Skip to main content
Chronic Stress logoLink to Chronic Stress
. 2017 Jun 22;1:2470547017710916. doi: 10.1177/2470547017710916

Neurobiology of Chronic Stress-Related Psychiatric Disorders: Evidence from Molecular Imaging Studies

Margaret T Davis 1,2,, Sophie E Holmes 1,2, Robert H Pietrzak 1,2,3, Irina Esterlis 1,2,3
PMCID: PMC5976254  NIHMSID: NIHMS940862  PMID: 29862379

Abstract

Chronic stress accounts for billions of dollars of economic loss annually in the United States alone, and is recognized as a major source of disability and mortality worldwide. Robust evidence suggests that chronic stress plays a significant role in the onset of severe and impairing psychiatric conditions, including major depressive disorder, bipolar disorder, and posttraumatic stress disorder. Application of molecular imaging techniques such as positron emission tomography and single photon emission computed tomography in recent years has begun to provide insight into the molecular mechanisms by which chronic stress confers risk for these disorders. The present paper provides a comprehensive review and synthesis of all positron emission tomography and single photon emission computed tomography imaging publications focused on the examination of molecular targets in individuals with major depressive disorder, posttraumatic stress disorder, or bipolar disorder to date. Critical discussion of discrepant findings and broad strengths and weaknesses of the current body of literature is provided. Recommended future directions for the field of molecular imaging to further elucidate the neurobiological substrates of chronic stress-related disorders are also discussed. This article is part of the inaugural issue for the journal focused on various aspects of chronic stress.

Keywords: depression, posttraumatic stress disorder, bipolar disorder, positron emission tomography, single photon emission computed tomography

Introduction

The experience of chronic psychological stress is associated with a variety of serious physical, financial, and emotional consequences at both individual and societal levels.1 The World Health Organization has labeled stress as a “worldwide epidemic” in recognition of the magnitude of its deleterious effects.2 The American Institute of Stress estimates that the effects of chronic stress cost US companies over 300 billion dollars annually.3 More gravely, stress and resultant changes in affect are associated with increased morbidity and mortality,4 including increased rates of mental illness and suicide.5 Robust evidence has linked the experience of chronic stress to onset of major depressive disorder (MDD), which is the leading cause of disability worldwide.1,6,7 Likewise, onset of bipolar (BD) and posttraumatic stress (PTSD) disorders, both of which are associated with substantial emotional, physical, and financial burden,8,9 has been linked to chronic stress. Unfortunately, the molecular mechanisms by which chronic stress confers risk for mental health problems are poorly understood, hindering the development of critical interventions to resolve and prevent chronic stress-related disorders.

Application of Positron Emission Tomography and Single Photon Emission

Computed tomography (SPECT) has begun to provide information concerning how the brain changes following chronic stress on a molecular level. Positron emission tomography (PET) and SPECT molecular imaging techniques allow for the quantification of biochemical processes, including those known to play a pivotal role in the pathophysiology of psychiatric disorders in the living human brain.10 Both techniques rely on the injection of a “radiotracer,” a physiologically active compound with affinity for a specific target, which has been tagged with a small amount of radioactive isotope. This review is part of the journal’s inaugural issue, which is designed to comprehensively describe the neurobiological and neuroclinical effects of chronic stress. The role of this review is to summarize the existing molecular imaging work in psychiatric conditions known to be associated with chronic stress—MDD, BD, and PTSD—and recommend directions for future research.

Methods

Separate searches of the PubMed database were conducted for each disorder. In each case, the following standard prompts were used: “positron emission tomography” OR “PET” OR“single photon emission computed tomography” OR “SPECT” OR “single photon emission tomography” OR “SPET”). Distinct specifiers were then added for each of the three disorders: AND (“depression” OR “MDD”), AND (“bipolar disorder” OR “mania”), AND (“posttraumatic stress disorder” OR “PTSD”). Searches for MDD, BD, and PTSD initially yielded 2, 302, 447, and 158 PubMed article results, respectively. Studies were included in this review if they satisfied the following criteria: (1) articles focused on the use of PET or SPECT techniques, (2) articles describing original research, (3) articles presenting in vivo results in human participants, and (4) articles focused on the imaging of specific molecule/receptor types (i.e. excluding glucose metabolism and cerebral blood flow). Following careful review, 126 MDD, 22 BD, and 15 PTSD articles were retained.

Results

Major Depressive Disorder

A substantial body of research supports the assertion that stress, both chronic and acute, is causally related to the onset of MDD.11 Recent research suggests that exposure to chronic or “every day” stress in financial, occupational, or personal settings alone, or in combination with acute stress exposure is more powerfully predictive of MDD than acute stress exposure alone.1214 While short-term stress can be adaptive, prolonged, or chronic exposure to stress can lead to long-term dysregulation of many physiological and neurochemical processes. PET and SPECT research has provided more precise insight into the nature of the chronic stress-related alterations associated with the onset and persistence of MDD (Table 1).

Table 1.

Radiotracers used for in vivo molecular imaging in individuals with major depressive disorder.

Molecular target (receptor) MDD Radiotracer (full name) Abbreviation(s) Main findings
Serotonin synthesis a-[11C]methyl-L-tryptophan a-[11C]- MTrp ↓Serotonin synthesis in MDD1517
5-hydroxytryptophan (5-HTP) labelled with 11C in the ß position [ß-11C]5-HTP ↓Serotonin synthesis in MDD18,19
Serotonin transporter (SERT) (3-amino-4-(2-dimethylami-nomethyl- phenylsulfanyl)-benzonitrile) [11C]DASB ↓SERT availability in MDD;2022 mixed findings23,24
(11)C-(+)-6β -(4Methylthiophenyl)- 1,2,3,5,6alpha,10β hexahydropyrrolo [2,1-a] isoquinoline [11C]McN5652 ↓SERT availability in MDD;25,26 ↑SERT availability in MDD27,28
[123I] 2-((2-((dimethylamino)methyl) phenyl) thio)-5-iodophenylamine (aDaM) [123I]-ADAM ↓SERT availability in MDD;29–,32 ↑midbrain SERT availability in suicide attempters33
N, N-dimethyl-2-(2-amino-4- [18F]fluorophenylthio)benzylamine (4-[18F]-aDaM) (4-[18F]-ADAM) Suicidal ideation positively correlated with SERT availability34
Carbon-11 labeled 2β-carbomethoxy-3β- [4′-((Z)-2-iodoethenyl)phenyl]nortropane [11C]-ZIENT PET ↓SERT availability in MDD suicide attempters35
Serotonin 1A (5HT1A) receptors carbon11–labeledN-(2-1-4-2-methoxyphenyl)-1piperazinyl)ethyl))- N-(2-pyridyl)-cyclohexanecarboxamide [11C]WAY-100635 ↑5HT1A availability in MDD26,3640; ↓5HT1A availability in MDD4148
No-carrier-added 4-(2′-methoxyphenyl)- 1-[2′-(N-2-pyridinyl)-p-[18F] fluorobenzamido]ethylpiperazine) [18F]MPPF ↓5HT1A availability in MDD49
(cont.)
Serotonin 1B (5HT1B) receptors (R-1-[4-(2-methoxy-isopropyl)-phenyl]-3- [2-(4-methyl-piperazin-1-yl)benzyl]- pyrrolidin-2-one) [11C]P943 ↓5HT1B availability in MDD50
(5-methyl-8-(4-methyl-piperazin-1-yl)- 4-oxo-4H-chromene-2-carboxylic acid (4-morpholin-4-yl-phenyl)-amide) [11C]AZ10419369 ↓5HT1B availability in MDD;51 ↓5HT1B availability in MDD following therapy52
Serotonin 2A (5HT2A) receptors 123iodinated 4-amino-N-1- [3-(4-fluorophenoxy)propyl]-4-methyl-4- piperidinyl] 5-iodo-2-methoxybenzamide 123I-5-I-R91150 ↓5HT2A availability in MDD53
[18F]altanserin ↓5HT2A availability in MDD54,55
((R)-(+)-4 -(l-hydroxy-1- (2,3-dimethoxyphenyl)methyl)-N -2-(4-fluorophenylethyl)piperidine) labeled with 11C [11C]MDL 100,907 ↑5HT2A availability in individuals with a history of MDD56
[18F]fluoroethylspiperone [18F]FESP ↓5HT2A availability in drug-naïve MDD, not different from HC in SSRI responders57
[18F]setoperone ↓5HT2A availability in MDD;58,59 ↑5HT2A availability in MDD60,61
Serotonin receptor type 4 (5HT4) [11C]SB207145 ↓5HT4 availability in healthy individuals with family history of MDD62
SERT/dopamine transporter (DAT) 123I-labelled 2β-carbomethoxy-3 β-(4-iodophenyl)-tropane [123I]I-ß-CIT ↓SERT/DAT availability in MDD6366
[123I]nor-ß-CIT ↓SERT/DAT availability in MDD67,68
(2β-carbomethoxy-3β- (4- iodophenyl)-tropane [123I-labeled β -CIT Mixed results69,64
Dopamine synthesis [18F]fluorodopa [18F]DOPA ↓Dopamine synthesis70
Doapmine synthesis (cont) 11C-labeled 3,4-Dihydroxy-phenyl-L-alanine L-[11C]DOPA ↓Dopamine synthesis102
Dopamine transpoter (DAT) ((99 m)Tc-[2[[2-[[[3-(4-chlorophenyl)-8-methyl-8-azabicyclo[3,2,1]-oct-2-yl]-methyl](2-mercaptoethyl) amino]ethyl] amino]ethane-thiolato(3-)-N2,N2′,S2,S2]oxo-[1R-(exo-exo)])) [99mTc] TRODAT-1 ↓DAT availability in MDD7176
[11C]RTI-32 PET ↓DAT availability in MDD77
[123I]N-fluoropropyl-carbomethoxy-3β- (4-iodophenyl)tropane ↓DAT availability in MDD78
Dopamine type 2/3 receptors (D2/3) [123I]iodo-benzamide ([123I]IBZM) ↑D 2/3 availability in MDD;62,79 No differences in D 2/3 availability between MDD and HC80,74,81,82
[123I]epidepride ↑D 2/3 availability in MDD68
((S)-N-((1-ethyl-2-pyrrolidinyl)methyl)- 5- bromo-2,3-dimethoxybenzamide) [11C]FLB 457 No differences in D 2/3 availability between MDD and HC83
[11C]raclopride [11C]RAC ↑D1 availability in MDD24
Dopamine Type 1 Receptors (D1) 8-chloro-7-hydroxy-3-methyl-5- (7-benzofuranyl)-2,3,4,5-tetrahydro- IH-3-benzazepine) [11C]NNC-112 ↓D1 availability in MDD84
((R)-(+)-8-chloro-2,3,4,5-tetrahydro- 3- methyl-5-phenyl-1H-3-benzazepin-7-ol) [11C]SCH 23,390 ↓D1 availability in MDD with anger85
Metabotropic glutamate receptors type 5 (mGluR5) (3-(6-methyl-pyridin-2-ylethynyl)- cyclohex-2-enone-O-(11) C-methyl-oxime) [11C]ABP688 ↓mGluR5 availability in MDD86,87
[18F]FPEB No differences in mGluR5 availability between MDD and HC88
Translocator protein (TSPO) [11C]PBR28 No differences in TSPO availability between MDD (mild symptom severity) and HC89
fluorine F 18–labeled N-(2-(2- fluoroethoxy)benzyl)-N- (4-phenoxypyridin-3-yl)acetamide ([18F]FEPPA) ↑TSPO availability in MDD90,91
MAO-A ([11C]harmine) ↑MAO-A availability in MDD92,93
β-Amyloid F-florbetapir ↑ β-Amyloid availability in MDD94
N-Methyl-[11C]-2-(4′methyaminophenyl)- 6-hydroxybenzothiasole [11C]PIB-PET β-Amyloid availability correlated with MDD severity95
β2*-nAChR (123I-IA); [123I]-5-iodo-3-[2(S)-azetidinylmethoxy]pyridine [123I]5-I-A-85380 No differences in β2*-nAChR between MDD and HC96

Note. MDD, major depressive disorder; PET, positron emission tomography; MAO, monoamine oxidase; HC, healthy control.

