression without callous unemotional traits: a positron emission tomography study using (11)C-DASB and (11)C-MDL100907. Biol. Psychiatry. 2012; 72(12): 10041011.Google Scholar
Chameau, P, van Hooft, JA. Serotonin 5-HT(3) receptors in the central nervous system. Cell Tissue Res. 2006; 326(2): 573581.Google Scholar
De Deurwaerdère, P, Moison, D, Navailles, S, Porras, G, Spampinato, U. Regionally and functionally distinct serotonin3 receptors control in vivo dopamine outflow in the rat nucleus accumbens. J. Neurochem. 2005; 94(1): 140149.Google Scholar
Cervantes, MC, Delville, Y. Serotonin 5-HT1A and 5-HT3 receptors in an impulsive-aggressive phenotype. Behav. Neurosci. 2009; 123(3): 589598.Google Scholar
Ricci, LA, Grimes, JM, Melloni, RH Jr. Serotonin type 3 receptors modulate the aggression-stimulating effects of adolescent cocaine exposure in Syrian hamsters (Mesocricetus auratus). Behav. Neurosci. 2004; 118(5): 10971110.Google Scholar
Ricci, LA, Knyshevski, I, Melloni, RH Jr. Serotonin type 3 receptors stimulate offensive aggression in Syrian hamsters. Behav. Brain Res. 2005; 156(1): 1929.Google Scholar
Rudissaar, R, Pruus, K, Skrebuhhova, T, Allikmets, L, Matto, V. Modulatory role of 5-HT3 receptors in mediation of apomorphine-induced aggressive behaviour in male rats. Behav. Brain Res. 1999; 106(1–2): 9196.Google Scholar
McKenzie-Quirk, SD, Girasa, KA, Allan, AM, Miczek, KA. 5-HT(3) receptors, alcohol and aggressive behavior in mice. Behav. Pharmacol. 2005; 16(3): 163169.Google Scholar
Sellers, EM, Toneatto, T, Romach, MK, et al. Clinical efficacy of the 5-HT3 antagonist ondansetron in alcohol abuse and dependence. Alcohol Clin. Exp. Res. 1994; 18(4): 879885.Google Scholar
Johnson, BA, Ait-Daoud, N, Ma, JZ, Wang, Y. Ondansetron reduces mood disturbance among biologically predisposed, alcohol-dependent individuals. Alcohol Clin. Exp. Res. 2003; 27(11): 17731779.Google Scholar
Myrick, H, Anton, RF, Li, X, et al. Effect of naltrexone and ondansetron on alcohol cue-induced activation of the ventral striatum in alcohol-dependent people. Arch. Gen. Psychiatry. 2008; 65(4): 466475.Google Scholar
Ducci, F, Enoch, MA, Yuan, Q, et al. HTR3B is associated with alcoholism with antisocial behavior and alpha EEG power—an intermediate phenotype for alcoholism and co-morbid behaviors. Alcohol. 2009; 43(1): 7384.Google Scholar
Melke, J, Westberg, L, Nilsson, S, et al. A polymorphism in the serotonin receptor 3A (HTR3A) gene and its association with harm avoidance in women. Arch. Gen. Psychiatry. 2003; 60(10): 10171023.Google Scholar
Gatt, JM, Williams, LM, Schofield, PR, et al. Impact of the HTR3A gene with early life trauma on emotional brain networks and depressed mood. Depress. Anxiety. 2010; 27(8): 752759.Google Scholar
Iidaka, T, Ozaki, N, Matsumoto, A, et al. A variant C178T in the regulatory region of the serotonin receptor gene HTR3A modulates neural activation in the human amygdala. J. Neurosci. 2005; 25(27): 64606466.Google Scholar
Schlüter, T, Winz, O, Henkel, K, et al. The impact of dopamine on aggression: an [18F]-FDOPA PET Study in healthy males. J. Neurosci. 2013; 33(43): 16, 88916, 896.Google Scholar
Buckholtz, JW, Treadway, MT, Cowan, RL, et al. Mesolimbic dopamine reward system hypersensitivity in individuals with psychopathic traits. Nat. Neurosci. 2010; 13(4): 419421.Google Scholar
Wagner, S, Baskaya, O, Anicker, NJ, et al. The catechol o-methyltransferase (COMT) val(158)met polymorphism modulates the association of serious life events (SLE) and impulsive aggression in female patients with borderline personality disorder (BPD). Acta Psychiatr. Scand. 2010; 122(2): 110117.Google Scholar
Bhakta, SG, Zhang, JP, Malhotra, AK. The COMT Met158 allele and violence in schizophrenia: a meta-analysis. Schizophr. Res. 2012; 140(1–3): 192197.Google Scholar
Flory, JD, Xu, K, New, AS, et al. Irritable assault and variation in the COMT gene. Psychiatr. Genet. 2007; 17(6): 344346.Google Scholar
Soyka, M, Zill, P, Koller, G, et al. Val158Met COMT polymorphism and risk of aggression in alcohol dependence. Addict. Biol. 2015; 20(1): 197204.Google Scholar
Vevera, J, Stopkova, R, Bes, M, et al. COMT polymorphisms in impulsively violent offenders with antisocial personality disorder. Neuro. Endocrinol. Lett. 2009; 30(6): 753756.Google Scholar
Hirata, Y, Zai, CC, Nowrouzi, B, Beitchman, JH, Kennedy, JL. Study of the catechol-o-methyltransferase (COMT) gene with high aggression in children. Aggress. Behav. 2013; 39(1): 4551.Google Scholar
Fresan, A, Camarena, B, Apiquian, R, et al. Association study of MAO-A and DRD4 genes in schizophrenic patients with aggressive behavior. Neuropsychobiology. 2007; 55(3–4): 171175.Google Scholar
