The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Reviews and OverviewsFull Access

A Brain-Centric Model of Suicidal Behavior

The suicide death toll is alarmingly high, outnumbering deaths from war and homicide worldwide (1). Many factors contribute to suicide death, making it difficult to prevent because it is much more than a response to life stressors. Campaigns to lower U.S. suicide rates by 20% by 2025, and even to zero, confront hard realities such as not knowing why the annual suicide rate in the United States has risen every year since 2000 (2). The concurrent fall in suicide rates worldwide (1) is largely driven by restricting access to one highly lethal suicide method, namely, pesticides, which are commonly used in a few very populous countries, including China, India, and Sri Lanka (35). That approach saves lives without addressing the reasons people want to die by suicide. Currently empirically proven suicide prevention methods (Figure 1) include 1) better training of nonpsychiatrist physicians in the diagnosis and treatment of depression, the most common psychiatric disorder associated with suicide (6); 2) better outreach during the highest risk period, after suicidal patients are discharged from hospital or the emergency department (7); and 3) means restriction, focused on the commonest methods in each country (8). To prevent suicide in the short term, we need to scale up the use of these proven methods. To use these methods more effectively, we need to know who is at risk and when the risk is greater. Progress in both domains has stalled.

FIGURE 1.

FIGURE 1. The stress-diathesis model of suicidal behaviora

a CBT=cognitive-behavioral therapy; BDNF=brain-derived neurotrophic factor; dACC=dorsal anterior cingulate cortex; DBT=dialectical behavioral therapy; dlPFC=dorsolateral prefrontal cortex; dmPFC=dorsomedial prefrontal cortex; DST=dexamethasone suppression test; GR=glucocorticoid receptor; HPA axis=hypothalamic-pituitary-adrenal axis; IFG=inferior frontal gyrus; mOFC=medial orbitofrontal cortex; MR=mineralocorticoid receptor; OFC=orbitofrontal cortex; rACC=rostral anterior cingulate cortex; rTMS=repetitive transcranial magnetic stimulation; SNRIs=serotonin-norepinephrine reuptake inhibitors; SSRIs=selective serotonin reuptake inhibitors; tDCS=transcranial direct current stimulation; TrkB=Tropomyosin-related kinase B; TSST=Trier Social Stress Test; vlPFC=ventrolateral prefrontal cortex; vmPFC=ventromedial prefrontal cortex.

Rehashing older approaches does not improve suicide risk assessment or prevention (9). While universal approaches like training primary care physicians to manage depression and means restriction work (10), for targeted prevention approaches we need to find new risk detection methods. The explosion of new information about the causes of suicidal behavior (11) needs to be built into a heuristic descriptive model that integrates potential predictors and prevention targets with a hypothetical explanatory model. A model for suicidal behavior must reconcile two major observations. First, suicidal behavior is found in many psychiatric disorders and not just in mood disorders and borderline personality disorder, where it is part of the diagnostic symptom list. Second, only a minority of psychiatric patients make suicide attempts, and the presence or absence of suicidal behavior is not simply determined by the presence of a psychiatric illness or the overall severity of illness (1214).

Suicidal ideation is much more prevalent than suicidal behavior, and it often does not progress to suicidal behavior (15). Suicide prevention measures might prevent the transitioning from ideation to behavior if we understood the factors involved (13, 16). Suicidal behavior risk is moderated by a set of psychopathological and cognitive traits that are different from the psychopathology of associated psychiatric illnesses. We term the interaction of internal and external stressors with the traits that form a diathesis for suicide risk the stress-diathesis model of suicidal behavior (13, 14). In this model, we seek to integrate clinical and cognitive correlates of suicidal behavior with their neural circuitry and the biological basis of abnormalities in this circuitry to help explain the pathogenesis of the diathesis elements. We then link these elements to pharmacologic, psychotherapeutic, and brain modulatory approaches to suicide prevention (Figure 1).

Pathways to Suicidal Behavior: a Stress-Diathesis Model

The stress-diathesis model (13, 14) depicts suicidal behavior as a consequence of an interaction between acute stressors or proximal risk factors and a diathesis or distal factors (Figure 1). Stressors can be external, in the form of relationship or financial problems, or internal, such as a major depressive episode. The diathesis refers to a set of suicide-related traits that moderates the likelihood of suicidal behavior in response to stressors. The neurobiological correlates of these traits may be potential biomarkers for suicide risk, separate from biomarkers of co-occurring psychiatric disorders. Identified suicide-related, risk-moderating traits (Figure 1) include 1) excessive subjective distress when depressed and attentional bias toward negative stimuli; 2) altered decision making involving less delayed discounting and less executive control resulting in impulsive-aggressive tendencies and favoring acting on emotions; 3) an array of neuropsychological abnormalities, including learning difficulties, cognitive rigidity, and memory problems; and 4) social distortions (1726).

  1. Subjective distress is linked to familial transmission of suicidal behavior and encompasses hopelessness and subjective depression, which are more severe in depressed suicide attempters compared with depressed nonattempters (27). Others use terms like psych-ache and emotional pain to refer to a similar phenomenon (28, 29). Joiner’s tripartite model of suicidal behavior (30) describes a tolerance to the pain involved in suicide, sometimes developed by prior nonfatal suicide attempts, that could encompass weighing the emotional pain being suffered against the anticipated pain of a suicide attempt. Other models of suicide, like the integrated motivational-volitional model (16), examine the threshold between ideation and action, also emphasizing the role of emotion and cognition.

  2. Decision making is multifaceted. Linked to suicidal behavior are impaired probabilistic learning assessed by monetary reward and punishments (31), risky decision tendency under uncertainty (32), and a bias for active responding when escaping an aversive state (33). Decision making differs in high-lethality suicide attempters relative to low-lethality attempters because the former are generally more deliberate and planned. High-lethality attempters have greater impairment of reward learning, hampering the search for alternative solutions (34), but they have less delayed discounting (i.e., they can suppress the desire for immediate satisfaction in favor of delayed reward) (35) and perform better on an object alternation task, indicating greater executive control and response organization (25). Impaired decision making (36) and related brain circuit changes (37) are also observed in healthy relatives of suicide decedents, consistent with familial transmission of an endophenotype for suicide risk.

  3. Suicide attempters exhibit less language fluency and learning (38) and impaired deterministic learning in the context of complex unambiguous environments (39). These learning deficits scale with attempt lethality and are partially explained by impaired cognitive control (34), the most consistent deficit found in suicide attempters (17, 22, 24). Memory deficits in suicide attempters include impaired working memory and long-term memory and a shift from specific to more general autobiographical memory (18, 23).

  4. Suicide risk is increased by cognitive distortions related to social reward and exclusion (40). A perceived lack of reciprocally caring relationships, also known as thwarted belongingness, may deter help-seeking (41), amplify sensitivity to social rejection (42), and impair social integration (43), and all enhance suicidal ideation and behavior.

Genetic and Epigenetic Causes of the Diathesis in the Stress-Diathesis Model

Human genetic studies have made three major observations: 1) heritability of fatal or nonfatal suicidal behavior and even ideation is moderate; 2) heritability of suicidal behavior is independent of major psychiatric syndromes; and 3) heritability of suicidal behavior is mediated by many gene variants of small effect (4447). After candidate gene studies stalled because effects were too small and difficult to replicate, rapidly expanding sample sizes for genome-wide association studies (GWASs) have identified some suicide-specific candidate genes that also mostly remain to be replicated (48, 49). A recent review reported 40 genes associated with suicidal behavior, independently of psychiatric diagnoses (50). Combining GWASs with transcriptome data indicates that suicide-related genes are associated with inflammation, the hypothalamic-pituitary-adrenal (HPA) axis, γ-aminobutyric acid (GABA) and glutamate neurotransmission, and neurogenesis (5156). Severity of suicide attempts is associated with genes involved in anerobic energy production, circadian clock regulation, and tyrosine catabolism (57). Suicide risk may be mediated by genetic and epigenetic effects of stress response genes (5861). Altered genome-wide DNA methylation reveals alteration at several genetic loci in the prefrontal cortex (PFC) and cerebellum, and more globally in the brain in suicide decedents compared with control subjects (6264).

Neurobiological Correlates of Suicide Risk–Related Diathesis Traits

Genetic and epigenetic effects are mediated through biological effects that are currently being discovered (Figure 1). The four major suicide risk–moderating trait domains are linked to different combinations of neural circuitry and neurotransmitter system abnormalities. These, in turn, are the results of altered stress responses, trophic/apoptotic effects, and inflammation.

Neural Circuitry of Suicidal Behavior and Suicidal Ideation

Although neuroimaging studies of suicidal behavior and ideation disagree about details, the main features of dysfunctional neural circuitry (Figure 1) include 1) a relationship of enhanced negative affective and self-referential processing networks (ventromedial prefrontal cortex [vmPFC], medial orbitofrontal cortex [OFC], rostral anterior cingulate cortex [rACC], insula, and ventral striatum) to suicidal ideation (65) that may also underlie excessive subjective distress; 2) structural and functional deficits in the dorsomedial prefrontal cortex (dmPFC), dorsolateral prefrontal cortex (dlPFC), ventrolateral prefrontal cortex (vlPFC), and dorsal anterior cingulate cortex (dACC) that correlate with severity of subjective depression (66) and contribute to less top-down control over vmPFC regions, resulting in impaired decision making and, in turn, suicidal behavior (67); 3) differential activation of the OFC to pleasant versus negative facial expressions, perhaps related to excessive distress and social distortions; 4) serotonergic release deficits in the ventral PFC and ACC that are more prominent in high-lethality suicidal behavior and suicide death; and 5) abnormalities of glutamate and opioid systems that may affect memory, learning, and reward mechanisms.

Structural Brain Findings.

Suicide decedents have smaller gray matter volume in the dlPFC (68) and hippocampus (69, 70), perhaps related to impaired decision making and learning/memory functions, respectively. Gray matter volume changes in nonfatal suicidal behavior are less consistently reported (14, 71, 72) and may be confined to subgroups of suicide attempters. Higher-lethality suicide attempters have larger PFC and insula volumes compared with lower-lethality attempters and nonattempters (73), potentially related to superior planning abilities. Violent attempters have greater caudate volume, and suicide attempters with familial suicidal behavior have smaller volumes in temporal regions, dlPFC, and putamen, compared with other subgroups of attempters (72). Adolescent suicidal behavior mostly involves smaller ventral PFC regions, particularly the OFC (74), which correlates with greater attempt lethality (75).

Diffusion tensor imaging (DTI) examines fractional anisotropy (FA) as an index of white matter integrity. In mood disorders, a prior suicide attempt is associated with lower FA in frontal-subcortical connections (7679). Such findings may indicate impaired top-down executive control, explaining a correlation with attempt-related impulsivity (80) and number of suicide attempts (81, 82). DTI studies of suicidal ideation are inconclusive (77, 83, 84).

Functional Brain Activity and Connectivity.

Resting-state functional MRI (rs-fMRI) indicates lower functional connectivity in adolescent and adult suicide attempters, predominantly in the default mode network, which is implicated in self-referential processing and social cognition (8587). This functional circuitry deficit maps onto the DTI-identified structural connectivity deficits in default mode network hubs (e.g., vmPFC) and connections (e.g., genu of corpus callosum) (8082). Suicidal ideation relates to greater rs-fMRI activity in the vmPFC (88) and greater resting functional connectivity between the OFC and amygdala (89) and between the posterior cingulate cortex and middle temporal gyrus (90), perhaps related to negative self-reflection or rumination (91). However, other studies have reported lower rs-fMRI activity and functional connectivity (85, 86, 92, 93), probably due to use of different outcome measures of rs-fMRI activity, for example, regional homogeneity, fractional amplitude of low-frequency fluctuations among studies, and heterogeneity of suicidal ideation. Suicidal ideation–related increased frontal-subcortical connectivity may be confined to suicidal ideators who do not attempt suicide (94), because better executive control prevents acting on suicidal thoughts.

Task-based fMRI studies add the specificity of a task but also add variance as a result of the details of task implementation. Task-based fMRI studies are able to identify deficits in brain regions related to the suicide diathesis. Compared with suicide nonattempters, suicide attempters having the same diagnosis manifest less activity in the dlPFC, rACC, dACC, thalamus, and insula while performing decision-making tasks (e.g., the Iowa Gambling Task) (9599). Lower activity in the dACC and dlPFC is also detected in attempters during tasks evaluating executive function (e.g., the go/no-go task) (100). Recall of suicide episodes (mental pain and suicide action) is associated with less activity in the dlPFC and dmPFC and greater activity in the parahippocampal gyrus and cuneus (101). Immersion and reappraisal of negative autobiographical memories are associated with greater OFC activity in suicide attempters (102). Suicide attempters have lower activity in the supramarginal gyrus and posterior insula during the cyberball task, an fMRI experiment evoking reaction to social exclusion (103), perhaps reflecting distorted social perception. Suicide attempters also have less OFC and rACC activity in response to positive facial expressions (78) and greater lateral OFC activity in response to angry facial expressions (95, 97, 104). This may underlie excessive subjective distress in suicide attempters and may also explain poorer help-seeking (41), susceptibility to bullying (105), and admonishment from parents or teachers (106), all of which are suicide risk factors.