The majority of existing PET and SPECT research in MDD populations has focused on the functioning of two monoaminergic neurotransmitters: serotonin and dopamine. Focus on monoamine functioning in depression resulted in part from the monoaminergic hypothesis,97 which postulates that alterations in the function of monoaminergic neurotransmission play a causal role in the development of MDD.

Serotonin

Between 1991 and 2016, 71 molecular imaging studies meeting the criteria specified above were published. Strong evidence supports the role of the serotonergic (5HT) system, which plays a role in the regulation of sleep, stress responses, and affective cognition98 in the development and continuation of depressive symptoms.99 However, the exact nature of the relationship between 5HT functioning and MDD is still under debate. Both preclinical and postmortem evidence suggests that serotonergic dysfunction and specifically deficits in serotonin are central to the pathophysiology of MDD.100,101 In line with such findings, evidence from PET work suggests lower serotonin synthesis in MDD individuals relative to healthy control,1519 and that treatment with selective serotonin reuptake inhibitor (SSRI) resulted in increased serotonin synthesis.16

The serotonin transporter (SERT) plays a key role in modulation of brain 5HT levels via reuptake into presynaptic neurons,98 and is the primary target of action for many commonly prescribed antidepressant medications.102 Interest in SERT has been motivated in part by evidence suggesting that expression of the SERT gene (5HTT) may moderate emotional and behavioral responses to stress,103 such that individuals displaying a specific 5HTT polymorphism (associated with decreased serotonergic functioning in preclinical studies103,104) were more likely to develop MDD and suicidal behavior following stress.105 Evidence from a recent SPECT study similarly found lower SERT availability in the thalamus and that high levels of life stress interacted to predict depressive symptom severity, suggesting that SERT may play a specific role in the development of depression following chronic stress.106 Miller et al.25 observed lower SERT availability across brain regions in individuals reporting a history of child abuse who went on to develop MDD, but not PTSD, relative to MDD individuals without an abuse history. Further, support for the relationship between SERT and stress comes from an experimental study of HPA axis dysfunction, as measured by the dexamethasone suppression test,107 showing an association with reduction in SERT levels. Evidence points to lower SERT availability in multiple brain areas, most commonly the thalamus,20,29 and midbrain21,22,30,31,67 or brainstem,63,69 areas responsible for regulation of sleep/wake cycles which are frequently disrupted in MDD.

Importantly, SERT levels appear to be related to antidepressant treatment outcome and illness progression.32,108,109,110,111 Increase in midbrain SERT availability is associated with depressive symptom remission following antidepressant treatment,109,111,112 and Amsterdam et al.110 showed a significant increase in both midbrain and medial temporal lobe SERT availability in treatment responders following 12 weeks of cognitive behavioral therapy. Conversely, in participants whose MDD symptoms did not remit following a year of antidepressant use, lower SERT availability was detected in several brain regions in individuals with MDD relative to healthy controls.27

Of note, in vivo evidence for low SERT in MDD is strong but not universal. Some studies have reported no significant differences in SERT availability between MDD and healthy control groups,23,28,64,113,114 while others reported higher SERT availability in individuals with MDD relative to controls.65,113,115 No clear explanation for these discrepant findings is available, though low sample size due to cost in PET and SPECT studies, difficulty in recruiting these patients, and cross-sample variability (e.g. symptom severity, sex, race, medication) likely play a role. Notably, characteristics of some specific radiotracers may have biased findings in some cases. For example, both SPECT tracers, [123I]nor-β-CIT and [123I]β-CIT, have been shown to bind not only to SERT but to norepinephrine (NET) and dopamine (DAT) transporters.116 Thus, depending on the region, quantification of SERT might represent SERT and DAT/NET densities.

A large focus in the serotonergic literature has also been the 5HT1A receptor, which is a post-synaptic G-coupled protein receptor and the most common serotonergic receptor in the brain.105 The 5HT1A receptor is located both in brain areas with projections from the raphe nuclei (RN), where the serotonergic system is centralized,117 and in the RN itself. Preclinical research has shown that animals with higher numbers of 5HT1A receptors are more vulnerable to stress118 and display more “depressive” behavior (e.g. helplessness119). Similarly, some postmortem studies have shown higher 5HT1A density in select brain regions (e.g. midbrain dorsal RN) of individuals with MDD relative to healthy controls.49 Based on evidence from other modalities, it is reasonable to hypothesize observation of similarly increased 5HT1A availability in vivo.

To date all published 5HT1A studies have used PET, with all but one using the 5HT1A radiotracer [11C]WAY-100635 with somewhat mixed findings. Unlike cited preclinical and postmortem studies, there does not appear to be a consensus on whether 5HT1A is up or downregulated in depression: about half the published studies observed lower 5HT1A availability in MDD participants across various brain regions,4146 whereas the other half found evidence for higher levels of 5HT1A in MDD, particularly in treatment-resistant individuals,26,3639,47,48,120,121 and with only one study reporting no 5HT1A differences between MDD participants and controls.40 Interestingly, downregulation in 5HT1A appears to be associated with treatment response following SSRI treatment.39,121

Importantly, a specific, testable explanation for the observed variability in 5HT1A findings has been proposed. In molecular imaging, there are various ways to quantify receptor availability, one of which is by the use of a reference tissue, which should be a region that is devoid of the target molecule (commonly the cerebellum). This method has been implemented in the studies that report lower 5HT1A availability in MDD; however, it appears that the cerebellum is not devoid of 5HT1A and there is evidence of lower cerebellar 5HT1A availability in the control relative to the MDD groups,98 which would bias the results and lead to an underestimation of the true 5HT1A density in MDD. Illustrating this point, Hesselgrave and Parsey122 showed that the same data set showed lower 5HT1A availability in MDD when analyses were completed using the cerebellum as reference, and the opposite finding when the outcome measure was a blood derived input function. It has also been suggested that 5HT1A level varies temporally over the course of MDD, possibly in part as a function of antidepressant exposure.122 At present, all that can be concluded is that the preponderance of evidence suggests 5HT1A is elevated in MDD.

Like 5HT1A, the 5HT1B receptor is an autoreceptor present in the terminal regions of serotonergic neurons and plays a role in regulating, specifically decreasing when activated, the amount of serotonin in the synapse.98 Both clinical123 and preclinical124 studies have shown that 5HT1B agonists have antidepressant properties, suggesting that unlike 5HT1A, lower levels of 5HT1B may be associated with MDD symptom experience. Although limited, evidence is somewhat consistent with this hypothesis with two reports of lower 5HT1B availability in the ventral striatum and pallidum50 and the anterior cingulate cortex (ACC), subgenual prefrontal cortex (PFC), and hippocampus,51 respectively, but another report of lower 5HT1B availability in the dorsal brainstem following cognitive behavioral therapy for MDD.52

The 5HT2A receptor has received significant molecular imaging research attention, largely because SSRIs are known to act directly on 5HT2A receptors, leading to receptor downregulation.125 Importantly, the 5HT2A receptor has proven responsive to stress exposure; 5HT2A levels have been shown to increase following exposure to chronic stress in preclinical studies.126,127 Depressive behavior following stress has also been shown to increase in response to 5HT2A agonists,128,129 further implicating upregulated 5HT2A in the pathophysiology of MDD. In vivo molecular imaging studies, however, have shown mixed results. There are several reports of downregulated5355,57,58,60 5HT2A in MDD across several brain regions. However, there are also studies of no significant differences between untreated MDD participants and controls,59,130 and two showed higher levels of 5HT2A in the basal ganglia131 and frontal, parietal, and occipital cortices56 of unmedicated MDD participants relative to controls. The reports of higher 5HT2A density in MDD are in agreement with studies showing a reduction in cortical 5HT2A availability following treatment with tricyclic antidepressants.61,130 In attempting to account for mixed findings concerning 5HT2A in MDD, Ruhe et al.98 suggested that individuals with MDD may have lower 5HT2A availability in the hippocampus due to HPA axis dysregulation and higher 5HT2A availability in other areas (e.g. the frontal cortex), a pattern that has been shown in animals exposed to chronic stress. However, this proposed explanation has not yet been directly tested.132

Finally, a single PET study investigated the relationship between MDD risk and availability of the 5HT4 receptor,62 which has been implicated in recent preclinical studies as a potential target for rapidly acting antidepressant agents.133 The authors reported that healthy individuals with a family history of MDD have lower 5HT4 availability in the striatum, which was inversely correlated with the number of first degree relatives with MDD. These findings implicate the 5HT4 receptor as a potential target subserving familial risk for MDD, though replication and exploration of this receptor system in individuals with MDD is warranted.

Dopamine

Another monoaminergic neurotransmitter implicated in the pathophysiology of MDD is dopamine, which is known to be associated with emotion regulation, attention, motivation, and reward.134 Dopaminergic circuits contribute to the regulation of concentration and memory functioning, as well as the ability to experience pleasure, all of which are impaired in MDD.135 Dopaminergic dysfunction (i.e. reduced D2 receptor function), specifically in the nucleus accumbens, has also been linked directly to the experience of chronic stress, and subsequent development of depressive symptoms (anhedonia) in preclinical studies.80,136

As with serotonin, early molecular imaging of the dopamine system sought to examine the neurotransmitter’s synthesis in the brain, speculating in part based on results from clinical studies,137 that less dopamine synthesis would be observed in depressed individuals.98 This was verified by some PET studies in limited brain regions.19,138 A much larger number of studies have examined the functioning of dopamine transmitters (DAT) in MDD but have produced incongruent results. Specifically, genetic research has suggested that a DAT gene polymorphism, which increases DAT availability, is associated with higher risk for MDD.70,139 Several postmortem studies, however, have observed lower brain-wide DAT availability in MDD individuals. Findings in the molecular imaging literature are similarly mixed. Although there are reports of higher DAT availability in the striatum,71,140 basal ganglia,72 and other brain areas73 of MDD participants, others reported no significant differences between MDD and healthy control (HC),74,75 and three reported lower DAT availability in MDD in the striatum.76,78,141 Furthermore, Meyer et al.76 reported that a downregulation in DAT availability was related to more severe depressive symptoms. However, they did not detect a significant change in DAT availability before and after bupropion antidepressant therapy,76 suggesting that bupropion treatment did not appear to target this system or this system may not be significantly involved in MDD pathophysiology.

Measuring DAT with SPECT has also yielded some mixed results.28,63,6769,77,108,109 However, not all of these studies characterized their results as indicative of DAT availability because, as noted above, SPECT DAT tracers are recognized to have affinity for SERT and NET as well. Results of these studies should therefore be interpreted with caution. Furthermore, Camardese et al.66 highlighted the heterogeneity in presentation of MDD symptoms that can meet diagnosis and the potential effects of pharmacological treatment history and drug abuse as potential confounds affecting the observed variability in findings. As with SERT, inconsistent findings make it difficult to form a conclusion concerning the relationship between DAT and MDD at present.