Buchmann, AF, Zohsel, K, Blomeyer, D, et al. Interaction between prenatal stress and dopamine D4 receptor genotype in predicting aggression and cortisol levels in young adults. Psychopharmacology (Berl.). 2014; 231(16): 30893097.Google Scholar
Delville, Y, Mansour, KM, Ferris, CF. Serotonin blocks vasopressin-facilitated offensive aggression: interactions within the ventrolateral hypothalamus of golden hamsters. Physiol. Behav. 1996; 59(4–5): 813816.Google Scholar
Ferris, CF, Melloni, RH Jr, Koppel, G, et al. Vasopressin/serotonin interactions in the anterior hypothalamus control aggressive behavior in golden hamsters. J. Neurosci. 1997; 17(11): 43314340.Google Scholar
Ferris, CF, Potegal, M. Vasopressin receptor blockade in the anterior hypothalamus suppresses aggression in hamsters. Physiol. Behav. 1988; 44(2): 235239.Google Scholar
Bosch, OJ, Neumann, ID. Vasopressin released within the central amygdala promotes maternal aggression. Eur. J. Neurosci. 2010; 31(5): 883891.Google Scholar
Wersinger, SR, Caldwell, HK, Christiansen, M, Young, WS III. Disruption of the vasopressin 1b receptor gene impairs the attack component of aggressive behavior in mice. Genes Brain Behav. 2007; 6(7): 653660.Google Scholar
Wersinger, SR, Ginns, EI, O’Carroll, AM, Lolait, SJ, Young, WS III. Vasopressin V1b receptor knockout reduces aggressive behavior in male mice. Mol. Psychiatry. 2002; 7(9): 975984.Google Scholar
Wersinger, SR, Caldwell, HK, Martinez, L, et al. Vasopressin 1a receptor knockout mice have a subtle olfactory deficit but normal aggression. Genes Brain Behav. 2007; 6(6): 540551.Google Scholar
Fodor, A, Barsvari, B, Aliczki, M, et al. The effects of vasopressin deficiency on aggression and impulsiveness in male and female rats. Psychoneuroendocrinology. 2014; 47: 141150.Google Scholar
Uzefovsky, F, Shalev, I, Israel, S, Knafo, A, Ebstein, RP. Vasopressin selectively impairs emotion recognition in men. Psychoneuroendocrinology. 2012; 37(4): 576580.Google Scholar
Guastella, AJ, Kenyon, AR, Alvares, GA, Carson, DS, Hickie, IB. Intranasal arginine vasopressin enhances the encoding of happy and angry faces in humans. Biol. Psychiatry. 2010; 67(12): 12201222.Google Scholar
Lee, RJ, Coccaro, EF, Cremers, H, et al. A novel V1a receptor antagonist blocks vasopressin-induced changes in the CNS response to emotional stimuli: an fMRI study. Front. Syst. Neurosci. 2013; 7: 100.Google Scholar
Luppino, D, Moul, C, Hawes, DJ, Brennan, J, Dadds, MR. Association between a polymorphism of the vasopressin 1B receptor gene and aggression in children. Psychiatr. Genet. 2014; 24(5): 185190.Google Scholar
Zai, CC, Muir, KE, Nowrouzi, B, et al. Possible genetic association between vasopressin receptor 1B and child aggression. Psychiatry Res. 2012; 200(2–3): 784788.Google Scholar
Vogel, F, Wagner, S, Baskaya, O, et al. Variable number of tandem repeat polymorphisms of the arginine vasopressin receptor 1A gene and impulsive aggression in patients with borderline personality disorder. Psychiatr. Genet. 2012; 22(2): 105106.Google Scholar
McBurnett, K, Lahey, BB, Rathouz, PJ, Loeber, R. Low salivary cortisol and persistent aggression in boys referred for disruptive behavior. Arch. Gen. Psychiatry. 2000; 57(1): 3843.Google Scholar
Popma, A, Vermeiren, R, Geluk, CA, et al. Cortisol moderates the relationship between testosterone and aggression in delinquent male adolescents. Biol. Psychiatry. 2007; 61(3): 405411.Google Scholar
Kuepper, Y, Alexander, N, Osinsky, R, et al. Aggression–interactions of serotonin and testosterone in healthy men and women. Behav. Brain Res. 2010; 206(1): 93100.Google Scholar
Welker, KM, Lozoya, E, Campbell, JA, Neumann, CS, Carre, JM. Testosterone, cortisol, and psychopathic traits in men and women. Physiol. Behav. 2014; 129: 230236.Google Scholar
Denson, TF, Mehta, PH, Ho, TD. Endogenous testosterone and cortisol jointly influence reactive aggression in women. Psychoneuroendocrinology. 2013; 38(3): 416424.Google Scholar
Cima, M, Smeets, T, Jelicic, M. Self-reported trauma, cortisol levels, and aggression in psychopathic and non-psychopathic prison inmates. Biol. Psychol. 2008; 78(1): 7586.Google Scholar
Derntl, B, Windischberger, C, Robinson, S, et al. Amygdala activity to fear and anger in healthy young males is associated with testosterone. Psychoneuroendocrinology. 2009; 34(5): 687693.Google Scholar
Hermans, EJ, Ramsey, NF, van Honk, J. Exogenous testosterone enhances responsiveness to social threat in the neural circuitry of social aggression in humans. Biol. Psychiatry. 2008; 63(3): 263270.Google Scholar
van Wingen, G, Mattern, C, Verkes, RJ, Buitelaar, J, Fernández, G. Testosterone reduces amygdala-orbitofrontal cortex coupling. Psychoneuroendocrinology. 2010; 35(1): 105113.Google Scholar