Neurotransmitter Systems.

Deficient serotonin release as a potential suicide biomarker was discovered over 40 years ago (107). Low cerebrospinal fluid (CSF) serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) predicts suicide risk in depressed individuals, with an odds ratio of 4.6 (see reference 108 for a review). Deficient serotonin release is not due to serotonin biosynthesis, because suicide decedents have greater tryptophan hydroxylase 2 (the rate-limiting enzyme of serotonin synthesis) expression per neuron, and there are more brainstem serotonin neuron cell bodies (109, 110). The serotonin deficit likely involves less serotonin neuron firing and less release, because fatal and nonfatal suicide attempts are associated with up-regulated serotonin 1A autoreceptors that are located on serotonin neuron cell bodies and proximal dendrites (111, 112). Mouse studies show that greater expression of these autoreceptors produces a biological phenotype of less serotonin neuron firing and release and a behavioral phenotype manifesting giving-up behavior in the tail suspension test and the forced swim test (113). Other studies report up-regulated postsynaptic cortical serotonin 2A receptors that correlate with lifetime aggression severity (114). Less presynaptic serotonin transporter binding in major depression is seen throughout most of the PFC, but in suicide an additional deficit is confined to the ventral PFC and ACC, suggesting a role in decision making (115, 116). Serotonin dysfunction is more prominent in higher-lethality suicidal behavior (117), indicating localized PFC hypofunction (118). Greater raphe nucleus serotonin 1A receptor binding correlates with prior and future higher-lethality suicidal behavior (112, 119). Up-regulated serotonin 1A receptors may result from a 5-HT1A gene promoter polymorphism that causes overexpression of autoreceptors as a result of less binding of inhibitory transcription factors in serotonin neurons (120, 121). Childhood adversity may produce a phenocopy by epigenetic effects in the promotor and thereby mediate the effect on suicidal behavior risk via less serotonin release (108). Complex relationships also exist between the serotonin system and stress response. Childhood adversity may both up-regulate HPA stress responses (122) and reduce serotonin neuron firing (108) in parallel (123), the former amplifying neuroinflammation (124) and the latter dampening neurotrophic factors (125).

Less is known about the role of norepinephrine in suicide risk (reviewed in reference 126). Depressed suicide decedents have fewer brain-projecting noradrenergic neurons in the rostral locus coeruleus and higher β-adrenergic receptor binding in the PFC, both consistent with less noradrenergic activity. Moreover, depressed suicide attempters have lower urinary and plasma levels of 3-methoxy-4-hydroxyphenylglycol (MHGP), a major metabolite of norepinephrine, compared with control subjects, and CSF 3-methoxy-4-hydroxphenylglycol (MHPG) levels correlate negatively with lethality of future suicide attempts (127). Childhood adversity may sensitize norepinephrine release in response to a stressor in adulthood, as shown in rodents (128). Perhaps this excessive norepinephrine release, when combined with fewer noradrenergic neurons, is more likely to result in a depletion of norepinephrine and lower CSF MHPG in future suicide attempters (127).

Altered expression of glutamate receptor N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and kainite glutamate receptors is linked to suicidal behavior (129) and may play a role in the rapid-onset antidepressant and antisuicidal effects of ketamine (130135). Enhanced glutamatergic NMDA-mediated transmission may improve memory consolidation (136) but in excess may be neurotoxic (137). Expression of some glutamatergic genes is greater in suicide decedents with a history of major depression (138).

The kappa opioid receptor system may play a role in negative affect associated with suicidal ideation and may explain the possible antisuicidal effects of the kappa antagonist buprenorphine (139, 140). Kappa receptor agonists induce anxiogenic and dysphoric effects in humans (141143) and depressive-like behavior in animals (i.e., increased immobility in the forced swim test) (144, 145). Prodynorphin, the precursor to endogenous kappa opioid dynorphin, is elevated in the caudate nucleus in suicide decedents (146). Altered mu opioid receptor binding is also related to suicide risk (40, 147), perhaps via dysregulation of reward circuitry and social function (40).

Stress Response Systems

Response to stress involves a more immediate short-term noradrenergic system response and a more enduring HPA axis response. Feedback inhibition of resting cortisol level is through high-affinity mineralocorticoid receptors, and lower-affinity glucocorticoid receptors regulate stress-induced peaks of cortisol (123). Glucocorticoid receptors are subject to genetic effects via chaperone proteins like SKA1 and FKBP5 (58, 148, 149), as well as epigenetic changes (i.e., methylation) observed in response to maternal deprivation in rats (150) and in suicide decedents with a history of childhood adversity (60). Nonsuppression on the dexamethasone suppression test, which assesses this HPA axis glucocorticoid receptor feedback loop, predicts suicide (151153), but there is disagreement about nonfatal attempts (154158). Laboratory-induced psychosocial stressors, like the Trier Social Stress Test (TSST) (159), evaluate HPA axis response to social and environmental stressors (160). TSST studies find lower pretask baseline cortisol (161, 162) and blunted total cortisol output (162, 163) in suicide attempters compared with nonattempters, while heightened cortisol response to stress appears confined to suicide attempters exhibiting higher levels of impulsivity and aggression (164). Mixed results exist for the association between HPA axis abnormalities and suicidal ideation (163, 165167). Some authors attribute these inconsistencies to the heterogeneity of patterns and severity of suicidal thoughts, such that fluctuating patterns of suicidal ideation are associated with greater cortisol response to stress compared with more stable and chronic ideation patterns (168).

Neurotrophic and Apoptotic Pathways

Brain-derived neurotrophic factor (BDNF) regulates neuron survival, plasticity, and synaptic function (169174), plays an integral role in differentiation during development (175, 176), is affected by stress (177), and is associated with major depression and suicidal behavior (178). Expression of BDNF and BDNF receptor tyrosine kinase B genes is lower in suicide decedents (179181), and plasma BDNF levels are low in suicide attempters (182), perhaps reflecting a systemic genomic effect on BDNF expression. The BDNF gene Met allele, a part of BDNF functional polymorphism (rs6265), increases the risk for suicidal behavior (183, 184), particularly in depressed individuals (185) and in those exposed to early-life stress (186, 187), where it may do so by loss of ACC volume (188) and altered decision-making or depression circuitry. BDNF promoter/exon IV, which plays a critical role in BDNF gene regulation (189), is hypermethylated in Wernicke’s area in suicide decedents (190). Infant maltreatment increases DNA methylation and reduces BDNF gene expression in the PFC (191). Further, epigenetic regulation of TrkB-T1 (an astrocyte-specific variant of the BDNF receptor) is reported in suicide decedents through promoter methylation affecting regulation by miRNA miR-185 (192, 193). Low brain BDNF in the hippocampus of suicide decedents (180) may underlie its smaller size in suicide decedents (69) and attempters (194) and fewer mature granule neurons in its dentate gyrus (70). This may partly explain learning and memory components of suicide diathesis. Moreover, the dentate gyrus plays a role in emotion regulation through its connection to the PFC—which also shows lower neuron density and serotonin receptor binding in suicide decedents (68). These suicide risk–related neuron deficits may also be attributed to toxic effects of the excessive allostatic load due to glucocorticoids and abnormal stress response (137). Elevated glucocorticoids may result in loss of astrocytes and glutamate transporters. This, in turn, leads to a pathological accumulation of glutamate, with subsequent loss of neuronal density (137).

Neuroinflammation

Peripheral inflammatory biomarkers linked to suicide risk include elevated C-reactive protein (CRP), neutrophil-to-lymphocyte ratio, proinflammatory interleukins (IL), cytokines that regulate the immune response (e.g., IL-1β and IL-6), tumor necrosis factor-α (TNF-α), tissue growth factor-β1 (TGF-β1), and vascular endothelial growth factor (VEGF) and low levels of the anti-inflammatory IL-2, IL-4, and interferon-γ (IFN-γ) in suicidal individuals (generally suicide attempters) (195202). Elevated IL-6 correlates positively with suicide attempters’ impulsivity (203), thereby linking proinflammatory responses to an impulsive, reactive endophenotype for suicidal behavior. IL-1β negatively correlates, whereas IL-2 positively correlates, with OFC activation during experimental social exclusion (204), a known trigger of suicidal ideation and behavior. Quinolinic and kynurenic acids (which serve as an agonist and an antagonist of the excitotoxic glutamate receptors, respectively) are related to suicidal behavior (205207). Activated microglia (208, 209), the primary immune response cells in the brain, and brain translocator protein (TSPO) found in mitochondria of activated glial cells (210) are linked to suicide risk, independently of the presence of psychiatric disorders. Chronic inflammatory processes associated with latent Toxoplasma gondii (T. gondii) infection may increase suicide risk (211, 212), perhaps through amplification of impulsive aggressive traits (213), or accumulation of T. gondii in the amygdala (214), which is involved in emotional and physiological responses to stress.

Greater suicidal ideation severity is associated with higher CRP (197), IL-6, and IL-10 levels (215) and with activated microglia-related alterations of the tryptophan-kynurenine pathway (216). This suggests that regulation of inflammatory pathways may change with level of suicidal ideation and therefore risk (208). However, a recent study reports no association between suicidal ideation severity and cytokine pathway marker mRNA expression (217), and longitudinal studies are needed to determine whether fluctuating suicide risk correlates with severity of inflammation.

Stress-Diathesis-Model-Informed Suicide Prevention

Approaches targeting different components of the stress-diathesis model can help suicide prevention and treatment efforts. Targets for suicide prevention may be found in the domain of the stressors or in the processing and response to the stressors that involve the diathesis component (Figure 1), and include the perception of the stressor, the formulation of the response, and the actual response. The treatment or prevention target may be described in clinical or cognitive terms or in neurobiological or circuitry terms (Figure 1).

Stressors and Prevention

The amelioration of external stressors may be the goal of interventions by mental health professionals and social workers seeking to improve personal, family, and workplace relationships and to improve housing, nutrition, and employment status, but these approaches have not been shown to prospectively influence risk of suicidal behavior (10). Targeting internal stressors by training primary care providers in the management of major depressive episodes prevents suicide (6). Major depressive episodes account for about 60% of psychiatric diagnoses in suicide decedents (218), and major depressive episodes are mostly untreated at the time of death (219).

Diathesis and Prevention

Pharmacotherapy.

Antidepressants generally act on serotonergic and noradrenergic systems (220) and may help prevent suicide by effects on both the internal stressors of depression and anxiety and the diathesis by reducing hopelessness, reducing propensity for impulsive behavior in response to aggressive or suicidal urges and enhancing cognition. Lithium (221, 222) and clozapine (223, 224) have effects on the internal stressors of depression and acute psychosis. Because both medications reduce the risk of suicidal behavior independently of how effective they are for mood and psychosis (225), this indicates potential effects via the diathesis. The specific mechanisms of their antisuicidal diathesis effects are unknown and are not readily discoverable because lithium and clozapine have many different pharmacologic effects. Antiapoptotic effects of lithium (226) may increase gray matter volume in the dlPFC, where hypoactivity and cortical thinning are implicated in suicide risk. Clozapine blocks dopamine 2 and serotonin 2A receptors, potentially mediating antiaggressive and anti-impulsive properties (227).

Intravenous ketamine is a rapidly acting treatment that robustly reduces depressive symptoms and suicidal ideation in hours instead of weeks (130133). Intranasal ketamine (or esketamine) also shows promise as anti–suicidal ideation treatment (134) and is less invasive, but its absorption is more erratic, and, like intravenous ketamine, it must be administered in medical settings. The anti–suicidal ideation effect of ketamine is only partly explained by its antidepressant action, and effects on memory and cognition may also play a role (131133). The relative importance of ketamine diverting glutamatergic transmission to AMPA from NMDA glutamate receptors, opioid system activation (228), alteration of BDNF release (229), or up-regulation of insulin-like growth factor 2 in the hippocampus (230) in its lowering suicidal ideation is unknown. Ketamine has never been tested in terms of preventing suicidal behavior.

Psychotherapy.

Most studied are cognitive-behavioral therapy (CBT) and dialectical behavior therapy, and both can prevent suicide attempts (231, 232). CBT improves capacities for cognitive regulation of emotion and is associated with decreased activity in the amygdala-associated negative emotional reactivity (233) and enhanced activity of the emotion regulation network, including the subgenual ACC, medial PFC, and lingual gyrus (234). Dialectical behavior therapy prevents suicidal behavior without proven antidepressant benefit, indicating that it works via problem-solving and stress management or elements of the diathesis such as dampening amygdala reactivity to negative emotions (235).

Brief Interventions and Active Postdischarge Outreach in the Emergency Department or From Inpatient Units

After seeking help for suicidal thoughts or low-lethality suicide attempts in the emergency department, discharged patients have an elevated suicide attempt rate. The suicide death rate is 300 times higher in the first week and 200 times higher in the first month among patients discharged from inpatient psychiatric care compared with the general population (236). Suicide prediction models based on computational analyses of electronic health records may help identify individuals with the highest risk for suicide at time of discharge, the subgroup that would potentially benefit most from intensive treatment and follow-up (237, 238). A number of brief psychological and educational interventions are now available for use with individuals presenting to the emergency department with acute suicidal crisis (239). These brief interventions are inexpensive, easy to implement, and require limited staff resources. They seek to project a helpful option to a patient who gets into a crisis and overcome the social cognitive distortion that the social network is more hostile than helpful. Active outreach after discharge seeks the same goal, and both have been shown to reduce the postdischarge risk of suicidal behavior (7, 240242).