The majority of neuroimaging studies investigating dopamine receptor availability have focused primarily on D2/D3 receptors, in part due to these receptors being targets for antidepressant and antipsychotic therapy.136 Evidence from other modalities implicates possible downregulation of D2/3 receptors in MDD. Epigenetic studies have presented evidence for a link between chronic stress, D2 downregulation, and the development of depression.66 Similarly, preclinical studies have shown downregulation of D3 in animals experiencing stress and depression. However, of the 14 studies which have examined D2/3 availability using PET and SPECT in individuals with MDD, all but 3 failed to observe differences in both medicated44,81,142,143 and unmedicated26,43,73,79,142 MDD participants relative to HCs. The remaining three showed higher D2/3 availability in MDD in the striatum,82,131 and in the case of Lehto et al.,77 a positive correlation between MDD symptom severity and D2/3 availability in the temporal cortex. However, although in vivo evidence suggests no specific D2/3 dysregulation in MDD, alterations treatment with SSRIs appears to influence D2/3 availability. For example, Montgomery et al.24 reported lower D2/3 availability in the striatum of MDD participants using SSRIs, while two other studies observed increased D2/3 in the basal ganglia136 and striatum83 of SSRI treatment responders but not non-responders. Of note, while only two published studies have examined the D1 receptor in vivo in MDD, both found lower D1 availability in the striatum of MDD relative to control groups.85,144

Glutamate

More recently, there has been a major focus on the glutamatergic system in MDD. Glutamate is the primary excitatory neurotransmitter in the brain, with 80%–90% of synapses in the human brain being glutamatergic. Dysfunction of the glutamate system is thought to play a role in MDD84 and there is currently an intensive focus on the glutamate system as a target for novel treatments for MDD.84 Part of this focus has been on metabotropic glutamate receptors (mGluRs), G-protein-coupled receptors that mediate neuromodulatory effects of glutamate.145 Indeed, mGluR5 antagonists have consistently shown antidepressant effects in animal models of depression.146151 Lower levels of mGluR5 protein expression was also found in postmortem PFC tissue of individuals with MDD;152 however, another study did not detect differences using autoradiography.86 To date, there are four PET imaging studies that report on mGluR5 availability in MDD. Two smaller scale studies found lower levels of mGluR5 in various brain regions of those with unmedicated MDD including the PFC, ACC, and insula.152,153 A recent PET study focusing on late-life MDD found no differences in mGluR5 availability between older adults with MDD and matched controls.87 Similarly, a recent study by our group found no differences in mGluR5 availability in a large group of unmedicated MDD individuals compared to HCs. As part of the same study, we used MRS to assess measures of glutamate and found a negative correlation between mGluR5 availability and MRS measures of glutamate in the ACC.154 It is possible, therefore, that higher levels of glutamate are associated with a downregulation of mGluR5 in MDD. Furthermore, we recently observed that rapid downregulation of mGluR5 is associated with a reduction of somatic anxiety symptoms in MDD participants, suggesting this might be a treatment target for anxiety in MDD.153 Further work will be required to definitively determine whether mGluR5 is a viable treatment target for symptoms of MDD.

Inflammation

Convergent evidence indicates that inflammation plays a significant role in MDD.88,155,156 Inflammatory mechanisms have been proposed to underlie the link between chronic stress and depression,33 and meta-analyses have confirmed elevated inflammatory markers in individuals with MDD.157160 However, the specific role of inflammation in the brain is less clear. It is possible to measure neuroinflammation with PET and radioligands that bind to the translocator protein (TSPO), which is upregulated on activated microglia, the immune cells of the brain.161 To date, there are two published PET studies investigating TSPO in depression. The first found no difference in TSPO availability between depressed individuals with symptoms of mild severity and controls.162 The second larger study showed elevated microglial activation in medication-free depression of moderate to severe severity, specifically in the PFC, ACC, and insula.89 This finding has recently been replicated in a similar sample of medication-free depressed individuals.90 Interestingly, this study found elevated TSPO in depressed individuals with versus without current suicidal thinking, suggesting a role for neuroinflammation in suicidal ideation specifically. Indeed, the heterogeneity in MDD symptomatology may play a role in the expression of neuroinflammation in MDD and account for divergent findings. PET and the 40+ existing TSPO radioligands provide a unique opportunity for clarifying the role of neuroinflammation in the pathophysiology of MDD and evaluating the use of anti-inflammatory medications in treating its symptoms.

Other Neurotransmitters

In addition to those reviewed above, PET and SPECT have been used to measure other systems such as monoamine oxidase (MAO-A), β-Amyloid, nicotinic acetylcholine receptors (β2*-nAChR), and γ-aminobutyric acid (GABA)—benzodiazepine receptors (GABAA-BZR). MAO-A is an enzyme known to be responsible for the regulation of brain monoamine levels.91 Higher MAO-A availability is observed in MDD participants relative to HC,91,92 which would be in line with the studies showing lower monoamine neurotransmission in MDD.

β-Amyloid deposition in the brain, which is associated with the development of dementia, has been shown to correlate with history of lifetime MDD episode in postmortem work.93 A single PET study by Wu et al.93 showed higher β-Amyloid levels in depressed, but cognitively normal, older adults relative to age-matched HCs in multiple brain regions. A recent study by Yasuno et al.94 looking at older adults without clinical MDD also reported a positive correlation between β-Amyloid availability averaged across the brain and depressive symptom severity. The cholinergic system has long been identified to be dysregulated in MDD,95,163166 with the hypothesis of excesses in acetylcholine levels contributing to the experiences of depression. Specific to the neuroimaging literature, the β2*-nAChR has been identified as potentially relevant to MDD symptom expression,167 although clinical trials with targets to downregulate β2*-nAChR did not prove efficacious for treatment of MDD. Combined in vivo/postmortem study from our group verified that β2*-nAChR does not appear to be dysregulated in MDD; however, elevations in acetylcholine levels might indeed play a role in depression.168 Finally, a single study examined GABAA-BZR availability in MDD and found no differences in availability between MDD and control groups.96

PET and SPECT Literature: Suicidal behavior

Importantly, while suicidal behavior is most frequently investigated as an associated feature of MDD, evidence from clinical research suggests the existence of meaningful neurobiological differences between suicide attempters and non-attempters.169 For example, platelet studies showed that suicidal individuals had higher number for 5HT2A binding sites compared both to MDD non-attempters and HCs.169 Similarly, variation in the DRD2 gene affecting D2 receptor expression has been consistently associated with risk for suicidal behavior, but not for MDD.35 To date, seven published studies, all focused on the serotonergic system, have explicitly examined associations with current or historical suicidal behavior (ideation or attempt). Five studies focused on SERT availability in MDD participants with suicidal behavior with interesting if mixed results. For example, Yeh et al. observed a positive correlation between SERT availability and suicidal ideation across brain regions33 while two other studies observed lower SERT availability in the midbrain170 and putamen170 of MDD participants with a history of suicide attempts compared to both healthy controls and MDD non-attempters. In combination, these findings suggest that lower midbrain SERT availability might distinguish between MDD individuals with and without risk for suicide. Notably, Oquendo et al.171 failed to confirm midbrain SERT availability as a predictor of attempts in a recent longitudinal study. They did, however, observe that increased 5HT1A availability in the RN predicted both suicidal ideation and lethality of suicidal behavior.171 In a cross-sectional study, Sullivan et al. also found that 5HT1A availability in the RN correlated positively with the lethality of attempt.90 Thus, 5HT1A availability in the RN serve as a marker for suicide risk. Of note, a single PET study in high-lethality suicide attempters across psychiatric diagnoses found lower serotonin synthesis in the orbital and ventromedial PFC of attempters relative to HCs.172 These promising results underscore the need for more molecular imaging work in suicidal individuals.

Bipolar Disorder

Relationship Between BD and Chronic Stress

Stress is also thought to play a significant role in the development of bipolar disorder (BD). Stress can trigger the first manic/hypomanic episode, predict episode recurrence, and lead to less favorable outcomes.173177 Thus, there is significant interest in stress as a target for treatment and prevention strategies in BD. Molecular imaging can provide invaluable information on the molecular mechanisms underlying the link between stress and BD. To date, 22 PET and SPECT studies have been performed in BD (Table 2), and like MDD, the majority (n=14) have focused on the serotonergic system.

Table 2.

Radiotracers used for in vivo molecular imaging in individuals with bipolar disorder.

Molecular target (receptor) BD Radiotracer (full name) Abbreviation(s) Major findings
SERT (serotonin transporter) trans-1,2,3,5,6,10- -hexahydro-6-[4-(methylthio) phenyl]pyrrolo-[2,1-a]-isoquinoline [11C](+)-McNeil 5652 ↓SERT availability in BD178
[11C]-3-amino-4-(2-dimethylaminomethyl-phenylsulfanyl)-benzonitrile [11C]DASB Mixed findings65,34
[(123)I]-2-((2-((dimethylamino)methyl)phenyl)thio)- 5-iodophenylamine [123I]-ADAM ↓SERT availability in euthymic BD179184
Serotonin 2A receptor (5HT2A) [18F]-setoperone Mixed findings185,186
Serotonin type 1A receptor (5HT1A) [N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2- pyridinyl) cyclohexane carboxamide] [Carbonyl-C-11]WAY-100635 No differences in 5HT2A availability in euthymic BD;187 ↑5HT2A availability in BD depression123
[18F]trans- 4-fluoro-N-(2-[4-(2-methoxyphenyl) piperazino]-ethyl)-N- (2-pyridyl) cyclohexanecarboxamid [18F]FCWAY ↓5HT2A availability in BD depression188
Dopamine transporter (DAT) ((99 m)Tc-[2[[2-[[[3-(4-chlorophenyl)-8-methyl-8-azabicyclo[3,2,1]-oct-2-yl]-methyl](2-mercaptoethyl) amino]ethyl]amino]ethane-thiolato(3-)-N2,N2′,S2,S2]oxo-[1R-(exo-exo)])) [99mTc]TRODAT-1 ↑DAT availability in depressed189 and euthymic BD190
[O-methyl-11C]b -CFT [11C]CFT ↓DAT availability in BD across mood states
Dopamine type 2 teceptor (D2) [11C]raclopride. [11C]RAC No differences in D2 availability in manic BD191
Translocator protein (TSPO) [1-(2-chlorophenyl)-N-methyl-N-(1-methyl propyl)-3-isoquinoline carboxamide] [11C]-(R)-PK11195 ↑TSPO availability in euthymic BD
β2*-nAChR 5-iodo-3-(2(S) azetidinylmethoxy) pyridine [123I]5IA No differences in β2*-nAChR availability between depressed BD, euthymic BD, and HC192
Muscarinic type 2 (M2) receptor (fluorodopa F 18 [3-(3-[3-fluoroprop- ly]thio)-1,2,5-thiadiazol-4-yl]-1,2,5,6-tetrahydro-1- methylpyridine) [18F]FP-TZTP ↓M2 availability in depressed BD193,194

Note. BP, bipolar disorder.