Goetz, SM, Tang, L, Thomason, ME, et al. Testosterone rapidly increases neural reactivity to threat in healthy men: a novel two-step pharmacological challenge paradigm. Biol. Psychiatry. 2014; 76(4): 324331.Google Scholar
Grillo, CA, Risher, M, Macht, VA, et al. Repeated restraint stress-induced atrophy of glutamatergic pyramidal neurons and decreases in glutamatergic efflux in the rat amygdala are prevented by the antidepressant agomelatine. Neuroscience. 2015; 284: 430443.Google Scholar
Padival, MA, Blume, SR, Vantrease, JE, Rosenkranz, JA. Qualitatively different effect of repeated stress during adolescence on principal neuron morphology across lateral and basal nuclei of the rat amygdala. Neuroscience. 2015; 291: 128145.Google Scholar
Vyas, A, Mitra, R, Shankaranarayana Rao, BS, Chattarji, S. Chronic stress induces contrasting patterns of dendritic remodeling in hippocampal and amygdaloid neurons. J. Neurosci. 2002; 22(15): 68106818.Google Scholar
Vyas, A, Bernal, S, Chattarji, S. Effects of chronic stress on dendritic arborization in the central and extended amygdala. Brain Res. 2003; 965(1–2): 290294.Google Scholar
Gilabert-Juan, J, Castillo-Gomez, E, Perez-Rando, M, Molto, MD, Nacher, J. Chronic stress induces changes in the structure of interneurons and in the expression of molecules related to neuronal structural plasticity and inhibitory neurotransmission in the amygdala of adult mice. Exp. Neurol. 2011; 232(1): 3340.Google Scholar

References

McEvoy, JP. The costs of schizophrenia. J. Clin. Psychiatry. 2007; 68(Suppl 14): 47.Google Scholar
Torrey, EF. Stigma and violence: isn’t it time to connect the dots? Schizophr. Bull. 2011; 37(5): 892896.Google Scholar
Volavka, J, Citrome, L. Heterogeneity of violence in schizophrenia and implications for long-term treatment. Int. J. Clin. Pract. 2008; 62(8): 12371245.Google Scholar
Stahl, SM. Deconstructing violence as a medical syndrome: mapping psychotic, impulsive, and predatory subtypes to malfunctioning brain circuits. CNS Spectr. 2014; 19(5): 357365.Google Scholar
Witt, K, van Dorn, R, Fazel, S. Risk factors for violence in psychosis: systematic review and meta-regression analysis of 110 studies. PLoS One. 2013; 8(2): e55942.Google Scholar
Nolan, KA, Czobor, P, Roy, BB, et al. Characteristics of assaultive behavior among psychiatric inpatients. Psychiatr. Serv. 2003; 54(7): 10121016.Google Scholar
Dodge, KA, Lochman, JE, Harnish, JD, Bates, JE, Pettit, GS. Reactive and proactive aggression in school children and psychiatrically impaired chronically assaultive youth. J. Abnorm. Psychol. 1997; 106(1): 3751.Google Scholar
McDermott, BE, Holoyda, BJ. Assessment of aggression in inpatient settings. CNS Spectr. 2014; 19(05): 425431.Google Scholar
Nolan, KA, Volavka, J, Mohr, P, Czobor, P. Psychopathy and violent behavior among patients with schizophrenia or schizoaffective disorder. Psychiatr. Serv. 1999; 50(6): 787.Google Scholar
Patton, JH, Stanford, MS, Barratt, ES. Factor structure of the Barratt impulsiveness scale. J. Clin. Psychol. 1995; 51(6): 768774.Google Scholar
Nolan, KA, D’Angelo, D, Hoptman, MJ. Self-report and laboratory measures of impulsivity in patients with schizophrenia or schizoaffective disorder and healthy controls. Psychiatry Res. 2011; 187(1–2): 301303.Google Scholar
Barratt, ES. Impulsivity: integrating cognitive, behavioral, biological, and environmental data. In: McCowan, W, Johnson, J, Shure, M, eds. The Impulsive Client: Theory, Research, and Treatment. American Psychological Association; 1993: 3956.Google Scholar
Lipszyc, J, Schachar, R. Inhibitory control and psychopathology: a meta-analysis of studies using the stop signal task. J. Int. Neuropsychol. Soc. 2010; 16(6): 10641076.Google Scholar
Chambers, CD, Garavan, H, Bellgrove, MA. Insights into the neural basis of response inhibition from cognitive and clinical neuroscience. Neurosci. Biobehav. Rev. 2009; 33(5): 631646.Google Scholar
Enticott, PG, Ogloff, JRP, Bradshaw, JL, Daffern, M. Contrary to popular belief, a lack of behavioural inhibitory control may not be associated with aggression. Crim. Behav. Ment. Health. 2007; 17(3): 179183.Google Scholar
Whiteside, SP, Lynam, DR. The five factor model and impulsivity: using a structural model of personality to understand impulsivity. Personality and Individual Differences. 2001; 30(4): 669689.Google Scholar
Cyders, MA, Smith, GT. Mood-based rash action and its components: positive and negative urgency. Personality and Individual Differences. 2007; 43(4): 839850.Google Scholar
Hoptman, MJ, Antonius, D, Mauro, CJ, Parker, EM, Javitt, DC. Cortical thinning, functional connectivity, and mood-related impulsivity in schizophrenia: relationship to aggressive attitudes and behavior. Am. J. Psychiatry. 2014; 171(9): 943948.Google Scholar