Restriction of Lethal Means

Means restriction works because it seeks to make access to the most popular higher-lethality methods more difficult. It takes advantage of two key observations. The acute risk of acting on suicidal thoughts is brief, and the flexibility of the suicidal person in changing from one method to another is surprisingly limited. Studies of survivors of suicide attempts indicate that most suicide attempts are the result of a decision to act that was made minutes earlier, even if the method was planned months or more ahead (243, 244). Switching methods is known to be difficult because restricting access to a common method—such as household coal gas in the United Kingdom (245247) and firearms in Switzerland (248)—was followed by large reductions in suicide rates, mainly due to those methods, and very little method substitution even over several years. Forcing individuals to use a less lethal method may work because over 80% of suicide attempt survivors do not end up dying by suicide (249). So even after the combination of stress and diathesis leads to a suicide attempt, there are still interventions that can help.

The Future of Research and Suicide Prevention

Suicide as a Distinct Mental Disorder

Suicide is moderately heritable independently of the heritability of major psychiatric disorders (45, 46). This suicide-specific heritability is associated with suicide diathesis–related traits (36, 250). Further, the body of observations linking the suicide-related diathesis components to neurobiological abnormalities as described above supports the idea that suicidal behavior is not merely a symptom of a subset of psychiatric disorders, but should be coded as a distinct mental disorder (251). As a distinct diagnosis, it will be coded more consistently in medical records, raising clinician awareness of risk in individual patients, enhancing suicide research, and motivating the targeting of the suicide-related diathesis for prevention and treatment in suicidal patients.

Real-Time Monitoring of Acute Suicidal Crisis

While many of the distal or trait risk and protective factors for suicide and their underlying biological processes are extensively studied, we know little of the very short-term risk factors. Recent technological advances have begun to elucidate factors associated with acute suicidal processes that were difficult to observe by conventional assessment measures. Suicidal ideation, which usually precedes and may represent an alarm for imminent risk of a suicide attempt, goes unrecognized by even systematic periodic evaluation of outpatients. Around 60% of individuals denying suicidal ideation on weekly self-report measures reported suicidal thoughts using an ecological momentary assessment (EMA) technology that delivered questions on their smartphones six times a day over the same 1-week period (252). EMA also identifies a subgroup of suicidal individuals who report a more highly variable severity pattern of suicidal thoughts and spikes of suicidal thoughts when stressed (253). This pattern of suicidal ideation variability is associated with mood instability (254, 255) and a heightened stress response system (168), and it is a stable trait because it persists for 2 years (256). The suicidal behavior in this reactive group is just as lethal as in patients with stable but pronounced suicidal ideation (257). Different patterns of suicidal ideation suggest that a more nuanced approach to risk detection and prevention is needed (258) because otherwise suicidal ideation has modest sensitivity and low predictive value for suicide (259).

Combining EMA-collected real-time data on suicidal thoughts and behavior with “passive sensing,” which collects data from individuals’ smartphones, allows examination of the very short-term risk factors of suicide in multiple domains: emotional distress (via acoustic voice, sentiment in communicative language [e.g., via social media, text messaging], facial expression [in selfies], and music choice), and social dysfunction (self and social content in communications and language, patterns of online communication [frequency and diversity of online contacts], and geographic movement) (260). The wide availability of smartphones allows accumulation of massive amounts of such personal-level data that may lead to identification of a “phonotype” (261) or “screenotype” (262). These approaches have promise, but they lack replicated data on prediction of suicidal behavior risk (263). If successful, they may permit calibration of suicide prevention to fluctuating levels of risk, but much work remains to be done before this becomes a reality. In addition, smartphones have introduced self-guided digital interventions as novel suicide prevention methods that may prove as effective as face-to-face interventions (264).

Implicit Cognitions and Neuroimaging for Suicide Risk Detection

Future suicide decedents may deny suicidal thoughts as inpatients (265), and more so in initial evaluations and emergency department settings, when patients and clinicians are less known to each other (266). This may be done intentionally to thwart any intervention to prevent suicide, or because the person does not perceive a need for care (267). Others may be unconsciously using the defense mechanism of denial regarding suicidal impulses (268). The death/suicide implicit association test (IAT) has been proposed in such situations. The IAT predicts suicidal behavior in some prospective studies (269, 270), but not others (271). Neural decoding is an approach where machine-learning methods have been used to identify the neural activity pattern involved in the mental representation of a deceased loved one and to track unconscious deceased-related thinking (272). fMRI-measured neural representation of mental concept representations differentiates suicidal ideators from suicide attempters and healthy control subjects (273). The identification of a neural signature of suicidal ideation could be a biomarker for suicide risk even when suicidal ideation is denied or unrecognized.

Medication and Neuromodulation

Buprenorphine, a mu opioid receptor partial agonist (274276) and a kappa opioid receptor antagonist (277), is used for relapse prevention in opioid use disorder (278). Buprenorphine has antidepressant effects when used for both opioid use disorder and treatment-resistant depression (279284) and may also reduce suicidal ideation (139, 140, 283, 285, 286). Compared with placebo, low-dose buprenorphine reduces suicidal ideation in 2 weeks in individuals who have never used opioids (140), probably through modulating emotional pain/psych-ache. Few case reports demonstrate similar effect in individuals with opioid use disorder using higher dosages (139, 285, 286). By addressing both opioid relapse and comorbid depression and suicidality, buprenorphine should be further evaluated in reducing suicide risk via opioid overdose, which has been spiking in the United States over the past two decades.

Neuroimaging-delineated brain regions and networks involved in suicide risk represent plausible targets for neuromodulatory interventions. Some researchers report a decrease of suicidal ideation following treatment with repetitive transcranial magnetic stimulation (rTMS) (287), but results are preliminary because most studies have small sample sizes and lack control groups. Inhibition of the dlPFC by low-frequency rTMS can increase risky decision making in males (288), suggesting that enhancing dlPFC activity may reduce suicide risk by improved top-down cognitive control. Transcranial direct current stimulation (tDCS) is another promising, low-risk, noninvasive neuromodulation technique that has effects on inhibitory control, planning, delay discounting, and risk taking in healthy individuals (289291). It remains to be seen whether tDCS applied to the PFC can modulate impulsivity and suicide risk (292).

The role of inflammation in the pathogenesis of depression and suicidal behavior suggests use of anti-inflammatory drugs for depressive symptoms and suicidal ideation. None have been shown to improve suicidal ideation, but minocycline decreases microglial activation in the PFC and depressive and anxiety-like traits in animals (293) and may reduce depressive symptoms (294). Celecoxib, a selective inhibitor of the cyclo-oxygenase 2 enzyme, may be effective as an add-on treatment for unipolar depression (295). Glycogen synthase kinase-3 (GSK3), an enzyme that promotes inflammation, is linked to animal stress and aggression models, is elevated in the ventral PFC of depressed suicide decedents, and is inhibited by lithium (296).

No drugs targeting HPA axis dysregulation have been evaluated for suicide prevention. The glucocorticoid receptor antagonist mifepristone (297) and cortisol synthesis inhibitors such as metyrapone and ketoconazole (298) do not show distinct benefit for depression but may warrant testing in suicidal individuals with glucocorticoid hypofunction.

Conclusions

New suicide prevention targets may be identified in seeking clinical and cognitive correlates of suicidal behavior and by an understanding of the altered brain circuitry and pathogenesis underlying the stress-diathesis model of suicidal behavior. The goal is to enhance identification of high-risk patients and to determine when their risk spikes in order to focus prevention on those patients at those times. A biological approach offers biomarkers to guide a personalized medical approach to prevention of suicide, a way to track fluctuating risk, and a way to track treatment effects. Such biomarkers assume greater importance if they can detect risk when very suicidal patients choose to withhold information of a plan and intent to suicide in order to avoid being thwarted.

Department of Psychiatry, Columbia University Irving Medical Center, Columbia University, New York (Mann, Rizk); Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York (Mann, Rizk); Department of Radiology, Columbia University Irving Medical Center, New York (Mann).
Send correspondence to Dr. Mann ().

Supported by NIMH grant P50MH090964 (principal investigator, Dr. Mann).

Dr. Mann receives royalties for commercial use of the Columbia-Suicide Severity Rating Scale from the Research Foundation for Mental Hygiene. Dr. Rizk reports no financial relationships with commercial interests.

Dr. Rizk is supported by the Paul Janssen Postdoctoral Fellowship in Translational Neuroscience from Columbia University and a NARSAD Young Investigator Award from the Brain and Behavior Research Foundation.

References

1 World Health Organization: World Health Statistics 2019: Monitoring Health for the SDGs. Geneva, World Health Organization, 2019Google Scholar

2 Centers for Disease Control and Prevention: National Center for Injury Prevention and Control: Web-Based Injury Statistics Query and Reporting System (WISQARS), 2020 (https://www.cdc.gov/injury/wisqars/index.html)Google Scholar

3 Gunnell D, Fernando R, Hewagama M, et al.: The impact of pesticide regulations on suicide in Sri Lanka. Int J Epidemiol 2007; 36:1235–1242Crossref, MedlineGoogle Scholar

4 Vijayakumar L, Satheesh-Babu R: Does “no pesticide” reduce suicides? Int J Soc Psychiatry 2009; 55:401–406Crossref, MedlineGoogle Scholar

5 Page A, Liu S, Gunnell D, et al.: Suicide by pesticide poisoning remains a priority for suicide prevention in China: analysis of national mortality trends 2006–2013. J Affect Disord 2017; 208:418–423Crossref, MedlineGoogle Scholar

6 Szanto K, Kalmar S, Hendin H, et al.: A suicide prevention program in a region with a very high suicide rate. Arch Gen Psychiatry 2007; 64:914–920Crossref, MedlineGoogle Scholar

7 Doupnik SK, Rudd B, Schmutte T, et al.: Association of suicide prevention interventions with subsequent suicide attempts, linkage to follow-up care, and depression symptoms for acute care settings: a systematic review and meta-analysis. JAMA Psychiatry (Online ahead of print, June 17, 2020)Google Scholar

8 Mann JJ, Michel CA: Prevention of firearm suicide in the United States: what works and what is possible. Am J Psychiatry 2016; 173:969–979LinkGoogle Scholar

9 Franklin JC, Ribeiro JD, Fox KR, et al.: Risk factors for suicidal thoughts and behaviors: a meta-analysis of 50 years of research. Psychol Bull 2017; 143:187–232Crossref, MedlineGoogle Scholar

10 Zalsman G, Hawton K, Wasserman D, et al.: Suicide prevention strategies revisited: 10-year systematic review. Lancet Psychiatry 2016; 3:646–659Crossref, MedlineGoogle Scholar

11 Astraud LP, Bridge JA, Jollant F: Thirty years of publications in suicidology: a bibliometric analysis. Arch Suicide Res (Online ahead of print, April 1, 2020)Google Scholar

12 Nordentoft M, Mortensen PB, Pedersen CB: Absolute risk of suicide after first hospital contact in mental disorder. Arch Gen Psychiatry 2011; 68:1058–1064Crossref, MedlineGoogle Scholar

13 Mann JJ, Waternaux C, Haas GL, et al.: Toward a clinical model of suicidal behavior in psychiatric patients. Am J Psychiatry 1999; 156:181–189LinkGoogle Scholar

14 van Heeringen K, Mann JJ: The neurobiology of suicide. Lancet Psychiatry 2014; 1:63–72Crossref, MedlineGoogle Scholar

15 Nock MK, Borges G, Bromet EJ, et al.: Cross-national prevalence and risk factors for suicidal ideation, plans, and attempts. Br J Psychiatry 2008; 192:98–105Crossref, MedlineGoogle Scholar

16 O’Connor RC, Kirtley OJ: The integrated motivational-volitional model of suicidal behaviour. Philos Trans R Soc Lond B Biol Sci 2018; 373:20170268Crossref, MedlineGoogle Scholar

17 Richard-Devantoy S, Berlim MT, Jollant F: A meta-analysis of neuropsychological markers of vulnerability to suicidal behavior in mood disorders. Psychol Med 2014; 44:1663–1673Crossref, MedlineGoogle Scholar

18 Richard-Devantoy S, Berlim MT, Jollant F: Suicidal behaviour and memory: a systematic review and meta-analysis. World J Biol Psychiatry 2015; 16:544–566Crossref, MedlineGoogle Scholar

19 Richard-Devantoy S, Ding Y, Turecki G, et al.: Attentional bias toward suicide-relevant information in suicide attempters: a cross-sectional study and a meta-analysis. J Affect Disord 2016; 196:101–108Crossref, MedlineGoogle Scholar

20 Richard-Devantoy S, Olié E, Guillaume S, et al.: Decision-making in unipolar or bipolar suicide attempters. J Affect Disord 2016; 190:128–136Crossref, MedlineGoogle Scholar