PET and SPECT Literature: BD

Serotonin

Serotoninergic function has long been thought to be central to the development of bipolar disorder.58 Prange et al.195 proposed what they termed the “permissive hypothesis of 5HT function” in bipolar disorder, which stated that deficient serotonergic functioning underlies bipolar disorder, and that manic and depressive episodes specifically would be marked by low 5HT. Supporting this proposal, a recent meta-analysis confirmed two gene SNPs related to the production of tryptophan hydroxylase-2, a rate-limiting enzyme for 5HT in the brain, are consistently associated with the development of bipolar disorder.196 However, existing PET and SPECT imaging evidence has not yet substantiated the permissive hypothesis. It appears that there is a variability in SERT availability across mood states in BD.98 During euthymia, there is lower SERT availability consistently measured across studies.179182,184,197 During depression, studies appear to diverge in their findings, with reports of no differences in SERT availability in BD compared to HC,184 lower SERT availability in thalamus, putamen, amygdala of depressed BD participants,34 and higher SERT availability in the thalamus compared to MDD participants, but lower availability in the RN relative to controls.178 No studies investigating SERT availability in manic BD participants have been published to date.

Both 5HT1A and 5HT2A are target receptors for medications used to treat BD (e.g. lamotrigine198 and ziprasidone,199 respectively). Further, genetic studies have found that the presence of specific SNPs in the 5HT1A gene predict both MDD and BD,200 suggesting a possible similar pattern of 5HT1A upregulation in both disorders. By contrast, a postmortem study observed decreased 5HT2A mRNA levels in the hippocampal region of BD individuals relative to MDD and HC.201 Quantitation of 5HT1A and 5HT2A receptors in BD in vivo has led to divergence in findings,187,188,202 including lower and higher receptor availability during depression, and no differences during euthymia.187 5HT2A availability has been examined in bipolar mania only, with observations of lower availability relative to HC in all cortical regions,185 and no alterations in 5HT2A availability following three to five weeks treatment with mood stabilizers.186 More research across mood states is needed to increase understanding of the potential involvement of both receptors in BD. Of note, researchers have observed that while serotonin modulation plays a role in antidepressant action in bipolar depression, based on examination of the most effective agents for BD depression, it is not sufficient for symptom relief.198 It is therefore crucial that molecular imaging research in BD examine other receptor systems.

Dopamine

Dopaminergic dysfunction is also implicated in the development and expression of BD.98 Researchers have suggested that increased dopaminergic function underlies mania, while the opposite is true for depressive episodes in BD.203 Many antipsychotic drugs are commonly prescribed to treat mania block dopamine receptors, decreasing levels of dopamine in the brain.204 Indeed drugs known to increase levels of dopamine (e.g. some tricyclic antidepressants) have also been shown to induce mania in BD individuals.205,206 Two postmortem studies have observed upregulation of D2 in the dlPFC of BD.207,208 However, to date, only one molecular imaging study has examined D2 in BD during mania, finding no differences in D2 receptor availability in the striatum of manic BD participants relative to HCs.191 The remainder of molecular imaging studies have focused on DAT, dysfunction of which, genetic research suggests, is associated with risk for BD.209,210 Interestingly, DAT findings in BD diverge based on imaging modality used. More specifically, two studies using SPECT and [99mTc]TRODAT-1 (a non-selective DAT tracer98) observed higher DAT availability in the striatum of both depressed189 and euthymic190 BD participants relative to HC. By contrast, using PET and a DAT selective tracer ([11C]CFT), Anand et al.208 observed lower striatal DAT in BD across mood states relative to HC. As with serotonin, more research is needed to further clarify the relationship between dopaminergic neurotransmission and BD.

Inflammation

Mounting evidence suggests that inflammation plays a key role in BD as well as MDD.211 For example, pro-inflammatory cytokines have been shown to be elevated in the blood of BD individuals, during periods of mania, depression, and euthymia, suggesting that low-grade inflammation may be a trait marker of BD.212 Preliminary evidence suggests the presence of inflammation in the brain as well as the periphery in BD, with one postmortem study showing upregulation of markers of neuroinflammation in the frontal cortex in BD.213 One study has investigated neuroinflammation in vivo using PET to date. Using [11C](R)PK11195, Haarman et al. reported elevated TSPO levels in the right hippocampus of euthymic-medicated BD participants compared to controls.214 Whether these medications impact neuroinflammation and whether level of measurable neuroinflammation differs across mood states remains to be determined.

Other Neurotransmitters

Cholinergic neurotransmitter systems have also been implicated in the pathophysiology of BD.193 Both muscarinic agonists and acetylcholinesterase inhibitors increase cholinergic neurotransmission, and have been shown to increase depressive and decrease manic symptoms.193 They have also been found to elicit erratic emotional responding—similar to that manifested in BD—when administered to HCs.215 Additionally, CHRM2, which encodes for muscarinic receptors (M2) is one of several candidate genes shown to be expressed more in the ACC of BD individuals relative to HC in a recent postmortem study. Three molecular imaging studies have examined the availability of muscarinic (mACh) and nicotinic (nACh) receptors in BD. Cannon et al. first showed lower M2 availability in the ACC of depressed BD participants relative to HC.193 They then showed that variation in the gene that encodes for M2, specifically, presence of the TT allele, was associated with lower M2 availability in the ACC of BD participants, and with more severe lifetime BD symptoms.194 However, we did not find lower β2*-nAChR in depressed BD participants compared to euthymic BD and HC individuals, although variability in ACh levels between depression and euthymia may interfere with receptor availability quantification.192 Alterations in ACh levels were not taken into account in the Cannon et al. studies; therefore, the role of the cholinergic receptors in the pathophysiology of BD needs further research.

Posttraumatic Stress Disorder

Relationship Between PTSD and Chronic Stress

PTSD is a psychiatric condition, which by definition occurs in direct response to acute stress exposure (i.e. exposure to trauma). However, while approximately 80% of individuals will be exposed to a traumatic event capable of conferring PTSD (per the DSM-56,21), only 8%–15% of the general population will go on to develop PTSD.8 One reliable predictor of developing PTSD following trauma exposure is historical exposure to chronic stress.217 As with MDD and BD, molecular imaging has the potential to provide crucial insights into the pathophysiology of PTSD and specifically its relationship to chronic stress (Table 3).

Table 3.

Radiotracers used for in vivo molecular imaging in individuals with posttraumatic stress disorder.

Molecular target (receptor) Radiotracer (full name) Abbreviation(s) Studies utilizing this tracer in vivo
Neurokinin 1 Receptor (NK1) (2 S,3 S)-N-[[2-[11C]Methoxy-5-[5-(trifluoromethyl)tetrazol-1-yl]phenyl]methyl]-2-phenyl-piperidin-3-amine [11C]GR205171
Serotonin transpoter (SERT) [11C]2-(2-(Dimethylaminomethyl)phenylthio)-5-fluoromethylphenylamine [11C]AFM ↓SERT availability in MDD218
[11C]-3-amino-4-(2-dimethylaminomethylphenylsulfanyl)- benzonitrile [11C]DASB ↓SERT availability in MDD219,220
Serotonin 1B receptor (5HT1B) (R-1-[4-(2-methoxy-isopropyl)-phenyl]-3-[2-(4-methyl-piperazin-1-yl)benzyl]-pyrrolidin-2-one) [11C]P943 ↓5HT1B in trauma exposed HC50
Serotonin 1A receptor (5HT1A) carbon11–labeledN-(2-(1-(4-(2-methoxyphenyl)-1piperazinyl)ethyl))- N-(2-pyridyl)- cyclohexanecarboxamide [carbonyl-C-11]WAY-100635 ↑5HT1A availability in MDD221
(N-{2-[4-(2-methoxyphenyl)piperazino]}-N-(2-pyridinyl)trans-4-fluorocyclohexanecarboxamide) [18F]CWAY No differences in 5HT1A availability in MDD222
Dopamine transporter (DAT) ((99 m)Tc-[2[[2-[[[3-(4-chlorophenyl)-8-methyl-8-azabicyclo[3,2,1]-oct-2-yl]-methyl](2-mercaptoethyl) amino]ethyl]amino]ethane-thiolato(3-)-N2,N2′,S2,S2]oxo-[1R-(exo-exo)])) [99mTC]-TRODAT-1 ↑DAT availability in PTSD223
GABAA-BZR [123I]iomazenil ↑ GABAA-BZR availability in PTSD224
[11C]flumazenil ↑ GABAA-BZR availability in PTSD225
Cannabinoid receptor 1 (CB1) ([11C]JHU75528) [11C]OMAR ↑CB1 availability in PTSD226,227
Norepinephrine Transporter (NET) [11C]methylreboxetine ([11C]MRB) ↓NET availability in PTSD relative to HC, but not trauma controls228
ß2*-nAChR (123I-5-IA; [123I]-5-iodo-3-[2(S)-azetidinylmethoxy]pyridine) [123I]5-IA-85380 ([123I]5-IA) ↑ ß2*-nAChR availability in PTSD229
K- Opioid Receptor [11C]LY2795050 ↓KOR availability associated with severity of loss symptoms in trauma survivors

PET and SPECT Literature: PTSD

Serotonin

As was the case with MDD and BD, altered serotonergic function has been implicated in the development of PTSD.203 More specifically, in contrast to reductions observed in MDD models, preclinical studies have reported increased 5HT synthesis in PTSD in multiple brain regions.231233 As in MDD, presence of the short SERT gene allele is a risk factor for PTSD, although a recent meta-analysis suggests that risk only extends to highly trauma exposed individuals.234 Published imaging data suggest lower SERT availability in PTSD as well as correlations between SERT and severity of PTSD-related symptomatology.218220

Focusing on presynaptic serotonin receptors, preclinical evidence suggests that alterations in 5HT1A function are associated with attentional bias to threat-related stimuli,235 and exposure to stress reduces functioning of the 5HT1B receptor.236 In line with preclinical findings, Sullivan et al.222 observed upregulation of 5HT1A in the amygdala, RN, and all cortical regions of individuals with PTSD with and without comorbid MDD relative to HCs. Of note, an earlier study by Bonne et al.221 observed no differences in 5HT1A availability in PTSD participants. However, Sullivan et al.222 argued that characteristics of the PET tracer used and issues with the analytic approach used might have contributed to the Bonne’s null result. Further, consistent with preclinical evidence, there is a finding of lower 5HT1B availability in trauma-exposed control participants relative to HCs with no previous trauma exposure.50 Earlier age of trauma exposure appears to be associated with both lower 5HT1B availability and PTSD symptom severity, suggesting that 5HT1B availability may be affected by trauma exposure, particularly during development.

Dopamine

Dopaminergic hyper-functioning has also been implicated in the development of PTSD, and specifically in the experience of hyperarousal symptoms.237 More specifically, a DAT gene polymorphism has been shown to be associated with risk for developing PTSD following trauma exposure.238 In keeping, researchers found higher availability of DAT in the striatum of PTSD participants relative to trauma-exposed control subjects.223

GABAA-BZR

Evidence has shown that exposure to trauma/acute stress is associated with reduction in GABAA benzodiazepine receptor (GABAA-BZR) density,239 which plays an important role in modulating nervous stress response in the central nervous system.225 In vivo quantification is in line with the animal studies reporting lower PFC GABAA-BZR availability in individuals with combat-related or nonrelated PTSD relative to HCs.224,225

Other Neurotransmitters

Other neurotransmitters have also been examined in trauma-related psychopathology: cannabinoid receptor 1 (CB1), NET, β2*-nAChR, and mGluR5. In line with preclinical reports of an association between chronic stress and altered CB1,240,241 Neumeister et al.226 reported elevated CB1 availability brain-wide in PTSD participants relative to both trauma-exposed controls and HCs. In an research domain criteria approach to examine the pathophysiology of PTSD, we reported that trauma and PTSD-related symptomology was associated with higher amygdalar CB1 availability.227 NET has also been implicated in the development of anxiety-related disorders like PTSD.242 A single PET study by Pietrzak et al.243 found lower NET availability in PTSD in the locus coeruleus relative to healthy controls. However, no significant differences in NET availability were observed in the trauma-exposed control group (compared to PTSD and HC groups).228 β2*-nAChRs availability, which has been implicated in regulation of memory and arousal which function abnormally in PTSD,244 appears to be upregulated in the mesiotemporal cortex of PTSD individuals relative to HCs. This receptor also appears to be implicated in the re-experiencing symptoms in PTSD.229 Finally, mGluR5 has emerged as a target of interest in PTSD245 due to its role in fear conditioning and emotion regulation. In a recent pilot study, we observed increased mGluR5 availability in PTSD participants relative to HC across cortical regions. MGluR5 availability was also correlated with severity of PTSD avoidance symptoms. The cause of this dysregulation is not clear; however, postmortem examination and preclinical data suggest deficits in glucocorticoid functioning may be impacting mGluR5 density. Based on this preliminary evidence, CB1, NET, β2*-nACh, and mGluR5 may all play roles in the pathophysiology of PTSD; however, the limited numbers of studies preclude firm conclusions.