Cyders, MA, Smith, GT. Emotion-based dispositions to rash action: positive and negative urgency. Psychol. Bull. 2008; 134(6): 807828.Google Scholar
Cyders, MA, Dzemidzic, M, Eiler, WJ, et al. Negative urgency mediates the relationship between amygdala and orbitofrontal cortex activation to negative emotional stimuli and general risk-taking. ereb Cortex. doi:10.1093/cercor/bhu123.Google Scholar
Enticott, PG, Ogloff, JR, Bradshaw, JL. Response inhibition and impulsivity in schizophrenia. Psychiatry Res. 2008; 157(1): 251254.Google Scholar
Enticott, PG, Ogloff, JR, Bradshaw, JL. Associations between laboratory measures of executive inhibitory control and self-reported impulsivity. Personality and Individual Differences. 2006; 41(2): 285294.Google Scholar
Pawliczek, CM, Derntl, B, Kellermann, T, et al. Inhibitory control and trait aggression: neural and behavioral insights using the emotional stop signal task. Neuroimage. 2013; 79: 264274.Google Scholar
Green, MF, Lee, J. Neural bases of emotional experience versus perception in schizophrenia. Biol. Psychiatry. 2012; 71(2): 9697.Google Scholar
Insel, TR, Cuthbert, BN, Garvey, MA, et al. Research domain criteria (RDoC): toward a new classification framework for research on mental disorders. Am. J. Psychiatry. 2010; 167(7): 748751.Google Scholar
Yudofsky, SC, Silver, JM, Jackson, W, Endicott, J, Williams, D. The Overt Aggression Scale for the objective rating of verbal and physical aggression. Am. J. Psychiatry. 1986; 143(1): 3539.Google Scholar
Buss, AH, Perry, M. The aggression questionnaire. J. Pers. Soc. Psychol. 1992; 63(3): 452459.Google Scholar
Taylor, SP. Aggressive behavior and physiological arousal as a function of provocation and the tendency to inhibit aggression. J. Pers. 1967; 35(2): 297310.Google Scholar
Cherek, DR, Moeller, FG, Dougherty, DM, Rhoades, H. Studies of violent and nonviolent male parolees: II. Laboratory and psychometric measurements of impulsivity. Biol. Psychiatry. 1997; 41(5): 523529.Google Scholar
Soyka, M. Neurobiology of aggression and violence in schizophrenia. Schizophr. Bull. 2011; 37(5): 913920.Google Scholar
Hoptman, MJ, Antonius, D. Neuroimaging correlates of aggression in schizophrenia: an update. Curr. Opin. Psychiatry. 2011; 24(2): 100106.Google Scholar
Weiss, EM. Neuroimaging and neurocognitive correlates of aggression and violence in schizophrenia. Scientifica. 2012; 2012: 158646.Google Scholar
Volavka, J. Neurobiology of Violence, 2nd edn. Washington, DC: American Psychiatric Association; 2001.Google Scholar
Hoptman, MJ, Volavka, J, Czobor, P, et al. Aggression and quantitative MRI measures of caudate in patients with chronic schizophrenia or schizoaffective disorder. J. Neuropsychiatry Clin. Neurosci. 2006; 18(4): 509515.Google Scholar
Hoptman, MJ, Volavka, J, Weiss, EM, et al. Quantitative MRI measures of orbitofrontal cortex in patients with chronic schizophrenia or schizoaffective disorder. Psychiatry Res. 2005; 140(2): 133145.Google Scholar
Chakos, MH, Lieberman, JA, Bilder, RM, et al. Increase in caudate nuclei volumes of first-episode schizophrenic patients taking antipsychotic drugs. Am. J. Psychiatry. 1994; 151(10): 14301436.Google Scholar
Rüsch, N, Spoletini, I, Wilke, M, et al. Inferior frontal white matter volume and suicidality in schizophrenia. Psychiatry Res. 2008; 164(3): 206214.Google Scholar
Narayan, VM, Narr, KL, Kumari, V, et al. Regional cortical thinning in subjects with violent antisocial personality disorder or schizophrenia. Am. J. Psychiatry. 2007; 164(9): 14181427.Google Scholar
Hoptman, MJ, Volavka, J, Johnson, G, et al. Frontal white matter microstructure, aggression, and impulsivity in men with schizophrenia: a preliminary study. Biol. Psychiatry. 2002; 52(1): 914.Google Scholar
Raine, A, Buchsbaum, MS, Stanley, J, et al. Selective reductions in prefrontal glucose metabolism in murderers. Biol. Psychiatry. 1994; 36(6): 365373.Google Scholar
Volkow, ND, Tancredi, LR, Grant, C, et al. Brain glucose metabolism in violent psychiatric patients: a preliminary study. Psychiatry Res. 1995; 61(4): 243253.Google Scholar
Wong, MTH, Fenwick, PBC, Lumsden, J, et al. Positron emission tomography in male violent offenders with schizophrenia. Psychiatry Res. 1997; 68(2): 111123.Google Scholar
Joyal, C, Putkonen, A, Mancini-Marie, A, et al. Violent persons with schizophrenia and comorbid disorders: a functional magnetic resonance imaging study. Schizophr. Res. 2007; 91(1): 97102.Google Scholar
Kumari, V, Das, M, Taylor, PJ, et al. Neural and behavioural responses to threat in men with a history of serious violence and schizophrenia or antisocial personality disorder. Schizophr. Res. 2009; 110(1): 4758.Google Scholar
Dolan, MC, Fullam, RS. Psychopathy and functional magnetic resonance imaging blood oxygenation level-dependent responses to emotional faces in violent patients with schizophrenia. Biol. Psychiatry. 2009; 66(6): 570577.Google Scholar