21 Keilp JG, Beers SR, Burke AK, et al.: Neuropsychological deficits in past suicide attempters with varying levels of depression severity. Psychol Med 2014; 44:2965–2974Crossref, MedlineGoogle Scholar

22 Keilp JG, Gorlyn M, Oquendo MA, et al.: Attention deficit in depressed suicide attempters. Psychiatry Res 2008; 159:7–17Crossref, MedlineGoogle Scholar

23 Keilp JG, Gorlyn M, Russell M, et al.: Neuropsychological function and suicidal behavior: attention control, memory, and executive dysfunction in suicide attempt. Psychol Med 2013; 43:539–551Crossref, MedlineGoogle Scholar

24 Keilp JG, Sackeim HA, Brodsky BS, et al.: Neuropsychological dysfunction in depressed suicide attempters. Am J Psychiatry 2001; 158:735–741LinkGoogle Scholar

25 Keilp JG, Wyatt G, Gorlyn M, et al.: Intact alternation performance in high lethality suicide attempters. Psychiatry Res 2014; 219:129–136Crossref, MedlineGoogle Scholar

26 Dombrovski AY, Hallquist MN: The decision neuroscience perspective on suicidal behavior: evidence and hypotheses. Curr Opin Psychiatry 2017; 30:7–14Crossref, MedlineGoogle Scholar

27 Mann JJ, Ellis SP, Currier D, et al.: Self-rated depression severity relative to clinician-rated depression severity: trait stability and potential role in familial transmission of suicidal behavior. Arch Suicide Res 2016; 20:412–425MedlineGoogle Scholar

28 Shneidman ES: Suicide as psychache. J Nerv Ment Dis 1993; 181:145–147Crossref, MedlineGoogle Scholar

29 Orbach I: Mental pain and suicide. Isr J Psychiatry Relat Sci 2003; 40:191–201MedlineGoogle Scholar

30 Van Orden KA, Witte TK, Cukrowicz KC, et al.: The interpersonal theory of suicide. Psychol Rev 2010; 117:575–600Crossref, MedlineGoogle Scholar

31 Bridge JA, Reynolds B, McBee-Strayer SM, et al.: Impulsive aggression, delay discounting, and adolescent suicide attempts: effects of current psychotropic medication use and family history of suicidal behavior. J Child Adolesc Psychopharmacol 2015; 25:114–123Crossref, MedlineGoogle Scholar

32 Ackerman JP, McBee-Strayer SM, Mendoza K, et al.: Risk-sensitive decision-making deficit in adolescent suicide attempters. J Child Adolesc Psychopharmacol 2015; 25:109–113Crossref, MedlineGoogle Scholar

33 Millner AJ, den Ouden HEM, Gershman SJ, et al.: Suicidal thoughts and behaviors are associated with an increased decision-making bias for active responses to escape aversive states. J Abnorm Psychol 2019; 128:106–118Crossref, MedlineGoogle Scholar

34 Dombrovski AY, Hallquist MN, Brown VM, et al.: Value-based choice, contingency learning, and suicidal behavior in mid- and late-life depression. Biol Psychiatry 2019; 85:506–516Crossref, MedlineGoogle Scholar

35 Dombrovski AY, Szanto K, Siegle GJ, et al.: Lethal forethought: delayed reward discounting differentiates high- and low-lethality suicide attempts in old age. Biol Psychiatry 2011; 70:138–144Crossref, MedlineGoogle Scholar

36 Hoehne A, Richard-Devantoy S, Ding Y, et al.: First-degree relatives of suicide completers may have impaired decision-making but functional cognitive control. J Psychiatr Res 2015; 68:192–197Crossref, MedlineGoogle Scholar

37 Ding Y, Pereira F, Hoehne A, et al.: Altered brain processing of decision-making in healthy first-degree biological relatives of suicide completers. Mol Psychiatry 2017; 22:1149–1154Crossref, MedlineGoogle Scholar

38 Huber RS, Hodgson R, Yurgelun-Todd DA: A qualitative systematic review of suicide behavior using the cognitive systems domain of the Research Domain Criteria (RDoC) framework. Psychiatry Res 2019; 282:112589Crossref, MedlineGoogle Scholar

39 McGirr A, Dombrovski AY, Butters MA, et al.: Deterministic learning and attempted suicide among older depressed individuals: cognitive assessment using the Wisconsin Card Sorting Task. J Psychiatr Res 2012; 46:226–232Crossref, MedlineGoogle Scholar

40 Lutz PE, Courtet P, Calati R: The opioid system and the social brain: implications for depression and suicide. J Neurosci Res 2020; 98:588–600Crossref, MedlineGoogle Scholar

41 Barnes LS, Ikeda RM, Kresnow MJ: Help-seeking behavior prior to nearly lethal suicide attempts. Suicide Life Threat Behav 2001; 32(suppl):68–75MedlineGoogle Scholar

42 Brown SL, Mitchell SM, Roush JF, et al.: Rejection sensitivity and suicide ideation among psychiatric inpatients: an integration of two theoretical models. Psychiatry Res 2019; 272:54–60Crossref, MedlineGoogle Scholar

43 Tsai AC, Lucas M, Kawachi I: Association between social integration and suicide among women in the United States. JAMA Psychiatry 2015; 72:987–993Crossref, MedlineGoogle Scholar

44 Roy B, Dwivedi Y: Understanding the neuroepigenetic constituents of suicide brain. Prog Mol Biol Transl Sci 2018; 157:233–262MedlineGoogle Scholar

45 Campos AI, Verweij KJH, Statham DJ, et al.: Genetic aetiology of self-harm ideation and behaviour. Sci Rep 2020; 10:9713Crossref, MedlineGoogle Scholar

46 Wender PH, Kety SS, Rosenthal D, et al.: Psychiatric disorders in the biological and adoptive families of adopted individuals with affective disorders. Arch Gen Psychiatry 1986; 43:923–929Crossref, MedlineGoogle Scholar

47 Statham DJ, Heath AC, Madden PA, et al.: Suicidal behaviour: an epidemiological and genetic study. Psychol Med 1998; 28:839–855Crossref, MedlineGoogle Scholar

48 Mullins N, Bigdeli TB, Børglum AD, et al.: GWAS of suicide attempt in psychiatric disorders and association with major depression polygenic risk scores. Am J Psychiatry 2019; 176:651–660LinkGoogle Scholar

49 Erlangsen A, Appadurai V, Wang Y, et al.: Genetics of suicide attempts in individuals with and without mental disorders: a population-based genome-wide association study. Mol Psychiatry (Online ahead of print, August 16, 2018)Google Scholar

50 Sokolowski M, Wasserman D: Genetic origins of suicidality? A synopsis of genes in suicidal behaviours, with regard to evidence diversity, disorder specificity, and neurodevelopmental brain transcriptomics. Eur Neuropsychopharmacol 2020; 37:1–11Crossref, MedlineGoogle Scholar

51 Galfalvy H, Haghighi F, Hodgkinson C, et al.: A genome-wide association study of suicidal behavior. Am J Med Genet B Neuropsychiatr Genet 2015; 168:557–563Crossref, MedlineGoogle Scholar

52 Pantazatos SP, Huang YY, Rosoklija GB, et al.: Whole-transcriptome brain expression and exon-usage profiling in major depression and suicide: evidence for altered glial, endothelial and ATPase activity. Mol Psychiatry 2017; 22:760–773Crossref, MedlineGoogle Scholar

53 Pantazatos SP, Andrews SJ, Dunning-Broadbent J, et al.: Isoform-level brain expression profiling of the spermidine/spermine N1-Acetyltransferase1 (SAT1) gene in major depression and suicide. Neurobiol Dis 2015; 79:123–134Crossref, MedlineGoogle Scholar

54 Haghighi F, Galfalvy H, Chen S, et al.: DNA methylation perturbations in genes involved in polyunsaturated fatty acid biosynthesis associated with depression and suicide risk. Front Neurol 2015; 6:92MedlineGoogle Scholar

55 Turecki G, Meaney MJ: Effects of the social environment and stress on glucocorticoid receptor gene methylation: a systematic review. Biol Psychiatry 2016; 79:87–96Crossref, MedlineGoogle Scholar

56 Coon H, Darlington TM, DiBlasi E, et al.: Genome-wide significant regions in 43 Utah high-risk families implicate multiple genes involved in risk for completed suicide. Mol Psychiatry (Online ahead of print, October 23, 2018)Google Scholar

57 Levey DF, Polimanti R, Cheng Z, et al.: Genetic associations with suicide attempt severity and genetic overlap with major depression. Transl Psychiatry 2019; 9:22Crossref, MedlineGoogle Scholar

58 Yin H, Galfalvy H, Pantazatos SP, et al.: Glucocorticoid receptor-related genes: genotype and brain gene expression relationships to suicide and major depressive disorder. Depress Anxiety 2016; 33:531–540Crossref, MedlineGoogle Scholar

59 Leszczyńska-Rodziewicz A, Szczepankiewicz A, Pawlak J, et al.: Association, haplotype, and gene-gene interactions of the HPA axis genes with suicidal behaviour in affective disorders. ScientificWorldJournal 2013; 2013:207361Crossref, MedlineGoogle Scholar

60 McGowan PO, Sasaki A, D’Alessio AC, et al.: Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat Neurosci 2009; 12:342–348Crossref, MedlineGoogle Scholar

61 Turecki G, Ernst C, Jollant F, et al.: The neurodevelopmental origins of suicidal behavior. Trends Neurosci 2012; 35:14–23Crossref, MedlineGoogle Scholar

62 Policicchio S, Washer S, Viana J, et al.: Genome-wide DNA methylation meta-analysis in the brains of suicide completers. Transl Psychiatry 2020; 10:69MedlineGoogle Scholar

63 Murphy TM, Mullins N, Ryan M, et al.: Genetic variation in DNMT3B and increased global DNA methylation is associated with suicide attempts in psychiatric patients. Genes Brain Behav 2013; 12:125–132Crossref, MedlineGoogle Scholar

64 Haghighi F, Xin Y, Chanrion B, et al.: Increased DNA methylation in the suicide brain. Dialogues Clin Neurosci 2014; 16:430–438MedlineGoogle Scholar

65 Schmaal L, van Harmelen AL, Chatzi V, et al.: Imaging suicidal thoughts and behaviors: a comprehensive review of 2 decades of neuroimaging studies. Mol Psychiatry 2020; 25:408–427Crossref, MedlineGoogle Scholar

66 Milak MS, Keilp J, Parsey RV, et al.: Regional brain metabolic correlates of self-reported depression severity contrasted with clinician ratings. J Affect Disord 2010; 126:113–124MedlineGoogle Scholar

67 Willeumier K, Taylor DV, Amen DG: Decreased cerebral blood flow in the limbic and prefrontal cortex using SPECT imaging in a cohort of completed suicides. Transl Psychiatry 2011; 1:e28Crossref, MedlineGoogle Scholar

68 Underwood MD, Kassir SA, Bakalian MJ, et al.: Neuron density and serotonin receptor binding in prefrontal cortex in suicide. Int J Neuropsychopharmacol 2012; 15:435–447Crossref, MedlineGoogle Scholar

69 Altshuler LL, Casanova MF, Goldberg TE, et al.: The hippocampus and parahippocampus in schizophrenia, suicide, and control brains. Arch Gen Psychiatry 1990; 47:1029–1034Crossref, MedlineGoogle Scholar

70 Boldrini M, Galfalvy H, Dwork AJ, et al.: Resilience is associated with larger dentate gyrus, while suicide decedents with major depressive disorder have fewer granule neurons. Biol Psychiatry 2019; 85:850–862Crossref, MedlineGoogle Scholar

71 van Heeringen K, Bijttebier S, Desmyter S, et al.: Is there a neuroanatomical basis of the vulnerability to suicidal behavior? A coordinate-based meta-analysis of structural and functional MRI studies. Front Hum Neurosci 2014; 8:824Crossref, MedlineGoogle Scholar

72 Jollant F, Wagner G, Richard-Devantoy S, et al.: Neuroimaging-informed phenotypes of suicidal behavior: a family history of suicide and the use of a violent suicidal means. Transl Psychiatry 2018; 8:120Crossref, MedlineGoogle Scholar

73 Rizk MM, Rubin-Falcone H, Lin X, et al.: Gray matter volumetric study of major depression and suicidal behavior. Psychiatry Res Neuroimaging 2019; 283:16–23Crossref, MedlineGoogle Scholar

74 Auerbach RP, Pagliaccio D, Allison GO, et al.: Neural correlates associated with suicide and nonsuicidal self-injury in youth. Biol Psychiatry (Online ahead of print, June 10, 2020)Google Scholar

75 Huber RS, Subramaniam P, Kondo DG, et al.: Reduced lateral orbitofrontal cortex volume and suicide behavior in youth with bipolar disorder. Bipolar Disord 2019; 21:321–329Crossref, MedlineGoogle Scholar

76 Jia Z, Wang Y, Huang X, et al.: Impaired frontothalamic circuitry in suicidal patients with depression revealed by diffusion tensor imaging at 3.0 T. J Psychiatry Neurosci 2014; 39:170–177Crossref, MedlineGoogle Scholar