Discussion

In the last 20 years, molecular imaging has made a substantial contribution to the understanding of the neurobiological alterations underlying chronic stress-related disorders. The reviewed studies have enriched our understanding of how and why existing psychotherapeutic agents affect neurotransmitter function and have implicated novel targets for optimizing treatment efficacy.246 To begin, they have confirmed the presence of in vivo alterations in serotonergic and dopaminergic functioning in at least some subgroups of MDD, BD, and PTSD individuals and provided preliminary evidence that other neurotransmitter systems play a role in these disorders.247 Areas of overlap between molecular imaging evidence and other research modalities (e.g. lower SERT in MDD) lend confidence to assertions concerning the role of some receptors and psychiatric diagnoses. Areas of discrepancy, however, encourage critical examination of explanations for such incongruity like that proposed and tested by Hesselgrave and Parsey122 to explain discrepant in vivo 5HT1A results in MDD.

While PET and SPECT have been confirmed as powerful and highly useful clinical research tools,228 as it stands, the molecular imaging literature on MDD, BD, and PTSD raises as many questions as it answers. One exciting novel application of molecular imaging with the potential to reduce variability in findings is transdiagnostic research. As noted, the experience of chronic stress conveys risk for complex and highly comorbid psychiatric diagnoses (e.g. MDD and PTSD). Heterogeneous symptom presentation within wide and overlapping diagnostic categories makes the systematic study of these disorders by obtaining a “representative sample” extremely challenging. Transdiagnostic designs could eliminate a significant source of error by basing analyses on the presence or absence of a specific symptoms in lieu of diagnosis (i.e. using research domain criteria248). In a recent example, Pietrzak et al.249 examined the availability of Kappa-opioid receptors (KORs) in individuals with a range of trauma-related pathology (including MDD and PTSD). Instead of examining the relationship between diagnostic status and receptor density, they used principal components analysis to group symptoms into transdiagnostic “threat” and “loss” categories, and observed that low KOR availability in the amygdalar-ACC-ventral striatal circuit was associated with severe loss, but not threat symptoms, suggesting the use of molecular imaging to investigate the relationship between receptor availability and transdiagnostic aspects of chronic stress-related psychopathology may be an important next step.

In summary, molecular imaging has provided unique insight into the neurobiological underpinnings of chronic stress-related disorders. Despite inconsistencies in specific findings, the wide-reaching impact of chronic stress, and subsequent disorders like MDD, BD, and PTSD, is evident. The ability to confirm preclinical and postmortem findings, and to quantify the action of pharmacological agents in vivo has allowed for identification of biomarkers to improve risk assessment, progress toward optimization of pharmacotherapy, and identification of promising new treatment targets.

Declaration of Conflicting Interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

The author(s) received no financial support for the research, authorship, and/or publication of this article.