Hoptman, MJ, D’Angelo, D, Catalano, D, et al. Amygdalofrontal functional disconnectivity and aggression in schizophrenia. Schizophr. Bull. 2010; 36(5): 10201028.Google Scholar
Szeszko, PR. Aggression in schizophrenia and its relationship to neural circuitry of urgency. Am. J. Psychiatry. 2014; 171(9): 897900.Google Scholar
Davidson, RJ, Putnam, KM, Larson, CL. Dysfunction in the neural circuitry of emotion regulation—a possible prelude to violence. Science. 2000; 289(5479): 591594.Google Scholar
Kring, AM, Werner, KH. Emotion regulation and psychopathology. In: Philippot, P, Feldman, RS, eds. The Regulation of Emotion. Mahwah, NJ: Lawrence Erlbaum Associates; 2004: 359385.Google Scholar
Gross, JJ, Jazaieri, H. Emotion, emotion regulation, and psychopathology: an affective science perspective. Clinical Psychological Science. 2014; 2(4): 387401.Google Scholar
Perry, Y, Henry, JD, Nangle, MR, Grisham, JR. Regulation of negative affect in schizophrenia: the effectiveness of acceptance versus reappraisal and suppression. J. Clin. Exp. Neuropsychol. 2012; 34(5): 497508.Google Scholar
Krakowski, MI, Czobor, P, Citrome, L, Bark, N, Cooper, TB. Atypical antipsychotic agents in the treatment of violent patients with schizophrenia and schizoaffective disorder. Arch. Gen. Psychiatry. 2006; 63(6): 622629.Google Scholar
Volavka, J, Czobor, P, Nolan, K, et al. Overt aggression and psychotic symptoms in patients with schizophrenia treated with clozapine, olanzapine, risperidone, or haloperidol. J. Clin. Psychopharmacol. 2004; 24(2): 225228.Google Scholar
Arango, V, Underwood, MD, Mann, JJ. Serotonin brain circuits involved in major depression and suicide. Prog. Brain Res. 2002; 136: 443453.Google Scholar
Citrome, L, Volavka, J. The psychopharmacology of violence: making sensible decisions. CNS Spectr. 2014; 19(5): 411418.Google Scholar
Biswal, BB, Mennes, M, Zuo, XN, et al. Toward discovery science of human brain function. Proc. Natl Acad. Sci. U S A. 2010; 107(10): 47344739.Google Scholar
Bersani, F, Minichino, A, Enticott, P, et al. Deep transcranial magnetic stimulation as a treatment for psychiatric disorders: a comprehensive review. Eur. Psychiatry. 2013; 28(1): 3039.Google Scholar
LaConte, SM, Peltier, SJ, Hu, XP. Real-time fMRI using brain-state classification. Hum. Brain Mapp. 2007; 28(10): 10331044.Google Scholar
Raichle, ME, MacLeod, AM, Snyder, AZ, et al. A default mode of brain function. Proc. Natl Acad. Sci. U S A. 2001; 98(2): 676682.Google Scholar
Gusnard, DA, Akbudak, E, Shulman, GL, Raichle, ME. Medial prefrontal cortex and self-referential mental activity: relation to a default mode of brain function. Proc. Natl Acad. Sci. U S A. 2001; 98(7): 42594264.Google Scholar
Ruiz, S, Lee, S, Soekadar, SR, et al. Acquired self-control of insula cortex modulates emotion recognition and brain network connectivity in schizophrenia. Hum. Brain Mapp. 2013; 34(1): 200212.Google Scholar
Hampson, M, Stoica, T, Saksa, J, et al. Real-time fMRI biofeedback targeting the orbitofrontal cortex for contamination anxiety. J. Vis. Exp. 2012; (59): 3535.Google Scholar
Sitaram, R, Caria, A, Veit, R, et al. Volitional control of the anterior insula in criminal psychopaths using realtime fMRI neurofeedback: a pilot study. Front. Behav. Neurosci. 2014; 8: 344.Google Scholar
Keers, R, Ullrich, S, DeStavola, BL, Coid, JW. Association of violence with emergence of persecutory delusions in untreated schizophrenia. Am. J. Psychiatry. 2014; 171(3): 332339.Google Scholar
Coid, JW, Ullrich, S, Kallis, C, et al. The relationship between delusions and violence: findings from the East London first episode psychosis study. JAMA Psychiatry. 2013; 70(5): 465471.Google Scholar
Johnson, SL, Carver, CS, Joormann, J. Impulsive responses to emotion as a transdiagnostic vulnerability to internalizing and externalizing symptoms. J. Affect. Disord. 2013; 150(3): 872878.Google Scholar
Dambacher, F, Sack, AT, Lobbestael, J, et al. Out of control evidence for anterior insula involvement in motor impulsivity and reactive aggression. Soc. Cogn. Affect. Neurosci. doi:10.1093/scan/nsu077.Google Scholar
Dambacher, F, Sack, AT, Lobbestael, J, et al. The role of right prefrontal and medial cortex in response inhibition: Interfering with action restraint and action cancellation using transcranial magnetic brain stimulation. J. Cogn. Neurosci. 2014; 26(8): 17751784.Google Scholar
Warburton, K. The new mission of forensic mental health services: managing violence as a medical syndrome in an environment that balances treatment and safety. CNS Spectr. 2014; 19(5): 368373.Google Scholar