77 Olvet DM, Peruzzo D, Thapa-Chhetry B, et al.: A diffusion tensor imaging study of suicide attempters. J Psychiatr Res 2014; 51:60–67Crossref, MedlineGoogle Scholar

78 Johnston JAY, Wang F, Liu J, et al.: Multimodal neuroimaging of frontolimbic structure and function associated with suicide attempts in adolescents and young adults with bipolar disorder. Am J Psychiatry 2017; 174:667–675LinkGoogle Scholar

79 Fan S, Lippard ETC, Sankar A, et al.: Gray and white matter differences in adolescents and young adults with prior suicide attempts across bipolar and major depressive disorders. J Affect Disord 2019; 245:1089–1097Crossref, MedlineGoogle Scholar

80 Mahon K, Burdick KE, Wu J, et al.: Relationship between suicidality and impulsivity in bipolar I disorder: a diffusion tensor imaging study. Bipolar Disord 2012; 14:80–89Crossref, MedlineGoogle Scholar

81 Cyprien F, de Champfleur NM, Deverdun J, et al.: Corpus callosum integrity is affected by mood disorders and also by the suicide attempt history: a diffusion tensor imaging study. J Affect Disord 2016; 206:115–124Crossref, MedlineGoogle Scholar

82 Lischke A, Domin M, Freyberger HJ, et al.: Structural alterations in the corpus callosum are associated with suicidal behavior in women with borderline personality disorder. Front Hum Neurosci 2017; 11:196MedlineGoogle Scholar

83 Taylor WD, Boyd B, McQuoid DR, et al.: Widespread white matter but focal gray matter alterations in depressed individuals with thoughts of death. Prog Neuropsychopharmacol Biol Psychiatry 2015; 62:22–28Crossref, MedlineGoogle Scholar

84 Myung W, Han CE, Fava M, et al.: Reduced frontal-subcortical white matter connectivity in association with suicidal ideation in major depressive disorder. Transl Psychiatry 2016; 6:e835Crossref, MedlineGoogle Scholar

85 Ordaz SJ, Goyer MS, Ho TC, et al.: Network basis of suicidal ideation in depressed adolescents. J Affect Disord 2018; 226:92–99Crossref, MedlineGoogle Scholar

86 Du L, Zeng J, Liu H, et al.: Fronto-limbic disconnection in depressed patients with suicidal ideation: a resting-state functional connectivity study. J Affect Disord 2017; 215:213–217Crossref, MedlineGoogle Scholar

87 Zhang S, Chen JM, Kuang L, et al.: Association between abnormal default mode network activity and suicidality in depressed adolescents. BMC Psychiatry 2016; 16:337MedlineGoogle Scholar

88 Lan MJ, Rizk MM, Pantazatos SP, et al.: Resting-state amplitude of low-frequency fluctuation is associated with suicidal ideation. Depress Anxiety 2019; 36:433–441Crossref, MedlineGoogle Scholar

89 Kang SG, Na KS, Choi JW, et al.: Resting-state functional connectivity of the amygdala in suicide attempters with major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2017; 77:222–227Crossref, MedlineGoogle Scholar

90 Chase HW, Segreti AM, Keller TA, et al.: Alterations of functional connectivity and intrinsic activity within the cingulate cortex of suicidal ideators. J Affect Disord 2017; 212:78–85Crossref, MedlineGoogle Scholar

91 Hamilton JP, Farmer M, Fogelman P, et al.: Depressive rumination, the default-mode network, and the dark matter of clinical neuroscience. Biol Psychiatry 2015; 78:224–230Crossref, MedlineGoogle Scholar

92 Kim K, Kim SW, Myung W, et al.: Reduced orbitofrontal-thalamic functional connectivity related to suicidal ideation in patients with major depressive disorder. Sci Rep 2017; 7:15772Crossref, MedlineGoogle Scholar

93 Li J, Duan X, Cui Q, et al.: More than just statics: temporal dynamics of intrinsic brain activity predicts the suicidal ideation in depressed patients. Psychol Med 2019; 49:852–860Crossref, MedlineGoogle Scholar

94 Minzenberg MJ, Lesh TA, Niendam TA, et al.: Conflict-related anterior cingulate functional connectivity is associated with past suicidal ideation and behavior in recent-onset psychotic major mood disorders. J Neuropsychiatry Clin Neurosci 2016; 28:299–305Crossref, MedlineGoogle Scholar

95 Olié E, Ding Y, Le Bars E, et al.: Processing of decision-making and social threat in patients with history of suicidal attempt: a neuroimaging replication study. Psychiatry Res 2015; 234:369–377Crossref, MedlineGoogle Scholar

96 Dombrovski AY, Szanto K, Clark L, et al.: Reward signals, attempted suicide, and impulsivity in late-life depression. JAMA Psychiatry 2013; 70:1MedlineGoogle Scholar

97 Pan LA, Hassel S, Segreti AM, et al.: Differential patterns of activity and functional connectivity in emotion processing neural circuitry to angry and happy faces in adolescents with and without suicide attempt. Psychol Med 2013; 43:2129–2142Crossref, MedlineGoogle Scholar

98 Jollant F, Lawrence NS, Olie E, et al.: Decreased activation of lateral orbitofrontal cortex during risky choices under uncertainty is associated with disadvantageous decision-making and suicidal behavior. Neuroimage 2010; 51:1275–1281Crossref, MedlineGoogle Scholar

99 Baek K, Kwon J, Chae JH, et al.: Heightened aversion to risk and loss in depressed patients with a suicide attempt history. Sci Rep 2017; 7:11228MedlineGoogle Scholar

100 Minzenberg MJ, Lesh TA, Niendam TA, et al.: Control-related frontal-striatal function is associated with past suicidal ideation and behavior in patients with recent-onset psychotic major mood disorders. J Affect Disord 2015; 188:202–209Crossref, MedlineGoogle Scholar

101 Reisch T, Seifritz E, Esposito F, et al.: An fMRI study on mental pain and suicidal behavior. J Affect Disord 2010; 126:321–325Crossref, MedlineGoogle Scholar

102 Silvers JA, Hubbard AD, Chaudhury S, et al.: Suicide attempters with borderline personality disorder show differential orbitofrontal and parietal recruitment when reflecting on aversive memories. J Psychiatr Res 2016; 81:71–78Crossref, MedlineGoogle Scholar

103 Olié E, Jollant F, Deverdun J, et al.: The experience of social exclusion in women with a history of suicidal acts: a neuroimaging study. Sci Rep 2017; 7:89MedlineGoogle Scholar

104 Jollant F, Lawrence NS, Giampietro V, et al.: Orbitofrontal cortex response to angry faces in men with histories of suicide attempts. Am J Psychiatry 2008; 165:740–748LinkGoogle Scholar

105 Gunn JF, Goldstein SE: Bullying and suicidal behavior during adolescence: a developmental perspective. Adolesc Res Rev 2016; 2:77–97Google Scholar

106 Akhtar Z, Alam M: Stress and suicidal ideation among school students. J Indian Acad Appl Psychol 2015; 41:236Google Scholar

107 Asberg M, Träskman L, Thorén P: 5-HIAA in the cerebrospinal fluid: a biochemical suicide predictor? Arch Gen Psychiatry 1976; 33:1193–1197Crossref, MedlineGoogle Scholar

108 Mann JJ: The serotonergic system in mood disorders and suicidal behaviour. Philos Trans R Soc Lond B Biol Sci 2013; 368:20120537Crossref, MedlineGoogle Scholar

109 Boldrini M, Underwood MD, Mann JJ, et al.: More tryptophan hydroxylase in the brainstem dorsal raphe nucleus in depressed suicides. Brain Res 2005; 1041:19–28Crossref, MedlineGoogle Scholar

110 Bach-Mizrachi H, Underwood MD, Tin A, et al.: Elevated expression of tryptophan hydroxylase-2 mRNA at the neuronal level in the dorsal and median raphe nuclei of depressed suicides. Mol Psychiatry 2008; 13:507–513Crossref, MedlineGoogle Scholar

111 Boldrini M, Underwood MD, Mann JJ, et al.: Serotonin-1A autoreceptor binding in the dorsal raphe nucleus of depressed suicides. J Psychiatr Res 2008; 42:433–442Crossref, MedlineGoogle Scholar

112 Sullivan GM, Oquendo MA, Milak M, et al.: Positron emission tomography quantification of serotonin(1A) receptor binding in suicide attempters with major depressive disorder. JAMA Psychiatry 2015; 72:169–178Crossref, MedlineGoogle Scholar

113 Richardson-Jones JW, Craige CP, Nguyen TH, et al.: Serotonin-1A autoreceptors are necessary and sufficient for the normal formation of circuits underlying innate anxiety. J Neurosci 2011; 31:6008–6018MedlineGoogle Scholar

114 Oquendo MA, Russo SA, Underwood MD, et al.: Higher postmortem prefrontal 5-HT2A receptor binding correlates with lifetime aggression in suicide. Biol Psychiatry 2006; 59:235–243Crossref, MedlineGoogle Scholar

115 Miller JM, Hesselgrave N, Ogden RT, et al.: Positron emission tomography quantification of serotonin transporter in suicide attempters with major depressive disorder. Biol Psychiatry 2013; 74:287–295Crossref, MedlineGoogle Scholar

116 Underwood MD, Kassir SA, Bakalian MJ, et al.: Serotonin receptors and suicide, major depression, alcohol use disorder, and reported early life adversity. Transl Psychiatry 2018; 8:279MedlineGoogle Scholar

117 Mann JJ, Malone KM: Cerebrospinal fluid amines and higher-lethality suicide attempts in depressed inpatients. Biol Psychiatry 1997; 41:162–171Crossref, MedlineGoogle Scholar

118 Oquendo MA, Placidi GP, Malone KM, et al.: Positron emission tomography of regional brain metabolic responses to a serotonergic challenge and lethality of suicide attempts in major depression. Arch Gen Psychiatry 2003; 60:14–22Crossref, MedlineGoogle Scholar

119 Oquendo MA, Galfalvy H, Sullivan GM, et al.: Positron emission tomographic imaging of the serotonergic system and prediction of risk and lethality of future suicidal behavior. JAMA Psychiatry 2016; 73:1048–1055Crossref, MedlineGoogle Scholar

120 Albert PR, Vahid-Ansari F, Luckhart C: Serotonin-prefrontal cortical circuitry in anxiety and depression phenotypes: pivotal role of pre- and post-synaptic 5-HT1A receptor expression. Front Behav Neurosci 2014; 8:199MedlineGoogle Scholar

121 Albert PR, Fiori LM: Transcriptional dys-regulation in anxiety and major depression: 5-HT1A gene promoter architecture as a therapeutic opportunity. Curr Pharm Des 2014; 20:3738–3750MedlineGoogle Scholar

122 Ladd CO, Huot RL, Thrivikraman KV, et al.: Long-term adaptations in glucocorticoid receptor and mineralocorticoid receptor mRNA and negative feedback on the hypothalamo-pituitary-adrenal axis following neonatal maternal separation. Biol Psychiatry 2004; 55:367–375Crossref, MedlineGoogle Scholar

123 Steinberg LJ, Mann JJ: Abnormal stress responsiveness and suicidal behavior: a risk phenotype. Biomarkers in Neuropsychiatry 2020; 2:100011CrossrefGoogle Scholar

124 Rohleder N: Acute and chronic stress induced changes in sensitivity of peripheral inflammatory pathways to the signals of multiple stress systems: 2011 Curt Richter Award Winner. Psychoneuroendocrinology 2012; 37:307–316Crossref, MedlineGoogle Scholar

125 Martinowich K, Lu B: Interaction between BDNF and serotonin: role in mood disorders. Neuropsychopharmacology 2008; 33:73–83Crossref, MedlineGoogle Scholar

126 Mathews DC, Richards EM, Niciu MJ, et al.: Neurobiological aspects of suicide and suicide attempts in bipolar disorder. Transl Neurosci 2013; 4MedlineGoogle Scholar

127 Galfalvy H, Currier D, Oquendo MA, et al.: Lower CSF MHPG predicts short-term risk for suicide attempt. Int J Neuropsychopharmacol 2009; 12:1327–1335Crossref, MedlineGoogle Scholar

128 Heim C, Nemeroff CB: The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies. Biol Psychiatry 2001; 49:1023–1039Crossref, MedlineGoogle Scholar

129 Sokolowski M, Wasserman J, Wasserman D: An overview of the neurobiology of suicidal behaviors as one meta-system. Mol Psychiatry 2015; 20:56–71Crossref, MedlineGoogle Scholar

130 Reinstatler L, Youssef NA: Ketamine as a potential treatment for suicidal ideation: a systematic review of the literature. Drugs R D 2015; 15:37–43Crossref, MedlineGoogle Scholar

131 Grunebaum MF, Ellis SP, Keilp JG, et al.: Ketamine versus midazolam in bipolar depression with suicidal thoughts: a pilot midazolam-controlled randomized clinical trial. Bipolar Disord 2017; 19:176–183Crossref, MedlineGoogle Scholar

132 Grunebaum MF, Galfalvy HC, Choo TH, et al.: Ketamine for rapid reduction of suicidal thoughts in major depression: a midazolam-controlled randomized clinical trial. Am J Psychiatry 2018; 175:327–335LinkGoogle Scholar