References

  • 1.Schmidt MV, Sterlemann V, Muller MB. Chronic stress and individual vulnerability. Ann N Y Acad Sci 2008; 1148: 174–183. [DOI] [PubMed] [Google Scholar]
  • 2.Organization WH. Task shifting: rational redistribution of tasks among health workforce teams: global recommendations and guidelines. 2007. http://apps.who.int/iris/handle/10665/43821. Accessed May 16, 2017.
  • 3.Rosch PJ. The quandary of job stress compensation. Health Stress 2001; 3(1): 1–4. [Google Scholar]
  • 4.Kopp MS, Rethelyi J. Where psychology meets physiology: chronic stress and premature mortality—the Central-Eastern European health paradox. Brain Res Bull 2004; 62(5): 351–367. [DOI] [PubMed] [Google Scholar]
  • 5.Paykel ES. Life stress, depression and attempted suicide. J Human Stress 1976; 2(3): 3–12. [DOI] [PubMed] [Google Scholar]
  • 6.Insel T, Cuthbert B, Garvey M, Murray CJL, Lopez AD. Alternative projections of mortality and disability by cause 1990–2020: the global burden of disease study. Lancet 1997; 349: 1498–1504. [DOI] [PubMed] [Google Scholar]
  • 7.US Department of Health and Human Services. National Strategy for Suicide Prevention: Goals and Objectives for Action. Rockville. The Yale Textbook of Public Psychiatry. 2016; 349: 111.
  • 8.Kessler RC. Posttraumatic stress disorder: the burden to the individual and to society. J Clin Psychiatry 2000; 61(suppl 5): 4–12. [PubMed] [Google Scholar]
  • 9.Kupfer DJ. The increasing medical burden in bipolar disorder. JAMA 2005; 293(20): 2528–2530. [DOI] [PubMed] [Google Scholar]
  • 10.Mier W, Mier D. Advantages in functional imaging of the brain. Front Hum Neurosci 2015; 9: 249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.McEwen BS. Protection and damage from acute and chronic stress: allostasis and allostatic overload and relevance to the pathophysiology of psychiatric disorders. Ann N Y Acad Sci 2004; 1032: 1–7. [DOI] [PubMed] [Google Scholar]
  • 12.Cohen S, Janicki-Deverts D, Miller GE. Psychological stress and disease. JAMA 2007; 298(14): 1685–1687. [DOI] [PubMed] [Google Scholar]
  • 13.McGonagle KA, Kessler RC. Chronic stress, acute stress, and depressive symptoms. Am J Community Psychol 1990; 18(5): 681–706. [DOI] [PubMed] [Google Scholar]
  • 14.Thoits PA. Stress and health: major findings and policy implications. J Health Soc Behav 2010; 51(Suppl): S41–53. [DOI] [PubMed] [Google Scholar]
  • 15.Frey BN, Skelin I, Sakai Y, Nishikawa M, Diksic M. Gender differences in α-[11 C] MTrp brain trapping, an index of serotonin synthesis, in medication-free individuals with major depressive disorder: a positron emission tomography study. Psychiatry Res Neuroimaging 2010; 183(2): 157–166. [DOI] [PubMed] [Google Scholar]
  • 16.Berney A, Nishikawa M, Benkelfat C, Debonnel G, Gobbi G, Diksic M. An index of 5-HT synthesis changes during early antidepressant treatment: α-[11 C] methyl-l-tryptophan PET study. Neurochem Int 2008; 52(4): 701–708. [DOI] [PubMed] [Google Scholar]
  • 17.Rosa-Neto P, Diksic M, Okazawa H, et al. Measurement of brain regional α-[11C] Methyl-L-Tryptophan trapping as a measure of serotonin synthesis in medication-free patients with major depression. Arch Gen Psychiatry 2004; 61(6): 556–563. [DOI] [PubMed] [Google Scholar]
  • 18.Agren H, Reibring L, Hartvig P, et al. Low brain uptake of L [11C] 5-hydroxytryptophan in major depression: a positron emission tomography study on patients and healthy volunteers. Acta Psychiatrica Scandinavica 1991; 83(6): 449–455. [DOI] [PubMed] [Google Scholar]
  • 19.Ågren H, Reibring L, Hartvig P, et al. PET studies with L-[11C] 5-HTP and L-[11C] dopa in brains of healthy volunteers and patients with major depression. Clin Neuropharmacol 1992; 15(Part A): 235A–236A. [DOI] [PubMed] [Google Scholar]
  • 20.Reimold M, Batra A, Knobel A, et al. Anxiety is associated with reduced central serotonin transporter availability in unmedicated patients with unipolar major depression: a [11C]DASB PET study. Mol Psychiatry 2008; 13(6): 606–613, 557. [DOI] [PubMed] [Google Scholar]
  • 21.Hahn A, Haeusler D, Kraus C, et al. Attenuated serotonin transporter association between dorsal raphe and ventral striatum in major depression. Hum Brain Mapp 2014; 35(8): 3857–3866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Selvaraj S, Murthy NV, Bhagwagar Z, et al. Diminished brain 5-HT transporter binding in major depression: a positron emission tomography study with [11C]DASB. Psychopharmacology (Berl) 2011; 213(2–3): 555–562. [DOI] [PubMed] [Google Scholar]
  • 23.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(4): 287–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Montgomery AJ, Stokes P, Kitamura Y, Grasby PM. Extrastriatal D 2 and striatal D 2 receptors in depressive illness: pilot PET studies using [11 C] FLB 457 and [11 C] raclopride. J Affect Disord 2007; 101(1): 113–122. [DOI] [PubMed] [Google Scholar]
  • 25.Miller JM, Kinnally EL, Ogden RT, Oquendo MA, Mann JJ, Parsey RV. Reported childhood abuse is associated with low serotonin transporter binding in vivo in major depressive disorder. Synapse 2009; 63(7): 565–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Moses-Kolko EL, Wisner KL, Price JC, et al. Serotonin 1A receptor reductions in postpartum depression: a positron emission tomography study. Fertil Steril 2008; 89(3): 685–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Miller JM, Oquendo MA, Ogden RT, Mann JJ, Parsey RV. Serotonin transporter binding as a possible predictor of one-year remission in major depressive disorder. J Psychiatr Res 2008; 42(14): 1137–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ruhe HG, Booij J, Reitsma JB, Schene AH. Serotonin transporter binding with [123I]beta-CIT SPECT in major depressive disorder versus controls: effect of season and gender. Eur J Nucl Med Mol Imaging 2009; 36(5): 841–849. [DOI] [PubMed] [Google Scholar]
  • 29.Ho PS, Ho KK, Huang WS, et al. Association study of serotonin transporter availability and SLC6A4 gene polymorphisms in patients with major depression. Psychiatry Res 2013; 212(3): 216–222. [DOI] [PubMed] [Google Scholar]
  • 30.Newberg AB, Amsterdam JD, Wintering N, et al. 123I-ADAM binding to serotonin transporters in patients with major depression and healthy controls: a preliminary study. J Nucl Med 2005; 46(6): 973–977. [PubMed] [Google Scholar]
  • 31.Newberg AB, Amsterdam JD, Wintering N, Shults J. Low brain serotonin transporter binding in major depressive disorder. Psychiatry Res 2012; 202(2): 161–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Blasi G, De Virgilio C, Papazacharias A, et al. Converging evidence for the association of functional genetic variation in the serotonin receptor 2a gene with prefrontal function and olanzapine treatment. JAMA Psychiatry 2013; 70(9): 921–930. [DOI] [PubMed] [Google Scholar]
  • 33.Köhler O, Benros ME, Nordentoft M, et al. Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: a systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry 2014; 71(12): 1381–1391. [DOI] [PubMed] [Google Scholar]
  • 34.Oquendo MA, Hastings RS, Huang Y-Y, et al. Brain serotonin transporter binding in depressed patients with bipolar disorder using positron emission tomography. Arch Gen Psychiatry 2007; 64(2): 201–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mandelli L, Serretti A. Gene environment interaction studies in depression and suicidal behavior: an update. Neurosci Biobehav Rev 2013; 37(10): 2375–2397. [DOI] [PubMed] [Google Scholar]
  • 36.Sargent PA, Kjaer KH, Bench CJ, et al. Brain serotonin1A receptor binding measured by positron emission tomography with [11C] WAY-100635: effects of depression and antidepressant treatment. Arch Gen Psychiatry 2000; 57(2): 174–180. [DOI] [PubMed] [Google Scholar]
  • 37.Parsey RV, Olvet DM, Oquendo MA, Huang Y-Y, Ogden RT, Mann JJ. Higher 5-HT1A receptor binding potential during a major depressive episode predicts poor treatment response: preliminary data from a naturalistic study. Neuropsychopharmacology 2006; 31(8): 1745–1749. [DOI] [PubMed] [Google Scholar]
  • 38.Parsey RV, Oquendo MA, Ogden RT, et al. Altered serotonin 1A binding in major depression: a [carbonyl-C-11] WAY100635 positron emission tomography study. Biol Psychiatry 2006; 59(2): 106–113. [DOI] [PubMed] [Google Scholar]
  • 39.Gray NA, Milak MS, DeLorenzo C, et al. Antidepressant treatment reduces serotonin-1A autoreceptor binding in major depressive disorder. Biol Psychiatry 2013; 74(1): 26–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Mickey BJ, Ducci F, Hodgkinson CA, Langenecker SA, Goldman D, Zubieta J-K. Monoamine oxidase A genotype predicts human serotonin 1A receptor availability in vivo. J Neurosci 2008; 28(44): 11354–11359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lothe A, Saoud M, Bouvard S, Redouté J, Lerond J, Ryvlin P. 5-HT 1A receptor binding changes in patients with major depressive disorder before and after antidepressant treatment: a pilot [18 F] MPPF positron emission tomography study. Psychiatry Res Neuroimaging 2012; 203(1): 103–104. [DOI] [PubMed] [Google Scholar]
  • 42.Drevets WC, Frank E, Price JC, et al. PET imaging of serotonin 1A receptor binding in depression. Biol Psychiatry 1999; 46(10): 1375–1387. [DOI] [PubMed] [Google Scholar]
  • 43.Hirvonen J, Karlsson H, Kajander J, et al. Decreased brain serotonin 5-HT1A receptor availability in medication-naive patients with major depressive disorder: an in-vivo imaging study using PET and [carbonyl-11C] WAY-100635. Int J Neuropsychopharmacol 2008; 11(4): 465–476. [DOI] [PubMed] [Google Scholar]
  • 44.Saijo T, Takano A, Suhara T, et al. Effect of electroconvulsive therapy on 5-HT1A receptor binding in patients with depression: a PET study with [11C] WAY 100635. Int J Neuropsychopharmacol 2010; 13(6): 785–791. [DOI] [PubMed] [Google Scholar]
  • 45.Meltzer CC, Price JC, Mathis CA, et al. Serotonin IA receptor binding and treatment response in late-life depression. Neuropsychopharmacology 2004; 29(12): 2258–2265. [DOI] [PubMed] [Google Scholar]
  • 46.Karlsson H, Hirvonen J, Kajander J, et al. Research letter: psychotherapy increases brain serotonin 5-HT 1A receptors in patients with major depressive disorder. Psychol Med 2010; 40(03): 523–528. [DOI] [PubMed] [Google Scholar]
  • 47.Bhagwagar Z, Montgomery AJ, Grasby PM, Cowen PJ. Lack of effect of a single dose of hydrocortisone on serotonin 1A receptors in recovered depressed patients measured by positron emission tomography with [11 C] WAY-100635. Biol Psychiatry 2003; 54(9): 890–895. [DOI] [PubMed] [Google Scholar]
  • 48.Bhagwagar Z, Rabiner E, Sargent P, Grasby P, Cowen P. Persistent reduction in brain serotonin1A receptor binding in recovered depressed men measured by positron emission tomography with & lsqb; 11C] WAY-100635. Mol Psychiatry 2004; 9(4): 386–392. [DOI] [PubMed] [Google Scholar]
  • 49.Stockmeier CA, Shapiro LA, Dilley GE, Kolli TN, Friedman L, Rajkowska G. Increase in serotonin-1A autoreceptors in the midbrain of suicide victims with major depression—postmortem evidence for decreased serotonin activity. J Neurosci 1998; 18(18): 7394–7401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Murrough JW, Henry S, Hu J, et al. Reduced ventral striatal/ventral pallidal serotonin1B receptor binding potential in major depressive disorder. Psychopharmacology 2011; 213(2–3): 547–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tiger M, Farde L, Rück C, et al. Low serotonin 1B receptor binding potential in the anterior cingulate cortex in drug-free patients with recurrent major depressive disorder. Psychiatry Res Neuroimaging 2016; 253: 36–42. [DOI] [PubMed] [Google Scholar]
  • 52.Tiger M, Rück C, Forsberg A, et al. Reduced 5-HT 1B receptor binding in the dorsal brain stem after cognitive behavioural therapy of major depressive disorder. Psychiatry Res Neuroimaging 2014; 223(2): 164–170. [DOI] [PubMed] [Google Scholar]
  • 53.Baeken C, De Raedt R, Bossuyt A, et al. The impact of HF-rTMS treatment on serotonin 2A receptors in unipolar melancholic depression. Brain Stimul 2011; 4(2): 104–111. [DOI] [PubMed] [Google Scholar]
  • 54.Biver F, Wikler D, Lotstra F, Damhaut P, Goldman S, Mendlewicz J. Serotonin 5-HT2 receptor imaging in major depression: focal changes in orbito-insular cortex. Br J Psychiatry 1997; 171(5): 444–448. [DOI] [PubMed] [Google Scholar]
  • 55.Mintun MA, Sheline YI, Moerlein SM, Vlassenko AG, Huang Y, Snyder AZ. Decreased hippocampal 5-HT 2A receptor binding in major depressive disorder: in vivo measurement with [18 F] altanserin positron emission tomography. Biol Psychiatry 2004; 55(3): 217–224. [DOI] [PubMed] [Google Scholar]