Stahl, SM, Morrissette, DA, Cummings, M, et al. California State Hospital Violence Assessment and Treatment (Cal-VAT). CNS Spectr. 2014; 19(5): 449465.Google Scholar

References

Dodge, KA. The structure and function of reactive and proactive aggression. In: Peppler, D, Rubin, K eds. The Development and Treatment of Childhood Aggression. Hillsdale, NJ: Erlbaum; 1991: 201218.Google Scholar
Coccaro, EF. Intermittent explosive disorder as a disorder of impulsive aggression for DSM-5. Am. J. Psychiatry. 2012; 169(6): 577588.Google Scholar
Berman, ME, Fallon, AE, Coccaro, EF. The relationship between personality psychopathology and aggressive behavior in research volunteers. J. Abnorm. Psychol. 1998; 107(4): 651658.Google Scholar
Kessler, RC, Coccaro, EF, Fava, M, et al. The prevalence and correlates of DSM-IV intermittent explosive disorder in the National Comorbidity Survey Replication. Arch. Gen. Psychiatry. 2006; 63(6): 669678.Google Scholar
Yanowitch, R, Coccaro, EF. The neurochemistry of human aggression. Adv. Genet. 2011; 75: 151169.Google Scholar
Coccaro, EF, Kavoussi, RJ, Hauger, RL, Cooper, TB, Ferris, CF. Cerebrospinal fluid vasopressin levels: correlates with aggression and serotonin function in personality-disordered subjects. Arch. Gen. Psychiatry. 1998; 55(8): 708714.Google Scholar
Coccaro, EF, Lee, R, Liu, T, Mathé, AA. CSF NPY correlates with aggression in human subjects. Biol. Psychiatry. 2012; 72(12): 9971003.Google Scholar
Coccaro, EF, Lee, R, Owens, MJ, Kinkead, B, Nemeroff, CB. Cerebrospinal fluid substance P-like immunoreactivity correlates with aggression in personality disordered subjects. Biol. Psychiatry. 2012; 72(3): 238243.Google Scholar
Coccaro, EF, Lee, R, Vezina, P. Cerebrospinal fluid glutamate concentration correlates with impulsive aggression in human subjects. J. Psychiatr. Res. 2013; 47(9): 12471253.Google Scholar
Coccaro, EF, Lee, R, Coussons-Read, M. Elevated plasma inflammatory markers in individuals with intermittent explosive disorder and correlation with aggression in humans. JAMA Psychiatry. 2014; 71(2): 158165.Google Scholar
Asberg, M, Schalling, D, Traskman-Bendz, L, Wagner, A. Psychobiology of suicide, impulsivity, and related phenomena. In Davis, K ed. Psychopharmacology: The Third Generation of Progress. New York: Raven Press; 1987: 665668.Google Scholar
Bourne, HR, Bunney, WE, Colburn, RW, et al. Noradrenaline, 5-hydroxytryptamine, and 5-hydroxyindoleacetic acid hindbrains of suicidal patients. Lancet. 1968; 2(7572): 805808.Google Scholar
Pare, CMB, Yeung, DPH, Price, K, Stacey, RS. 5-Hydroxytryptamine, noradrenaline, and dopamine in brainstem, hypothalamus, and caudate nucleus of controls and of patients committing suicide by coal-gas poisoning. Lancet. 1969; 2 (7612): 133135.Google Scholar
Shaw, DM, Eccleston, EG, Camps, FE. 5-Hydroxytryptamine in the hind-brain of depressive suicides. Br. J. Psychiatry. 1967; 113(505): 14071411.Google Scholar
Stanley, M, Traskman-Bendz, L, Dorovini-Zis, K. Correlations between aminergic metabolites simultaneously obtained from human CSF and brain. Life Sci. 1985; 37(14): 12791286.Google Scholar
Asberg, M, Träskman, L, Thorén, P. 5-HIAA in the cerebrospinal fluid: a biochemical suicide predictor? Arch. Gen. Psychiatry. 1976; 33(10): 11931197.Google Scholar
Brown, GL, Goodwin, FK, Ballenger, JC, Goyer, PF, Major, LF. Aggression in humans correlates with cerebrospinal fluid amine metabolites. Psychiatry Res. 1979; 1(2): 131139.Google Scholar
Brown, GL, Ebert, MH, Goyer, PF, et al. Aggression, suicide, and serotonin: relationships to CSF aminemetabolites. Am. J. Psychiatry. 1982; 139(6): 741746.Google Scholar
Lidberg, L, Tuck, JR, Asberg, M, Scalia-Tomba, GP, Bertilsson, L. Homicide, suicide, and CSF 5-HIAA. Acta Psychiatr. Scand. 1985; 71(3): 230236.Google Scholar
Lester, D. The concentration of neurotransmitter metabolites in cerebrospinal fluid of suicidal individuals: a meta-analysis. Pharmacopsychiatry. 1995; 28(2): 4550.Google Scholar
Coccaro, EF, Kavoussi, RJ, Cooper, TB, Hauger, RL. Central serotonin activity and aggression: inverse relationship with prolactin response to d-fenfluramine, but not CSF 5-HIAA concentration, in human subjects. Am. J. Psychiatry. 1997; 154(10): 14301435.Google Scholar
Coccaro, EF, Kavoussi, RJ, Trestman, RL, et al. Serotonin function in human subjects: intercorrelations among central 5-HT indices and aggressiveness. Psychiatry Res. 1997; 73(1–2): 114.Google Scholar
Hibbeln, JR, Umhau, JC, George, DT, et al. Plasma total cholesterol concentrations do not predict cerebrospinal fluid neurotransmitter metabolites: implications for the biophysical role of highly unsaturated fatty acids. Am. J. Clin. Nutr. 2000; 71(1 Suppl): 331S338S.Google Scholar
Simeon, D, Stanley, B, Frances, A, et al. Self-mutilation in personality disorders: psychological and biological correlates. Am. J. Psychiatry. 1992; 149(2): 221226.Google Scholar