133 Murrough JW, Soleimani L, DeWilde KE, et al.: Ketamine for rapid reduction of suicidal ideation: a randomized controlled trial. Psychol Med 2015; 45:3571–3580Crossref, MedlineGoogle Scholar

134 Canuso CM, Singh JB, Fedgchin M, et al.: Efficacy and safety of intranasal esketamine for the rapid reduction of symptoms of depression and suicidality in patients at imminent risk for suicide: results of a double-blind, randomized, placebo-controlled study. Am J Psychiatry 2018; 175:620–630LinkGoogle Scholar

135 Milak MS, Rashid R, Dong Z, et al.: Assessment of relationship of ketamine dose with magnetic resonance spectroscopy of Glx and GABA responses in adults with major depression: a randomized clinical trial. JAMA Netw Open 2020; 3:e2013211Crossref, MedlineGoogle Scholar

136 Morris RG: NMDA receptors and memory encoding. Neuropharmacology 2013; 74:32–40MedlineGoogle Scholar

137 Rajkowska G, Stockmeier CA: Astrocyte pathology in major depressive disorder: insights from human postmortem brain tissue. Curr Drug Targets 2013; 14:1225–1236Crossref, MedlineGoogle Scholar

138 Gray AL, Hyde TM, Deep-Soboslay A, et al.: Sex differences in glutamate receptor gene expression in major depression and suicide. Mol Psychiatry 2015; 20:1057–1068Crossref, MedlineGoogle Scholar

139 Ahmadi J, Jahromi MS, Ehsaei Z: The effectiveness of different singly administered high doses of buprenorphine in reducing suicidal ideation in acutely depressed people with co-morbid opiate dependence: a randomized, double-blind, clinical trial. Trials 2018; 19:462MedlineGoogle Scholar

140 Yovell Y, Bar G, Mashiah M, et al.: Ultra-low-dose buprenorphine as a time-limited treatment for severe suicidal ideation: a randomized controlled trial. Am J Psychiatry 2016; 173:491–498LinkGoogle Scholar

141 Pfeiffer A, Brantl V, Herz A, et al.: Psychotomimesis mediated by kappa opiate receptors. Science 1986; 233:774–776MedlineGoogle Scholar

142 González D, Riba J, Bouso JC, et al.: Pattern of use and subjective effects of Salvia divinorum among recreational users. Drug Alcohol Depend 2006; 85:157–162MedlineGoogle Scholar

143 Kumor KM, Haertzen CA, Johnson RE, et al.: Human psychopharmacology of ketocyclazocine as compared with cyclazocine, morphine, and placebo. J Pharmacol Exp Ther 1986; 238:960–968MedlineGoogle Scholar

144 Carlezon WA Jr, Béguin C, DiNieri JA, et al.: Depressive-like effects of the kappa-opioid receptor agonist salvinorin A on behavior and neurochemistry in rats. J Pharmacol Exp Ther 2006; 316:440–447MedlineGoogle Scholar

145 Land BB, Bruchas MR, Schattauer S, et al.: Activation of the kappa opioid receptor in the dorsal raphe nucleus mediates the aversive effects of stress and reinstates drug seeking. Proc Natl Acad Sci USA 2009; 106:19168–19173Crossref, MedlineGoogle Scholar

146 Hurd YL, Herman MM, Hyde TM, et al.: Prodynorphin mRNA expression is increased in the patch vs matrix compartment of the caudate nucleus in suicide subjects. Mol Psychiatry 1997; 2:495–500Crossref, MedlineGoogle Scholar

147 Scarr E, Money TT, Pavey G, et al.: Mu opioid receptor availability in people with psychiatric disorders who died by suicide: a case control study. BMC Psychiatry 2012; 12:126MedlineGoogle Scholar

148 Roy A, Gorodetsky E, Yuan Q, et al.: Interaction of FKBP5, a stress-related gene, with childhood trauma increases the risk for attempting suicide. Neuropsychopharmacology 2010; 35:1674–1683Crossref, MedlineGoogle Scholar

149 Brent D, Melhem N, Ferrell R, et al.: Association of FKBP5 polymorphisms with suicidal events in the Treatment of Resistant Depression in Adolescents (TORDIA) study. Am J Psychiatry 2010; 167:190–197LinkGoogle Scholar

150 Ladd CO, Thrivikraman KV, Huot RL, et al.: Differential neuroendocrine responses to chronic variable stress in adult Long Evans rats exposed to handling-maternal separation as neonates. Psychoneuroendocrinology 2005; 30:520–533Crossref, MedlineGoogle Scholar

151 Mann JJ, Currier D, Stanley B, et al.: Can biological tests assist prediction of suicide in mood disorders? Int J Neuropsychopharmacol 2006; 9:465–474Crossref, MedlineGoogle Scholar

152 Coryell W, Schlesser M: The dexamethasone suppression test and suicide prediction. Am J Psychiatry 2001; 158:748–753LinkGoogle Scholar

153 Yerevanian BI, Feusner JD, Koek RJ, et al.: The dexamethasone suppression test as a predictor of suicidal behavior in unipolar depression. J Affect Disord 2004; 83:103–108Crossref, MedlineGoogle Scholar

154 Jokinen J, Nordström AL, Nordström P: ROC analysis of dexamethasone suppression test threshold in suicide prediction after attempted suicide. J Affect Disord 2008; 106:145–152Crossref, MedlineGoogle Scholar

155 Jokinen J, Nordström P: HPA axis hyperactivity and attempted suicide in young adult mood disorder inpatients. J Affect Disord 2009; 116:117–120Crossref, MedlineGoogle Scholar

156 Fountoulakis KN, Iacovides A, Fotiou F, et al.: Neurobiological and psychological correlates of suicidal attempts and thoughts of death in patients with major depression. Neuropsychobiology 2004; 49:42–52MedlineGoogle Scholar

157 Pitchot W, Scantamburlo G, Pinto E, et al.: Vasopressin-neurophysin and DST in major depression: relationship with suicidal behavior. J Psychiatr Res 2008; 42:684–688Crossref, MedlineGoogle Scholar

158 Black DW, Monahan PO, Winokur G: The relationship between DST results and suicidal behavior. Ann Clin Psychiatry 2002; 14:83–88Crossref, MedlineGoogle Scholar

159 Kirschbaum C, Pirke KM, Hellhammer DH: The “Trier Social Stress Test”: a tool for investigating psychobiological stress responses in a laboratory setting. Neuropsychobiology 1993; 28:76–81Crossref, MedlineGoogle Scholar

160 McGirr A, Diaconu G, Berlim MT, et al.: Personal and family history of suicidal behaviour is associated with lower peripheral cortisol in depressed outpatients. J Affect Disord 2011; 131:368–373Crossref, MedlineGoogle Scholar

161 Keilp JG, Stanley BH, Beers SR, et al.: Further evidence of low baseline cortisol levels in suicide attempters. J Affect Disord 2016; 190:187–192Crossref, MedlineGoogle Scholar

162 Melhem NM, Keilp JG, Porta G, et al.: Blunted HPA axis activity in suicide attempters compared to those at high risk for suicidal behavior. Neuropsychopharmacology 2016; 41:1447–1456Crossref, MedlineGoogle Scholar

163 O’Connor DB, Green JA, Ferguson E, et al.: Cortisol reactivity and suicidal behavior: investigating the role of hypothalamic-pituitary-adrenal axis responses to stress in suicide attempters and ideators. Psychoneuroendocrinology 2017; 75:183–191Crossref, MedlineGoogle Scholar

164 Stanley B, Michel CA, Galfalvy HC, et al.: Suicidal subtypes, stress responsivity, and impulsive aggression. Psychiatry Res 2019; 280:112486Crossref, MedlineGoogle Scholar

165 López-Ibor JJJ Jr, Saiz-Ruiz J, Pérez de los Cobos JC: Biological correlations of suicide and aggressivity in major depressions (with melancholia): 5-hydroxyindoleacetic acid and cortisol in cerebral spinal fluid, dexamethasone suppression test, and therapeutic response to 5-hydroxytryptophan. Neuropsychobiology 1985; 14:67–74Crossref, MedlineGoogle Scholar

166 Pfennig A, Kunzel HE, Kern N, et al.: Hypothalamus-pituitary-adrenal system regulation and suicidal behavior in depression. Biol Psychiatry 2005; 57:336–342Crossref, MedlineGoogle Scholar

167 Giletta M, Calhoun CD, Hastings PD, et al.: Multi-level risk factors for suicidal ideation among at-risk adolescent females: the role of hypothalamic-pituitary-adrenal axis responses to stress. J Abnorm Child Psychol 2015; 43:807–820MedlineGoogle Scholar

168 Rizk MM, Galfalvy H, Singh T, et al.: Toward subtyping of suicidality: brief suicidal ideation is associated with greater stress response. J Affect Disord 2018; 230:87–92Crossref, MedlineGoogle Scholar

169 Huang EJ, Reichardt LF: Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci 2001; 24:677–736Crossref, MedlineGoogle Scholar

170 McAllister AK: Neurotrophins and neuronal differentiation in the central nervous system. Cell Mol Life Sci 2001; 58:1054–1060MedlineGoogle Scholar

171 Poo MM: Neurotrophins as synaptic modulators. Nat Rev Neurosci 2001; 2:24–32MedlineGoogle Scholar

172 Berton O, McClung CA, Dileone RJ, et al.: Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 2006; 311:864–868Crossref, MedlineGoogle Scholar

173 Tsankova NM, Berton O, Renthal W, et al.: Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nat Neurosci 2006; 9:519–525Crossref, MedlineGoogle Scholar

174 Morse JK, Wiegand SJ, Anderson K, et al.: Brain-derived neurotrophic factor (BDNF) prevents the degeneration of medial septal cholinergic neurons following fimbria transection. J Neurosci 1993; 13:4146–4156MedlineGoogle Scholar

175 Alcántara S, Pozas E, Ibañez CF, et al.: BDNF-modulated spatial organization of Cajal-Retzius and GABAergic neurons in the marginal zone plays a role in the development of cortical organization. Cereb Cortex 2006; 16:487–499MedlineGoogle Scholar

176 Engelhardt M, Di Cristo G, Berardi N, et al.: Differential effects of NT-4, NGF, and BDNF on development of neurochemical architecture and cell size regulation in rat visual cortex during the critical period. Eur J Neurosci 2007; 25:529–540MedlineGoogle Scholar

177 Roceri M, Cirulli F, Pessina C, et al.: Postnatal repeated maternal deprivation produces age-dependent changes of brain-derived neurotrophic factor expression in selected rat brain regions. Biol Psychiatry 2004; 55:708–714Crossref, MedlineGoogle Scholar

178 Russo-Neustadt A: Brain-derived neurotrophic factor, behavior, and new directions for the treatment of mental disorders. Semin Clin Neuropsychiatry 2003; 8:109–118MedlineGoogle Scholar

179 Karege F, Vaudan G, Schwald M, et al.: Neurotrophin levels in postmortem brains of suicide victims and the effects of antemortem diagnosis and psychotropic drugs. Brain Res Mol Brain Res 2005; 136:29–37Crossref, MedlineGoogle Scholar

180 Dwivedi Y, Rizavi HS, Conley RR, et al.: Altered gene expression of brain-derived neurotrophic factor and receptor tyrosine kinase B in postmortem brain of suicide subjects. Arch Gen Psychiatry 2003; 60:804–815Crossref, MedlineGoogle Scholar

181 Banerjee R, Ghosh AK, Ghosh B, et al.: Decreased mRNA and protein expression of BDNF, NGF, and their receptors in the hippocampus from suicide: an analysis in human postmortem brain. Clin Med Insights Pathol 2013; 6:1–11Crossref, MedlineGoogle Scholar

182 Kim Y-K, Lee H-P, Won S-D, et al.: Low plasma BDNF is associated with suicidal behavior in major depression. Prog Neuropsychopharmacol Biol Psychiatry 2007; 31:78–85Crossref, MedlineGoogle Scholar

183 Iga J, Ueno S, Yamauchi K, et al.: The Val66Met polymorphism of the brain-derived neurotrophic factor gene is associated with psychotic feature and suicidal behavior in Japanese major depressive patients. Am J Med Genet B Neuropsychiatr Genet 2007; 144B:1003–1006MedlineGoogle Scholar

184 Schenkel LC, Segal J, Becker JA, et al.: The BDNF Val66Met polymorphism is an independent risk factor for high lethality in suicide attempts of depressed patients. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:940–944MedlineGoogle Scholar

185 Sarchiapone M, Carli V, Roy A, et al.: Association of polymorphism (Val66Met) of brain-derived neurotrophic factor with suicide attempts in depressed patients. Neuropsychobiology 2008; 57:139–145Crossref, MedlineGoogle Scholar

186 Pregelj P, Nedic G, Paska AV, et al.: The association between brain-derived neurotrophic factor polymorphism (BDNF Val66Met) and suicide. J Affect Disord 2011; 128:287–290Crossref, MedlineGoogle Scholar