  • 56.Bhagwagar Z, Hinz R, Taylor M, Fancy S, Cowen P, Grasby P. Increased 5-HT 2A receptor binding in euthymic, medication-free patients recovered from depression: a positron emission study with [11 C] MDL 100,907. Am J Psychiatry 2006; 163(9): 1580–1587. [DOI] [PubMed] [Google Scholar]
  • 57.Messa C, Colombo C, Moresco RM, et al. 5-HT2A receptor binding is reduced in drug-naive and unchanged in SSRI-responder depressed patients compared to healthy controls: a PET study. Psychopharmacology 2003; 167(1): 72–78. [DOI] [PubMed] [Google Scholar]
  • 58.Yatham LN, Liddle PF, Shiah I, -S, et al. Brain serotonin2 receptors in major depression: a positron emission tomography study. Arch Gen Psychiatry 2000; 57(9): 850–858. [DOI] [PubMed] [Google Scholar]
  • 59.Meyer JH, Kapur S, Houle S, et al. Prefrontal cortex 5-HT2 receptors in depression: an [18F] setoperone PET imaging study. Am J Psychiatry 1999; 156(7): 1029–1034. [DOI] [PubMed] [Google Scholar]
  • 60.Meyer JH, McMain S, Kennedy SH, et al. Dysfunctional attitudes and 5-HT2 receptors during depression and self-harm. Am J Psychiatry 2003; 160(1): 90–99. [DOI] [PubMed] [Google Scholar]
  • 61.Yatham LN, Liddle PF, Dennie J, et al. Decrease in brain serotonin 2 receptor binding in patients with major depression following desipramine treatment: a positron emission tomography study with fluorine-18–labeled setoperone. Arch Gen Psychiatry 1999; 56(8): 705–711. [DOI] [PubMed] [Google Scholar]
  • 62.Madsen K, Torstensen E, Holst KK, et al. Familial risk for major depression is associated with lower striatal 5-HT4 receptor binding. Int J Neuropsychopharmacol 2014; 18: pyu034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Staley JK, Sanacora G, Tamagnan G, et al. Sex differences in diencephalon serotonin transporter availability in major depression. Biol Psychiatry 2006; 59(1): 40–47. [DOI] [PubMed] [Google Scholar]
  • 64.Meyer JH, Wilson AA, Sagrati S, et al. Serotonin transporter occupancy of five selective serotonin reuptake inhibitors at different doses: an [11C] DASB positron emission tomography study. Am J Psychiatry 2004; 161(5): 826–835. [DOI] [PubMed] [Google Scholar]
  • 65.Cannon DM, Ichise M, Rollis D, et al. Elevated serotonin transporter binding in major depressive disorder assessed using positron emission tomography and [11 C] DASB; comparison with bipolar disorder. Biol Psychiatry 2007; 62(8): 870–877. [DOI] [PubMed] [Google Scholar]
  • 66.Camardese G, Di Giuda D, Di Nicola M, et al. Imaging studies on dopamine transporter and depression: a review of literature and suggestions for future research. J Psychiatr Res 2014; 51: 7–18. [DOI] [PubMed] [Google Scholar]
  • 67.Joensuu M, Tolmunen T, Saarinen PI, et al. Reduced midbrain serotonin transporter availability in drug-naive patients with depression measured by SERT-specific [(123)I] nor-beta-CIT SPECT imaging. Psychiatry Res 2007; 154(2): 125–131. [DOI] [PubMed] [Google Scholar]
  • 68.Neumeister A, Willeit M, Praschak-Rieder N, et al. Dopamine transporter availability in symptomatic depressed patients with seasonal affective disorder and healthy controls. Psychol Med 2001; 31(08): 1467–1473. [DOI] [PubMed] [Google Scholar]
  • 69.Malison RT, Price LH, Berman R, et al. Reduced brain serotonin transporter availability in major depression as measured by [123 I]-2β-carbomethoxy-3β-(4-iodophenyl) tropane and single photon emission computed tomography. Biol Psychiatry 1998; 44(11): 1090–1098. [DOI] [PubMed] [Google Scholar]
  • 70.Mill J, Asherson P, Browes C, D’Souza U, Craig I. Expression of the dopamine transporter gene is regulated by the 3′ UTR VNTR: evidence from brain and lymphocytes using quantitative RT PCR. Am J Med Genet 2002; 114(8): 975–979. [DOI] [PubMed] [Google Scholar]
  • 71.Amsterdam JD, Newberg AB, Soeller I, Shults J. Greater striatal dopamine transporter density may be associated with major depressive episode. J Affect Disord 2012; 141(2): 425–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Brunswick DJ, Amsterdam JD, Mozley PD, Newberg A. Greater availability of brain dopamine transporters in major depression shown by [99mTc] TRODAT-1 SPECT imaging. Am J Psychiatry 2003; 160(10): 1836–1841. [DOI] [PubMed] [Google Scholar]
  • 73.Yang YK, Yeh TL, Yao WJ, et al. Greater availability of dopamine transporters in patients with major depression—a dual-isotope SPECT study. Psychiatry Res Neuroimaging 2008; 162(3): 230–235. [DOI] [PubMed] [Google Scholar]
  • 74.Hsieh PC, Lee IH, Yeh TL, et al. Distribution volume ratio of serotonin and dopamine transporters in euthymic patients with a history of major depression—a dual-isotope SPECT study. Psychiatry Res Neuroimaging 2010; 184(3): 157–161. [DOI] [PubMed] [Google Scholar]
  • 75.Árgyelán M, Szabó Z, Kanyó B, et al. Dopamine transporter availability in medication free and in bupropion treated depression: a 99m Tc-TRODAT-1 SPECT study. J Affect Disord 2005; 89(1): 115–123. [DOI] [PubMed] [Google Scholar]
  • 76.Meyer JH, Krüger S, Wilson AA, et al. Lower dopamine transporter binding potential in striatum during depression. Neuroreport 2001; 12(18): 4121–4125. [DOI] [PubMed] [Google Scholar]
  • 77.Lehto S, Tolmunen T, Joensuu M, et al. Midbrain binding of [123 I] nor-β-CIT in atypical depression. Prog Neuropsychopharmacol Biol Psychiatry 2006; 30(7): 1251–1255. [DOI] [PubMed] [Google Scholar]
  • 78.Wu H, Lou C, Huang Z, Shi G. SPECT imaging of dopamine transporters with 99mTc-TRODAT-1 in major depression and Parkinson’s disease. J Neuropsychiatry Clin Neurosci 2011; 23(1): 63–67. [DOI] [PubMed] [Google Scholar]
  • 79.Parsey RV, Oquendo MA, Zea-Ponce Y, et al. Dopamine D 2 receptor availability and amphetamine-induced dopamine release in unipolar depression. Biol Psychiatry 2001; 50(5): 313–322. [DOI] [PubMed] [Google Scholar]
  • 80.Papp M, Klimek V, Willner P. Parallel changes in dopamine D 2 receptor binding in limbic forebrain associated with chronic mild stress-induced anhedonia and its reversal by imipramine. Psychopharmacology 1994; 115(4): 441–446. [DOI] [PubMed] [Google Scholar]
  • 81.de Kwaasteniet BP, Pinto C, Ruhe EH, van Wingen GA, Booij J, Denys D. Striatal dopamine D 2/3 receptor availability in treatment resistant depression. PloS One 2014; 9(11): e113612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Meyer JH, McNeely HE, Sagrati S, et al. Elevated putamen D 2 receptor binding potential in major depression with motor retardation: an [11 C] raclopride positron emission tomography study. Am J Psychiatry 2006; 163(9): 1594–1602. [DOI] [PubMed] [Google Scholar]
  • 83.Larisch R, Klimke A, Vosberg H, Löffler S, Gaebel W, Müller-Gärtner H-W. In vivo evidence for the involvement of dopamine-D 2 receptors in striatum and anterior cingulate gyrus in major depression. Neuroimage 1997; 5(4): 251–260. [DOI] [PubMed] [Google Scholar]
  • 84.Sanacora G, Treccani G, Popoli M. Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 2012; 62(1): 63–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Dougherty DD, Bonab AA, Ottowitz WE, et al. Decreased striatal D1 binding as measured using PET and [11C] SCH 23,390 in patients with major depression with anger attacks. Depress Anxiety 2006; 23(3): 175–177. [DOI] [PubMed] [Google Scholar]
  • 86.Matosin N, Fernandez-Enright F, Frank E, et al. Metabotropic glutamate receptor mGluR2/3 and mGluR5 binding in the anterior cingulate cortex in psychotic and nonpsychotic depression, bipolar disorder and schizophrenia: implications for novel mGluR-based therapeutics. J Psychiatry Neurosci 2014; 39(6): 407–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.DeLorenzo C, Sovago J, Gardus J, et al. Characterization of brain mGluR5 binding in a pilot study of late-life major depressive disorder using positron emission tomography and [ 11C] ABP688. Transl Psychiatry 2015; 5(12): e693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zunszain PA, Hepgul N, Pariante CM. Inflammation and depression. In: Philip J Cowen, Trevor Sharp, Jennifer YF Lau. (eds). Behavioral Neurobiology of Depression and Its Treatment, Berlin, Germany: Springer, 2012, pp. 135–151. [Google Scholar]
  • 89.Setiawan E, Wilson AA, Mizrahi R, et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry 2015; 72(3): 268–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Holmes S, Hinz R, Gregory C, et al. Microglial activation in anterior cingulate in major depression and a role for inflammation in suicidal thinking: a PET study. Biol Psychiatry. Under Review. [DOI] [PubMed]
  • 91.Meyer JH, Ginovart N, Boovariwala A, et al. Elevated monoamine oxidase a levels in the brain: an explanation for the monoamine imbalance of major depression. Arch Gen Psychiatry 2006; 63(11): 1209–1216. [DOI] [PubMed] [Google Scholar]
  • 92.Meyer JH, Wilson AA, Sagrati S, et al. Brain monoamine oxidase A binding in major depressive disorder: relationship to selective serotonin reuptake inhibitor treatment, recovery, and recurrence. Arch Gen Psychiatry 2009; 66(12): 1304–1312. [DOI] [PubMed] [Google Scholar]
  • 93.Wu K-Y, Hsiao T, Chen C-S, et al. Increased brain amyloid deposition in patients with a lifetime history of major depression: evidenced on 18F-florbetapir (AV-45/Amyvid) positron emission tomography. Eur J Nucl Med Mol Imaging 2014; 41(4): 714–722. [DOI] [PubMed] [Google Scholar]
  • 94.Yasuno F, Kazui H, Morita N, et al. High amyloid-β deposition related to depressive symptoms in older individuals with normal cognition: a pilot study. Int J Geriatr Psychiatry 2016; 31(8): 920–928. [DOI] [PubMed] [Google Scholar]
  • 95.Mineur YS, Mose TN, Blakeman S, Picciotto MR. Hippocampal α7 nicotinic acetylcholine receptors contribute to modulation of depression-like behavior in C57BL/6 J mice. Br J Pharmacol. 8 April 2017. [DOI] [PMC free article] [PubMed]
  • 96.Croarkin PE, Levinson AJ, Daskalakis ZJ. Evidence for GABAergic inhibitory deficits in major depressive disorder. Neurosci Biobehav Rev 2011; 35(3): 818–825. [DOI] [PubMed] [Google Scholar]
  • 97.Schildkraut JJ. The catecholamine hypothesis of affective disorders: a review of supporting evidence. Am J Psychiatry 1965; 122(5): 509–522. [DOI] [PubMed] [Google Scholar]
  • 98.Ruhé HG, Visser AK, Frokjaer VG, Haarman BBC, Klein HC, Booij J. Molecular imaging of depressive disorders. In: Rudi AJO Dierckx, Andreas Otte, Erik FJ de Vries, Aren van Waarde, Johan A den Boer. (eds). PET and SPECT in Psychiatry, Berlin, Germany: Springer, 2014, pp. 93–172. [Google Scholar]
  • 99.Cai N, Chang S, Li Y, et al. Molecular signatures of major depression. Curr Biol 2015; 25(9): 1146–1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Domínguez-López S, Howell R, Gobbi G. Characterization of serotonin neurotransmission in knockout mice: implications for major depression. Rev Neurosci 2012; 23(4): 429–443. [DOI] [PubMed] [Google Scholar]
  • 101.Kambeitz JP, Howes OD. The serotonin transporter in depression: meta-analysis of in vivo and post mortem findings and implications for understanding and treating depression. J Affect Disord 2015; 186: 358–366. [DOI] [PubMed] [Google Scholar]
  • 102.Pirker W, Asenbaum S, Kasper S, et al. β-CIT SPECT demonstrates blockade of 5HT-uptake sites by citalopram in the human brain in vivo. J Neural Transm Gen Sect 1995; 100(3): 247–256. [DOI] [PubMed] [Google Scholar]
  • 103.Bennett AJ, Lesch KP, Heils A, et al. Early experience and serotonin transporter gene variation interact to influence primate CNS function. Mol Psychiatry 2002; 7(1): 118. [DOI] [PubMed] [Google Scholar]
  • 104.Caspi A, Sugden K, Moffitt TE, et al. Influence of life stress on depression: moderation by a polymorphism in the 5-HTT gene. Science 2003; 301(5631): 386–389. [DOI] [PubMed] [Google Scholar]
  • 105.Zhang K, Zhu Y, Zhu Y, et al. Molecular, functional, and structural imaging of major depressive disorder. Neurosci Bull 2016; 32(3): 273–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lin SH, Lee IH, Chen KC, et al. Serotonin transporter availability may moderate the association between perceiving stress and depressive tendencies—A SPECT with 5-HTTLPR genotyping study. Prog Neuropsychopharmacol Biol Psychiatry 2015; 61: 24–29. [DOI] [PubMed] [Google Scholar]
  • 107.Tsai A, McDonald AG, Rosenberg AE, Stamoulis C, Kleinman PK. Discordant radiologic and histological dimensions of the zone of provisional calcification in fetal piglets. Pediatr Radiol 2013; 43(12): 1606–1614. [DOI] [PubMed] [Google Scholar]