Coccaro, EF, Lee, R. Cerebrospinal fluid 5-hydroxyindolacetic acid and homovanillic acid: reciprocal relationships with impulsive aggression in human subjects. J. Neural. Transm. 2010; 117(2): 241248.Google Scholar
Coccaro, EF, Siever, LJ, Klar, HM, et al. Serotonergic studies in patients with affective and personality disorders: correlates with suicidal and impulsive aggressive behavior. Arch. Gen. Psychiatry. 1989; 46(7): 587599.Google Scholar
Coccaro, EF, Lee, R, Kavoussi, RJ. Aggression, suicidality, and intermittent explosive disorder: serotonergic correlates in personality disorder and healthy control subjects. Neuropsychopharmacology. 2010; 35(2): 435444.Google Scholar
Suarez, EC, Krishnan, KR. The relation of free plasma tryptophan to anger, hostility, and aggression in a nonpatient sample of adult men and women. Ann. Behav. Med. 2006; 31(3): 254260.Google Scholar
Goveas, JS, Csernansky, JG, Coccaro, EF. Platelet serotonin content correlates inversely with life history of aggression in personality-disordered subjects. Psychiatry Res. 2004; 126(1): 2332.Google Scholar
Coccaro, E, Kavoussi, R, Sheline, Y, Lish, J, Csernansky, J. Impulsive aggression in personality disorder correlates with tritiated paroxetine binding in the platelet. Arch. Gen. Psychiatry. 1996; 53(6): 531536.Google Scholar
Coccaro, EF, Kavoussi, RJ, Sheline, YI, Berman, ME, Csernansky, JG. Impulsive aggression in personality disorder correlates with platelet 5-HT2A receptor binding. Neuropsychopharmacology. 1997; 16(3): 211216.Google Scholar
Coccaro, EF, Lee, R, Kavoussi, RJ. Inverse relationship between numbers of 5-HT transporter binding sites and life history of aggression and intermittent explosive disorder. J. Psychiatr. Res. 2009; 44(3): 137142.Google Scholar
Marseille, R, Lee, R, Coccaro, EF. Inter-relationship between different platelet measures of 5-HT and their relationship to aggression in human subjects. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2012; 36(2): 277281.Google Scholar
New, AS, Trestman, RL, Mitropoulou, V, et al. Serotonergic function and self-injurious behavior in personality disorder patients. Psychiatry Res. 1997; 69(1): 1726.Google Scholar
Paris, J, Zweig-Frank, H, Kin, NM, et al. Neurobiological correlates of diagnosis and underlying traits in patients with borderline personality disorder compared with normal controls. Psychiatry Res. 2004; 121(3): 239252.Google Scholar
O’Keane, V, Moloney, E, O’Neill, H, et al. Blunted prolactin responses to d-fenfluramine in sociopathy: evidence for subsensitivity of central serotonergic function. Br. J. Psychiatry. 1992; 160(5): 643646.Google Scholar
Moeller, FG, Steinberg, JL, Petty, F, et al. Serotonin and impulsive/aggressive behavior in cocaine dependent subjects. Prog. Neuropsychopharmacol. Biol. Psychiatry. 1994; 18(6): 10271035.Google Scholar
Moss, HB, Yao, JK, Panzak, GL. Serotonergic responsivity and behavioral dimensions in antisocial personality disorder with substance abuse. Biol. Psychiatry. 1990; 28(4): 325338.Google Scholar
Coccaro, EF, Berman, ME, Kavoussi, RJ, Hauger, RL. Relationship of prolactin response to d-fenfluramine to behavioral and questionnaire assessments of aggression in personality-disordered men. Biol. Psychiatry. 1996; 40(3): 157164.Google Scholar
Moeller, FG, Allen, T, Cherek, DR, et al. Ipsapirone neuroendocrine challenge: relationship to aggression as measured in the human laboratory. Psychiatry Res. 1998; 81(1): 3138.Google Scholar
Pihl, RO, Young, SN, Harden, P, et al. Acute effect of altered tryptophan levels and alcohol on aggression in normal human males. Psychopharmacology (Berl.). 1995; 119(4): 353360.Google Scholar
Cleare, AJ, Bond, AJ. The effect of tryptophan depletion and enhancement on subjective and behavioural aggression in normal male subjects. Psychopharmacology (Berl.). 1995; 118(1): 7281.Google Scholar
Dougherty, DM, Bjork, JM, Marsh, D, Moeller, FG. Influence of trait hostility on tryptophan depletion-induced laboratory aggression. Psychiatry Res. 1999; 88(3): 227232.Google Scholar
Linnoila, M, Virkkunen, M, Scheinin, M, et al. Low cerebrospinal fluid 5-hydroxyindoleacetic acid concentration differentiates impulsive from nonimpulsive violent behavior. Life Sci. 1983; 33(26): 26092614.Google Scholar
Spoont, MR. Modulatory role of serotonin in neural information processing: implications for human psychopathology. Psychol. Bull. 1992; 112(2): 330350.Google Scholar
Linnoila, VM, Virkkunen, M. Aggression, suicidality, and serotonin. J. Clin. Psychiatry. 1992; 53(Suppl): 4651.Google Scholar
Coccaro, EF, Kavoussi, RJ, Lesser, JC. Self- and other-directed human aggression: the role of the central serotonergic system. Int. Clin. Psychopharmacol. 1992; 6(Suppl 6): 7083.Google Scholar