187 Youssef MM, Underwood MD, Huang YY, et al.: Association of BDNF Val66Met polymorphism and brain BDNF levels with major depression and suicide. Int J Neuropsychopharmacol 2018; 21:528–538Crossref, MedlineGoogle Scholar

188 Gerritsen L, Tendolkar I, Franke B, et al.: BDNF Val66Met genotype modulates the effect of childhood adversity on subgenual anterior cingulate cortex volume in healthy subjects. Mol Psychiatry 2012; 17:597–603Crossref, MedlineGoogle Scholar

189 Dennis KE, Levitt P: Regional expression of brain derived neurotrophic factor (BDNF) is correlated with dynamic patterns of promoter methylation in the developing mouse forebrain. Brain Res Mol Brain Res 2005; 140:1–9MedlineGoogle Scholar

190 Keller S, Sarchiapone M, Zarrilli F, et al.: Increased BDNF promoter methylation in the Wernicke area of suicide subjects. Arch Gen Psychiatry 2010; 67:258–267Crossref, MedlineGoogle Scholar

191 Roth TL, Lubin FD, Funk AJ, et al.: Lasting epigenetic influence of early-life adversity on the BDNF gene. Biol Psychiatry 2009; 65:760–769Crossref, MedlineGoogle Scholar

192 Ernst C, Chen ES, Turecki G: Histone methylation and decreased expression of TrkB.T1 in orbital frontal cortex of suicide completers. Mol Psychiatry 2009; 14:830–832Crossref, MedlineGoogle Scholar

193 Maussion G, Yang J, Yerko V, et al.: Regulation of a truncated form of tropomyosin-related kinase B (TrkB) by Hsa-miR-185* in frontal cortex of suicide completers. PLoS One 2012; 7:e39301Crossref, MedlineGoogle Scholar

194 Colle R, Chupin M, Cury C, et al.: Depressed suicide attempters have smaller hippocampus than depressed patients without suicide attempts. J Psychiatr Res 2015; 61:13–18Crossref, MedlineGoogle Scholar

195 Courtet P, Jaussent I, Genty C, et al.: Increased CRP levels may be a trait marker of suicidal attempt. Eur Neuropsychopharmacol 2015; 25:1824–1831Crossref, MedlineGoogle Scholar

196 Cáceda R, Griffin WST, Delgado PL: A probe in the connection between inflammation, cognition, and suicide. J Psychopharmacol 2018; 32:482–488Crossref, MedlineGoogle Scholar

197 Chang CC, Tzeng NS, Kao YC, et al.: The relationships of current suicidal ideation with inflammatory markers and heart rate variability in unmedicated patients with major depressive disorder. Psychiatry Res 2017; 258:449–456Crossref, MedlineGoogle Scholar

198 Ekinci O, Ekinci A: The connections among suicidal behavior, lipid profile, and low-grade inflammation in patients with major depressive disorder: a specific relationship with the neutrophil-to-lymphocyte ratio. Nord J Psychiatry 2017; 71:574–580Crossref, MedlineGoogle Scholar

199 Black C, Miller BJ: Meta-analysis of cytokines and chemokines in suicidality: distinguishing suicidal versus nonsuicidal patients. Biol Psychiatry 2015; 78:28–37Crossref, MedlineGoogle Scholar

200 Ducasse D, Olié E, Guillaume S, et al.: A meta-analysis of cytokines in suicidal behavior. Brain Behav Immun 2015; 46:203–211Crossref, MedlineGoogle Scholar

201 Serafini G, Parisi VM, Aguglia A, et al.: A specific inflammatory profile underlying suicide risk? Systematic review of the main literature findings. Int J Environ Res Public Health 2020; 17:2393CrossrefGoogle Scholar

202 Keaton SA, Madaj ZB, Heilman P, et al.: An inflammatory profile linked to increased suicide risk. J Affect Disord 2019; 247:57–65Crossref, MedlineGoogle Scholar

203 Isung J, Aeinehband S, Mobarrez F, et al.: High interleukin-6 and impulsivity: determining the role of endophenotypes in attempted suicide. Transl Psychiatry 2014; 4:e470Crossref, MedlineGoogle Scholar

204 Conejero I, Jaussent I, Cazals A, et al.: Association between baseline pro-inflammatory cytokines and brain activation during social exclusion in patients with vulnerability to suicide and depressive disorder. Psychoneuroendocrinology 2019; 99:236–242MedlineGoogle Scholar

205 Erhardt S, Lim CK, Linderholm KR, et al.: Connecting inflammation with glutamate agonism in suicidality. Neuropsychopharmacology 2013; 38:743–752Crossref, MedlineGoogle Scholar

206 Bryleva EY, Brundin L: Kynurenine pathway metabolites and suicidality. Neuropharmacology 2017; 112(Pt B):324–330MedlineGoogle Scholar

207 Brundin L, Sellgren CM, Lim CK, et al.: An enzyme in the kynurenine pathway that governs vulnerability to suicidal behavior by regulating excitotoxicity and neuroinflammation. Transl Psychiatry 2016; 6:e865Crossref, MedlineGoogle Scholar

208 Suzuki H, Ohgidani M, Kuwano N, et al.: Suicide and microglia: recent findings and future perspectives based on human studies. Front Cell Neurosci 2019; 13:31MedlineGoogle Scholar

209 Schnieder TP, Trencevska I, Rosoklija G, et al.: Microglia of prefrontal white matter in suicide. J Neuropathol Exp Neurol 2014; 73:880–890MedlineGoogle Scholar

210 Holmes SE, Hinz R, Conen S, et al.: Elevated translocator protein in anterior cingulate in major depression and a role for inflammation in suicidal thinking: a positron emission tomography study. Biol Psychiatry 2018; 83:61–69Crossref, MedlineGoogle Scholar

211 Arling TA, Yolken RH, Lapidus M, et al.: Toxoplasma gondii antibody titers and history of suicide attempts in patients with recurrent mood disorders. J Nerv Ment Dis 2009; 197:905–908Crossref, MedlineGoogle Scholar

212 Sutterland AL, Kuin A, Kuiper B, et al.: Driving us mad: the association of Toxoplasma gondii with suicide attempts and traffic accidents: a systematic review and meta-analysis. Psychol Med 2019; 49:1608–1623Crossref, MedlineGoogle Scholar

213 Cook TB, Brenner LA, Cloninger CR, et al.: “Latent” infection with Toxoplasma gondii: association with trait aggression and impulsivity in healthy adults. J Psychiatr Res 2015; 60:87–94Crossref, MedlineGoogle Scholar

214 Vyas A, Kim SK, Giacomini N, et al.: Behavioral changes induced by Toxoplasma infection of rodents are highly specific to aversion of cat odors. Proc Natl Acad Sci USA 2007; 104:6442–6447Crossref, MedlineGoogle Scholar

215 O’Donovan A, Rush G, Hoatam G, et al.: Suicidal ideation is associated with elevated inflammation in patients with major depressive disorder. Depress Anxiety 2013; 30:307–314Crossref, MedlineGoogle Scholar

216 Bradley KA, Case JA, Khan O, et al.: The role of the kynurenine pathway in suicidality in adolescent major depressive disorder. Psychiatry Res 2015; 227:206–212MedlineGoogle Scholar

217 Rengasamy M, Zhong Y, Marsland A, et al.: Signaling networks in inflammatory pathways and risk for suicidal behavior. Brain Behav Immun Health 2020; 7:100122CrossrefGoogle Scholar

218 Cavanagh JT, Carson AJ, Sharpe M, et al.: Psychological autopsy studies of suicide: a systematic review. Psychol Med 2003; 33:395–405Crossref, MedlineGoogle Scholar

219 Oquendo MA, Malone KM, Ellis SP, et al.: Inadequacy of antidepressant treatment for patients with major depression who are at risk for suicidal behavior. Am J Psychiatry 1999; 156:190–194AbstractGoogle Scholar

220 Dranovsky A, Hen R: Hippocampal neurogenesis: regulation by stress and antidepressants. Biol Psychiatry 2006; 59:1136–1143Crossref, MedlineGoogle Scholar

221 Coppen A, Standish-Barry H, Bailey J, et al.: Does lithium reduce the mortality of recurrent mood disorders? J Affect Disord 1991; 23:1–7Crossref, MedlineGoogle Scholar

222 Felber W, Bauer M, Lewitzka U, et al.: Lithium clinics in Berlin and Dresden: a 50-year experience. Pharmacopsychiatry 2018; 51:166–171MedlineGoogle Scholar

223 Meltzer HY, Alphs L, Green AI, et al.: Clozapine treatment for suicidality in schizophrenia: International Suicide Prevention Trial (InterSePT). Arch Gen Psychiatry 2003; 60:82–91Crossref, MedlineGoogle Scholar

224 Vermeulen JM, van Rooijen G, van de Kerkhof MPJ, et al.: Clozapine and long-term mortality risk in patients with schizophrenia: a systematic review and meta-analysis of studies lasting 1.1-12.5 years. Schizophr Bull 2019; 45:315–329Crossref, MedlineGoogle Scholar

225 Cipriani A, Hawton K, Stockton S, et al.: Lithium in the prevention of suicide in mood disorders: updated systematic review and meta-analysis. BMJ 2013; 346:f3646Crossref, MedlineGoogle Scholar

226 Benedetti F, Radaelli D, Poletti S, et al.: Opposite effects of suicidality and lithium on gray matter volumes in bipolar depression. J Affect Disord 2011; 135:139–147Crossref, MedlineGoogle Scholar

227 Wilkowska A, Wiglusz MS, Cubała WJ: Clozapine: promising treatment for suicidality in bipolar disorder. Psychiatr Danub 2019; 31(suppl 3):574–578MedlineGoogle Scholar

228 Williams NR, Heifets BD, Blasey C, et al.: Attenuation of antidepressant effects of ketamine by opioid receptor antagonism. Am J Psychiatry 2018; 175:1205–1215LinkGoogle Scholar

229 Stenovec M, Lasič E, Božić M, et al.: Ketamine inhibits ATP-evoked exocytotic release of brain-derived neurotrophic factor from vesicles in cultured rat astrocytes. Mol Neurobiol 2016; 53:6882–6896MedlineGoogle Scholar

230 Grieco SF, Cheng Y, Eldar-Finkelman H, et al.: Up-regulation of insulin-like growth factor 2 by ketamine requires glycogen synthase kinase-3 inhibition. Prog Neuropsychopharmacol Biol Psychiatry 2017; 72:49–54MedlineGoogle Scholar

231 Hawton K, Witt KG, Salisbury TLT, et al.: Psychosocial interventions following self-harm in adults: a systematic review and meta-analysis. Lancet Psychiatry 2016; 3:740–750Crossref, MedlineGoogle Scholar

232 DeCou CR, Comtois KA, Landes SJ: Dialectical behavior therapy is effective for the treatment of suicidal behavior: a meta-analysis. Behav Ther 2019; 50:60–72Crossref, MedlineGoogle Scholar

233 Fu CH, Williams SC, Cleare AJ, et al.: Neural responses to sad facial expressions in major depression following cognitive behavioral therapy. Biol Psychiatry 2008; 64:505–512Crossref, MedlineGoogle Scholar

234 Rubin-Falcone H, Weber J, Kishon R, et al.: Longitudinal effects of cognitive behavioral therapy for depression on the neural correlates of emotion regulation. Psychiatry Res Neuroimaging 2018; 271:82–90Crossref, MedlineGoogle Scholar

235 Goodman M, Carpenter D, Tang CY, et al.: Dialectical behavior therapy alters emotion regulation and amygdala activity in patients with borderline personality disorder. J Psychiatr Res 2014; 57:108–116Crossref, MedlineGoogle Scholar

236 Chung D, Hadzi-Pavlovic D, Wang M, et al.: Meta-analysis of suicide rates in the first week and the first month after psychiatric hospitalisation. BMJ Open 2019; 9:e023883Crossref, MedlineGoogle Scholar

237 Kessler RC, Bauer MS, Bishop TM, et al.: Using administrative data to predict suicide after psychiatric hospitalization in the Veterans Health Administration system. Front Psychiatry 2020; 11:390MedlineGoogle Scholar

238 McCarthy JF, Bossarte RM, Katz IR, et al.: Predictive modeling and concentration of the risk of suicide: implications for preventive interventions in the US Department of Veterans Affairs. Am J Public Health 2015; 105:1935–1942Crossref, MedlineGoogle Scholar

239 Turecki G, Brent DA, Gunnell D, et al.: Suicide and suicide risk. Nat Rev Dis Primers 2019; 5:74Crossref, MedlineGoogle Scholar

240 Stanley B, Brown GK: Safety planning intervention: a brief intervention to mitigate suicide risk. Cognit Behav Pract 2012; 19:256–264CrossrefGoogle Scholar

241 Stanley B, Brown GK, Brenner LA, et al.: Comparison of the safety planning intervention with follow-up vs usual care of suicidal patients treated in the emergency department. JAMA Psychiatry 2018; 75:894–900Crossref, MedlineGoogle Scholar

242 Stanley B, Chaudhury SR, Chesin M, et al.: An emergency department intervention and follow-up to reduce suicide risk in the VA: acceptability and effectiveness. Psychiatr Serv 2016; 67:680–683LinkGoogle Scholar