Berman, ME, McCloskey, MS, Fanning, JR, Schumacher, JA, Coccaro, EF. Serotonin augmentation reduces response to attack in aggressive individuals. Psychol. Sci. 2009; 20(6): 714720.Google Scholar
Marks, DJ, Miller, SR, Schulz, KP, Newcorn, JH, Halperin, JM. The interaction of psychosocial adversity and biological risk in childhood aggression. Psychiatry Res. 2007; 151(3): 221230.Google Scholar
Duke, AA, Bègue, L, Bell, R, Eisenlohr-Moul, T. Revisiting the serotonin-aggression relation in humans: a meta-analysis. Psychol. Bull. 2013; 139(5): 11481172.Google Scholar
Cleare, AJ, Bond, AJ. Ipsapirone challenge in aggressive men shows an inverse correlation between 5-HT1A receptor function and aggression. Psychopharmacology (Berl.). 2000; 148(4): 344349.Google Scholar
Almeida, M, Lee, R, Coccaro, EF. Cortisol responses to ipsapirone challenge correlate with aggression, while basal cortisol levels correlate with impulsivity, in personality disorder and healthy volunteer subjects. J. Psychiatr. Res. 2010; 44(14): 874880.Google Scholar
Bortolato, M, Pivac, N, Muck Seler, D, et al. The role of the serotonergic system at the interface of aggression and suicide. Neuroscience. 2013; 236: 160185.Google Scholar
Olivier, B, van Oorschot, R. 5-HT1B receptors and aggression: a review. Eur. J. Pharmacol. 2005; 526(1–3): 207217.Google Scholar
Meyer, JH,Wilson, AA, Rusjan, P, et al. Serotonin2A receptor binding potential in people with aggressive and violent behaviour. J. Psychiatry Neurosci. 2008; 33(6): 499508.Google Scholar
Soloff, PH, Price, JC, Mason, NS, Becker, C, Meltzer, CC. Gender, personality, and serotonin-2A receptor binding in healthy subjects. Psychiatry Research: Neuroimaging. 2010; 181(1): 7784.Google Scholar
Rosell, DR, Thompson, JL, Slifstein, M, et al. Increased serotonin 2A receptor availability in the orbitofrontal cortex of physically aggressive personality disordered patients. Biol. Psychiatry. 2010; 67(12): 11541162.Google Scholar
Siever, LJ, Buchsbaum, MS, New, AS, et al. d,l-fenfluramine response in impulsive personality disorder assessed with [18F] fluorodeoxyglucose positron emission tomography. Neuropsychopharmacology. 1999; 20(5): 413423.Google Scholar
New, AS, Hazlett, EA, Buchsbaum, MS, et al. Blunted prefrontal cortical 18fluorodeoxyglucose positron emission tomography response to meta-chlorophenylpiperazine in impulsive aggression. Arch. Gen. Psychiatry. 2002; 59(7): 621629.Google Scholar
New, AS, Buchsbaum, MS, Hazlett, EA, et al. Fluoxetine increases relative metabolic rate in prefrontal cortex in impulsive aggression. Psychopharmacology (Berl.). 2004; 176(3–4): 451458.Google Scholar
Leyton, M, Okazawa, H, Diksic, M, et al. Brain regional alpha-[11C] methyl-L-tryptophan trapping in impulsive subjects with borderline personality disorder. Am. J. Psychiatry. 2001; 158(5): 775782.Google Scholar
Frankle, WG, Lombardo, I, New, AS, et al. Brain serotonin transporter distribution in subjects with impulsive aggressivity: a positron emission study with [11C]McN 5652. Am. J. Psychiatry. 2005; 162(5): 915923.Google Scholar
Koch, W, Schaaff, N, Pöpperl, G, et al. [I-123] ADAM and SPECT in patients with borderline personality disorder and healthy control subjects. J. Psychiatry Neurosci. 2007; 32(4): 234240.Google Scholar
Coccaro, EF, Lee, R, Kavoussi, RJ. A double-blind, randomized, placebo-controlled trial of fluoxetine in patients with intermittent explosive disorder. J. Clin. Psychiatry. 2009; 70(5): 653662.Google Scholar
George, DT, Phillips, MJ, Lifshitz, M, et al. Fluoxetine treatment of alcoholic perpetrators of domestic violence: a 12-week, double-blind, randomized, placebo-controlled intervention study. J. Clin. Psychiatry. 2011; 72(1): 6065.Google Scholar
Silva, H, Iturra, P, Solari, A, et al. Fluoxetine response in impulsive-aggressive behavior and serotonin transporter polymorphism in personality disorder. Psychiatr. Genet. 2010; 20(1): 2530.Google Scholar
Heiligenstein, JH, Coccaro, EJ, Potvin, JH, et al. Fluoxetine not associated with increased violence or aggression in controlled clinical trials. Ann. Clin. Psychiatry. 1992; 4(4): 285295.Google Scholar
Salzman, C, Wolfson, AN, Schatzberg, A, et al. Effect of fluoxetine on anger in symptomatic volunteers with borderline personality disorder. J. Clin. Psychopharmacol. 1995; 15(1): 2329.Google Scholar
Coccaro, EF, Kavoussi, RJ, Hauger, RL. Serotonin function and antiaggressive response to fluoxetine: a pilot study. Biol. Psychiatry. 1997; 42(7): 546552.Google Scholar
Coccaro, EF, Sripada, CS, Yanowitch, RN, Phan, KL. Corticolimbic function in impulsive aggressive behavior. Biol. Psychiatry. 2011; 69(12): 11531159.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×