243 Simon OR, Swann AC, Powell KE, et al.: Characteristics of impulsive suicide attempts and attempters. Suicide Life Threat Behav 2001; 32(suppl):49–59Crossref, MedlineGoogle Scholar

244 Chaudhury SR, Singh T, Burke A, et al.: Clinical correlates of planned and unplanned suicide attempts. J Nerv Ment Dis 2016; 204:806–811Crossref, MedlineGoogle Scholar

245 Lester D: Effects of detoxification of domestic gas on suicide in the Netherlands. Psychol Rep 1991; 68:202Crossref, MedlineGoogle Scholar

246 Kreitman N: The coal gas story: United Kingdom suicide rates, 1960–71. Br J Prev Soc Med 1976; 30:86–93MedlineGoogle Scholar

247 Hawton K, Bergen H, Simkin S, et al.: Long term effect of reduced pack sizes of paracetamol on poisoning deaths and liver transplant activity in England and Wales: interrupted time series analyses. BMJ 2013; 346:f403Crossref, MedlineGoogle Scholar

248 Reisch T, Steffen T, Habenstein A, et al.: Change in suicide rates in Switzerland before and after firearm restriction resulting from the 2003 “Army XXI” reform. Am J Psychiatry 2013; 170:977–984LinkGoogle Scholar

249 Owens D, Horrocks J, House A: Fatal and non-fatal repetition of self-harm. systematic review. Br J Psychiatry 2002; 181:193–199Crossref, MedlineGoogle Scholar

250 McGirr A, Alda M, Séguin M, et al.: Familial aggregation of suicide explained by cluster B traits: a three-group family study of suicide controlling for major depressive disorder. Am J Psychiatry 2009; 166:1124–1134LinkGoogle Scholar

251 Sisti D, Mann JJ, Oquendo MA: Toward a distinct mental disorder-suicidal behavior. JAMA Psychiatry (Online ahead of print, March 18, 2020)Google Scholar

252 Gratch I, Choo TH, Galfalvy H, et al.: Detecting suicidal thoughts: The power of ecological momentary assessment. Depress Anxiety (Online ahead of print, May 22, 2020)Google Scholar

253 Kleiman EM, Turner BJ, Fedor S, et al.: Digital phenotyping of suicidal thoughts. Depress Anxiety 2018; 35:601–608MedlineGoogle Scholar

254 Rizk MM, Choo TH, Galfalvy H, et al.: Variability in suicidal ideation is associated with affective instability in suicide attempters with borderline personality disorder. Psychiatry 2019; 82:173–178Crossref, MedlineGoogle Scholar

255 Peters EM, Dong LY, Thomas T, et al.: Instability of suicidal ideation in patients hospitalized for depression: an exploratory study using smartphone ecological momentary assessment. Arch Suicide Res (Online ahead of print, July 11, 2020)Google Scholar

256 Oquendo MA, Galfalvy HC, Choo TH, et al.: Highly variable suicidal ideation: a phenotypic marker for stress induced suicide risk. Mol Psychiatry (Online ahead of print, June 23, 2020)Google Scholar

257 Hall RC, Platt DE, Hall RC: Suicide risk assessment: a review of risk factors for suicide in 100 patients who made severe suicide attempts: evaluation of suicide risk in a time of managed care. Psychosomatics 1999; 40:18–27Crossref, MedlineGoogle Scholar

258 Bernanke JA, Stanley BH, Oquendo MA: Toward fine-grained phenotyping of suicidal behavior: the role of suicidal subtypes. Mol Psychiatry 2017; 22:1080–1081Crossref, MedlineGoogle Scholar

259 McHugh CM, Corderoy A, Ryan CJ, et al.: Association between suicidal ideation and suicide: meta-analyses of odds ratios, sensitivity, specificity, and positive predictive value. BJPsych Open 2019; 5:e18MedlineGoogle Scholar

260 Allen NB, Nelson BW, Brent D, et al.: Short-term prediction of suicidal thoughts and behaviors in adolescents: can recent developments in technology and computational science provide a breakthrough? J Affect Disord 2019; 250:163–169Crossref, MedlineGoogle Scholar

261 Rollman BL, Brent DA: Phonotype: a new taxonomy for mHealth research. J Gen Intern Med 2020; 35:1881–1883MedlineGoogle Scholar

262 Robinson TN, Reeves B, Ram N: Rollman and Brent: Phonotype. J Gen Intern Med 2020; 35:2479Crossref, MedlineGoogle Scholar

263 Kleiman EM, Glenn CR, Liu RT: Real-time monitoring of suicide risk among adolescents: potential barriers, possible solutions, and future directions. J Clin Child Adolesc Psychol 2019; 48:934–946MedlineGoogle Scholar

264 Torok M, Han J, Baker S, et al.: Suicide prevention using self-guided digital interventions: a systematic review and meta-analysis of randomised controlled trials. Lancet Digit Health 2020; 2:e25–e36Crossref, MedlineGoogle Scholar

265 Busch KA, Fawcett J, Jacobs DG: Clinical correlates of inpatient suicide. J Clin Psychiatry 2003; 64:14–19Crossref, MedlineGoogle Scholar

266 van Spijker BA, van Straten A, Kerkhof AJ: The effectiveness of a web-based self-help intervention to reduce suicidal thoughts: a randomized controlled trial. Trials 2010; 11:25Crossref, MedlineGoogle Scholar

267 Brook R, Klap R, Liao D, et al.: Mental health care for adults with suicide ideation. Gen Hosp Psychiatry 2006; 28:271–277Crossref, MedlineGoogle Scholar

268 Oquendo MA, Halberstam B, Mann JJ: Risk factors for suicidal behavior: utility and limitations of research instruments, in Standardized Evaluation in Clinical Practice. Edited by First MB. Washington, DC, American Psychiatric Publishing, 2003, pp 103–130Google Scholar

269 Nock MK, Park JM, Finn CT, et al.: Measuring the suicidal mind: implicit cognition predicts suicidal behavior. Psychol Sci 2010; 21:511–517Crossref, MedlineGoogle Scholar

270 Millner AJ, Augenstein TM, Visser KH, et al.: Implicit cognitions as a behavioral marker of suicide attempts in adolescents. Arch Suicide Res 2019; 23:47–63Crossref, MedlineGoogle Scholar

271 Glenn CR, Kleiman EM, Coppersmith DDL, et al.: Implicit identification with death predicts change in suicide ideation during psychiatric treatment in adolescents. J Child Psychol Psychiatry 2017; 58:1319–1329MedlineGoogle Scholar

272 Schneck N, Haufe S, Tu T, et al.: Tracking deceased-related thinking with neural pattern decoding of a cortical-basal ganglia circuit. Biol Psychiatry Cogn Neurosci Neuroimaging 2017; 2:421–429MedlineGoogle Scholar

273 Just MA, Pan L, Cherkassky VL, et al.: Machine learning of neural representations of suicide and emotion concepts identifies suicidal youth. Nat Hum Behav 2017; 1:911–919Crossref, MedlineGoogle Scholar

274 Dum J, Bläsig J, Herz A: Buprenorphine: demonstration of physical dependence liability. Eur J Pharmacol 1981; 70:293–300MedlineGoogle Scholar

275 Lee KO, Akil H, Woods JH, et al.: Differential binding properties of oripavines at cloned μ- and δ-opioid receptors. Eur J Pharmacol 1999; 378:323–330Crossref, MedlineGoogle Scholar

276 Greenwald M, Johanson CE, Bueller J, et al.: Buprenorphine duration of action: mu-opioid receptor availability and pharmacokinetic and behavioral indices. Biol Psychiatry 2007; 61:101–110Crossref, MedlineGoogle Scholar

277 Butler S: Buprenorphine: clinically useful but often misunderstood. Scand J Pain 2013; 4:148–152Crossref, MedlineGoogle Scholar

278 Lee JD, Nunes EV Jr, Novo P, et al.: Comparative effectiveness of extended-release naltrexone versus buprenorphine-naloxone for opioid relapse prevention (X:BOT): a multicentre, open-label, randomised controlled trial. Lancet 2018; 391:309–318Crossref, MedlineGoogle Scholar

279 Bodkin JA, Zornberg GL, Lukas SE, et al.: Buprenorphine treatment of refractory depression. J Clin Psychopharmacol 1995; 15:49–57Crossref, MedlineGoogle Scholar

280 Nyhuis PW, Gastpar M, Scherbaum N: Opiate treatment in depression refractory to antidepressants and electroconvulsive therapy. J Clin Psychopharmacol 2008; 28:593–595Crossref, MedlineGoogle Scholar

281 Ehrich E, Turncliff R, Du Y, et al.: Evaluation of opioid modulation in major depressive disorder. Neuropsychopharmacology 2015; 40:1448–1455Crossref, MedlineGoogle Scholar

282 Fava M, Memisoglu A, Thase ME, et al.: Opioid modulation with buprenorphine/samidorphan as adjunctive treatment for inadequate response to antidepressants: a randomized double-blind placebo-controlled trial. Am J Psychiatry 2016; 173:499–508LinkGoogle Scholar

283 Serafini G, Adavastro G, Canepa G, et al.: The efficacy of buprenorphine in major depression, treatment-resistant depression and suicidal behavior: a systematic review. Int J Mol Sci 2018; 19:2410CrossrefGoogle Scholar

284 Latif ZE, Šaltyte Benth J, Solli KK, et al.: Anxiety, depression, and insomnia among adults with opioid dependence treated with extended-release naltrexone vs buprenorphine-naloxone: a randomized clinical trial and follow-up study. JAMA Psychiatry 2019; 76:127–134Crossref, MedlineGoogle Scholar

285 Ahmadi J, Abtahi S: Eight-milligram single dose of buprenorphine as an expeditious treatment for severe suicidal ideation: a case report. Iran Red Crescent Med J 2017; 19:e59999CrossrefGoogle Scholar

286 Striebel JM, Kalapatapu RK: The anti-suicidal potential of buprenorphine: a case report. Int J Psychiatry Med 2014; 47:169–174Crossref, MedlineGoogle Scholar

287 Abdelnaim MA, Langguth B, Deppe M, et al.: Anti-suicidal efficacy of repetitive transcranial magnetic stimulation in depressive patients: a retrospective analysis of a large sample. Front Psychiatry 2020; 10:929Crossref, MedlineGoogle Scholar

288 Knoch D, Gianotti LR, Pascual-Leone A, et al.: Disruption of right prefrontal cortex by low-frequency repetitive transcranial magnetic stimulation induces risk-taking behavior. J Neurosci 2006; 26:6469–6472Crossref, MedlineGoogle Scholar

289 Dockery CA, Hueckel-Weng R, Birbaumer N, et al.: Enhancement of planning ability by transcranial direct current stimulation. J Neurosci 2009; 29:7271–7277Crossref, MedlineGoogle Scholar

290 Boggio PS, Zaghi S, Villani AB, et al.: Modulation of risk-taking in marijuana users by transcranial direct current stimulation (tDCS) of the dorsolateral prefrontal cortex (DLPFC). Drug Alcohol Depend 2010; 112:220–225Crossref, MedlineGoogle Scholar

291 Fecteau S, Knoch D, Fregni F, et al.: Diminishing risk-taking behavior by modulating activity in the prefrontal cortex: a direct current stimulation study. J Neurosci 2007; 27:12500–12505Crossref, MedlineGoogle Scholar

292 Teti Mayer J, Nicolier M, Gabriel D, et al.: Efficacy of transcranial direct current stimulation in reducing impulsivity in borderline personality disorder (TIMBER): study protocol of a randomized controlled clinical trial. Trials 2019; 20:347Crossref, MedlineGoogle Scholar

293 Schmidtner AK, Slattery DA, Gläsner J, et al.: Minocycline alters behavior, microglia, and the gut microbiome in a trait-anxiety-dependent manner. Transl Psychiatry 2019; 9:223Crossref, MedlineGoogle Scholar

294 Miyaoka T, Wake R, Furuya M, et al.: Minocycline as adjunctive therapy for patients with unipolar psychotic depression: an open-label study. Prog Neuropsychopharmacol Biol Psychiatry 2012; 37:222–226Crossref, MedlineGoogle Scholar

295 Faridhosseini F, Sadeghi R, Farid L, et al.: Celecoxib: a new augmentation strategy for depressive mood episodes: a systematic review and meta-analysis of randomized placebo-controlled trials. Hum Psychopharmacol 2014; 29:216–223Crossref, MedlineGoogle Scholar

296 Beurel E, Jope RS: Inflammation and lithium: clues to mechanisms contributing to suicide-linked traits. Transl Psychiatry 2014; 4:e488Crossref, MedlineGoogle Scholar

297 Block T, Petrides G, Kushner H, et al.: Mifepristone plasma level and glucocorticoid receptor antagonism associated with response in patients with psychotic depression. J Clin Psychopharmacol 2017; 37:505–511Crossref, MedlineGoogle Scholar

298 Lombardo G, Enache D, Gianotti L, et al.: Baseline cortisol and the efficacy of antiglucocorticoid treatment in mood disorders: a meta-analysis. Psychoneuroendocrinology 2019; 110:104420Crossref, MedlineGoogle Scholar