  • 108.Yeh YW, Ho PS, Kuo SC, et al. Disproportionate reduction of serotonin transporter may predict the response and adherence to antidepressants in patients with major depressive disorder: a positron emission tomography study with 4-[18F]-ADAM. Int J Neuropsychopharmacol 2015; 18(7): pyu120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Laasonen-Balk T, Viinamaki H, Kuikka JT, Husso-Saastamoinen M, Lehtonen J, Tiihonen J. 123I-beta-CIT binding and recovery from depression. A six-month follow-up study. Eur Arch Psychiatry Clin Neurosci 2004; 254(3): 152–155. [DOI] [PubMed] [Google Scholar]
  • 110.Amsterdam JD, Newberg AB, Newman CF, Shults J, Wintering N, Soeller I. Change over time in brain serotonin transporter binding in major depression: effects of therapy measured with [(123) I]-ADAM SPECT. J Neuroimaging 2013; 23(4): 469–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Lanzenberger R, Kranz GS, Haeusler D, et al. Prediction of SSRI treatment response in major depression based on serotonin transporter interplay between median raphe nucleus and projection areas. Neuroimage 2012; 63(2): 874–881. [DOI] [PubMed] [Google Scholar]
  • 112.Rominger A, Cumming P, Brendel M, et al. Altered serotonin and dopamine transporter availabilities in brain of depressed patients upon treatment with escitalopram: a [123 I] β-CIT SPECT study. Eur Neuropsychopharmacol 2015; 25(6): 873–881. [DOI] [PubMed] [Google Scholar]
  • 113.Reivich M, Amsterdam JD, Brunswick DJ, Yann Shiue C. PET brain imaging with [11C](+)McN5652 shows increased serotonin transporter availability in major depression. J Affect Disord 2004; 82(2): 321–327. [DOI] [PubMed] [Google Scholar]
  • 114.Catafau AM, Perez V, Plaza P, et al. Serotonin transporter occupancy induced by paroxetine in patients with major depression disorder: a 123I-ADAM SPECT study. Psychopharmacology (Berl) 2006; 189(2): 145–153. [DOI] [PubMed] [Google Scholar]
  • 115.Ichimiya T, Suhara T, Sudo Y, et al. Serotonin transporter binding in patients with mood disorders: a PET study with [11 C](+) McN5652. Biol Psychiatry 2002; 51(9): 715–722. [DOI] [PubMed] [Google Scholar]
  • 116.Baldwin RM, Zea-Ponce Y, Laurelle M, et al. Evaluation of the monoamine uptake site ligand [131I] methyl 3β-(4-Iodophenyl)-tropane-2β-carboxylate ([123I] β-CIT) in non-human primates: pharmacokinetics, biodistribution and SPECT brain imaging coregistered with MRI. Nucl Med Biol 1993; 20(5): 597–606. [DOI] [PubMed] [Google Scholar]
  • 117.Savitz JB, Drevets WC. Neuroreceptor imaging in depression. Neurobiol Dis 2013; 52: 49–65. [DOI] [PubMed] [Google Scholar]
  • 118.Richardson-Jones JW, Craige CP, Guiard BP, et al. 5-HT 1A autoreceptor levels determine vulnerability to stress and response to antidepressants. Neuron 2010; 65(1): 40–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Naudon L, El Yacoubi M, Vaugeois J-M, Leroux-Nicollet I, Costentin J. A chronic treatment with fluoxetine decreases 5-HT 1A receptors labeling in mice selected as a genetic model of helplessness. Brain Res 2002; 936(1): 68–75. [DOI] [PubMed] [Google Scholar]
  • 120.Parsey RV, Ogden RT, Miller JM, et al. Higher serotonin 1A binding in a second major depression cohort: modeling and reference region considerations. Biol Psychiatry 2010; 68(2): 170–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Miller JM, Hesselgrave N, Ogden RT, et al. Brain serotonin 1A receptor binding as a predictor of treatment outcome in major depressive disorder. Biol Psychiatry 2013; 74(10): 760–767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Hesselgrave N, Parsey RV. Imaging the serotonin 1A receptor using [11C] WAY100635 in healthy controls and major depression. Phil Trans R Soc B 2013; 368(1615): 20120004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Tatarczyńska E, Antkiewicz-Michaluk L, Kłodzińska A, Stachowicz K, Chojnacka-Wójcik E. Antidepressant-like effect of the selective 5-HT 1B receptor agonist CP 94253: a possible mechanism of action. Eur J Pharmacol 2005; 516(1): 46–50. [DOI] [PubMed] [Google Scholar]
  • 124.Redrobe J, Bourin M. Evidence of the activity of lithium on 5-HT1B receptors in the mouse forced swimming test: comparison with carbamazepine and sodium valproate. Psychopharmacology 1999; 141(4): 370–377. [DOI] [PubMed] [Google Scholar]
  • 125.Carr GV, Lucki I. The role of serotonin receptor subtypes in treating depression: a review of animal studies. Psychopharmacology 2011; 213(2–3): 265–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Couch Y, Anthony DC, Dolgov O, et al. Microglial activation, increased TNF and SERT expression in the prefrontal cortex define stress-altered behaviour in mice susceptible to anhedonia. Brain Behav Immun 2013; 29: 136–146. [DOI] [PubMed] [Google Scholar]
  • 127.Benekareddy M, Vadodaria KC, Nair AR, Vaidya VA. Postnatal serotonin type 2 receptor blockade prevents the emergence of anxiety behavior, dysregulated stress-induced immediate early gene responses, and specific transcriptional changes that arise following early life stress. Biol Psychiatry 2011; 70(11): 1024–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Alam M, Huang HD, Shahzad SA, et al. Percutaneous coronary intervention vs. coronary artery bypass graft surgery for unprotected left main coronary artery disease in the drug-eluting stents era–an aggregate data meta-analysis of 11,148 patients. Circ J 2013; 77(2): 372–382. [DOI] [PubMed] [Google Scholar]
  • 129.Couch Y, Trofimov A, Markova N, et al. Low-dose lipopolysaccharide (LPS) inhibits aggressive and augments depressive behaviours in a chronic mild stress model in mice. J Neuroinflammation 2016; 13(1): 108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Attar-Lévy D, Martinot J-L, Blin J, et al. The cortical serotonin 2 receptors studied with positron-emission tomography and [18 F]-setoperone during depressive illness and antidepressant treatment with clomipramine. Biol Psychiatry 1999; 45(2): 180–186. [DOI] [PubMed] [Google Scholar]
  • 131.D’haenen HA, Bossuyt A. Dopamine D 2 receptors in depression measured with single photon emission computed tomography. Biol Psychiatry 1994; 35(2): 128–132. [DOI] [PubMed] [Google Scholar]
  • 132.Dwivedi Y, Mondal AC, Payappagoudar GV, Rizavi HS. Differential regulation of serotonin (5HT) 2A receptor mRNA and protein levels after single and repeated stress in rat brain: role in learned helplessness behavior. Neuropharmacology 2005; 48(2): 204–214. [DOI] [PubMed] [Google Scholar]
  • 133.Vidal R, Castro E, Pilar-Cuellar F, et al. Serotonin 5-HT4 receptors: a new strategy for developing fast acting antidepressants? Curr Pharm Des 2014; 20(23): 3751–3762. [DOI] [PubMed] [Google Scholar]
  • 134.Nutt DJ. Relationship of neurotransmitters to the symptoms of major depressive disorder. J Clin Psychiatry 2008; 69(suppl E1): 4–7. [PubMed] [Google Scholar]
  • 135.Dunlop BW, Nemeroff CB. The role of dopamine in the pathophysiology of depression. Arch Gen Psychiatry 2007; 64(3): 327–337. [DOI] [PubMed] [Google Scholar]
  • 136.Klimke A, Larisch R, Janz A, Vosberg H, Müller-Gärtner H-W, Gaebel W. Dopamine D 2 receptor binding before and after treatment of major depression measured by [123 I] IBZM SPECT. Psychiatry Res Neuroimaging 1999; 90(2): 91–101. [DOI] [PubMed] [Google Scholar]
  • 137.Bowers M, Heninger G, Gerbode F. Cerebrospinal fluid 5-hydroxyindoleacetic acid and homovanillic acid in psychiatric patients. Int J Neuropharmacol 1969; 8(3): 255–262. [DOI] [PubMed] [Google Scholar]
  • 138.Martinot M-LP, Bragulat V, Artiges E, et al. Decreased presynaptic dopamine function in the left caudate of depressed patients with affective flattening and psychomotor retardation. Am J Psychiatry 2001; 158(2): 314–316. [DOI] [PubMed] [Google Scholar]
  • 139.Opmeer EM, Kortekaas R, Aleman A. Depression and the role of genes involved in dopamine metabolism and signalling. Prog Neurobiol 2010; 92(2): 112–133. [DOI] [PubMed] [Google Scholar]
  • 140.Hsiao M-C, Lin K-J, Liu C-Y, Schatz DB. The interaction between dopamine transporter function, gender differences, and possible laterality in depression. Psychiatry Res Neuroimaging 2013; 211(1): 72–77. [DOI] [PubMed] [Google Scholar]
  • 141.Sarchiapone M, Carli V, Camardese G, et al. Dopamine transporter binding in depressed patients with anhedonia. Psychiatry Res Neuroimaging 2006; 147(2): 243–248. [DOI] [PubMed] [Google Scholar]
  • 142.Shah P, Ogilvie A, Goodwin G, Ebmeier K. Clinical and psychometric correlates of dopamine D 2 binding in depression. Psychol Med 1997; 27(06): 1247–1256. [DOI] [PubMed] [Google Scholar]
  • 143.Ebert D, Loew T, Feistel H, Pirner A. Dopamine and depression—striatal dopamine D2 receptor SPECT before and after antidepressant therapy. Psychopharmacology 1996; 126(1): 91–94. [DOI] [PubMed] [Google Scholar]
  • 144.Cannon DM, Klaver JM, Peck SA, Rallis-Voak D, Erickson K, Drevets WC. Dopamine type-1 receptor binding in major depressive disorder assessed using positron emission tomography and [ 11C] NNC-112. Neuropsychopharmacology 2009; 34(5): 1277–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Krystal JH, Mathew SJ, D’Souza DC, Garakani A, Gunduz-Bruce H, Charney DS. Potential psychiatric applications of metabotropic glutamate receptor agonists and antagonists. CNS Drugs 2010; 24(8): 669–693. [DOI] [PubMed] [Google Scholar]
  • 146.Brodkin J, Busse C, Sukoff SJ, Varney MA. Anxiolytic-like activity of the mGluR5 antagonist MPEP: a comparison with diazepam and buspirone. Pharmacol Biochem Behav 2002; 73(2): 359–366. [DOI] [PubMed] [Google Scholar]
  • 147.Pałucha A, Brański P, Szewczyk B, Wierońska JM, Kłak K, Pilc A. Potential antidepressant-like effect of MTEP, a potent and highly selective mGluR5 antagonist. Pharmacol Biochem Behav 2005; 81(4): 901–906. [DOI] [PubMed] [Google Scholar]
  • 148.Belozertseva I, Kos T, Popik P, Danysz W, Bespalov A. Antidepressant-like effects of mGluR1 and mGluR5 antagonists in the rat forced swim and the mouse tail suspension tests. Eur Neuropsychopharmacol 2007; 17(3): 172–179. [DOI] [PubMed] [Google Scholar]
  • 149.Li X, Need AB, Baez M, Witkin JM. Metabotropic glutamate 5 receptor antagonism is associated with antidepressant-like effects in mice. J Pharm Exp Ther 2006; 319(1): 254–259. [DOI] [PubMed] [Google Scholar]
  • 150.Tatarczyńska E, Kłodzińska A, Chojnacka-Wójcik E, et al. Potential anxiolytic and antidepressant like effects of MPEP, a potent, selective and systemically active mGlu5 receptor antagonist. Br J Pharmacol 2001; 132(7): 1423–1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Pilc A, Kłodzińska A, Brański P, et al. Multiple MPEP administrations evoke anxiolytic-and antidepressant-like effects in rats. Neuropharmacology 2002; 43(2): 181–187. [DOI] [PubMed] [Google Scholar]
  • 152.Deschwanden A, Karolewicz B, Feyissa AM, et al. Reduced metabotropic glutamate receptor 5 density in major depression determined by [11C] ABP688 PET and postmortem study. Am J Psychiatry 2011; 168(7): 727–734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Esterlis I. Ketamine-induced reduction in mGluR5 availability is associated with an antidepressant response: an [11C]ABP688 and PET imaging study in depression. Mol Psychiatry. Under Review. [DOI] [PMC free article] [PubMed]
  • 154.Abdallah CG, Jonas Hannestad, Graeme F Mason, Sophie E Holmes, Nicole DellaGioia, Gerard Sanacora, et al. mGluR5 and glutamate involvement in MDD: a multimodal imaging study [published online ahead of print April 4, 2017]. Biol Psychiatr: Cognit Neurosci Neuroimaging. 2017. [DOI] [PMC free article] [PubMed]
  • 155.Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nature Rev Neurosci 2008; 9(1): 46–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Bufalino C, Hepgul N, Aguglia E, Pariante CM. The role of immune genes in the association between depression and inflammation: a review of recent clinical studies. Brain Behav Immun 2013; 31: 31–47. [DOI] [PubMed] [Google Scholar]
  • 157.Zorrilla EP, Luborsky L, McKay JR, et al. The relationship of depression and stressors to immunological assays: a meta-analytic review. Brain Behav Immun 2001; 15(3): 199–226. [DOI] [PubMed] [Google Scholar]
  • 158.Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med 2009; 71(2): 171–186. [DOI] [PubMed] [Google Scholar]
  • 159.Dowlati Y, Herrmann N, Swardfager W, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry 2010; 67(5): 446–457. [DOI] [PubMed] [Google Scholar]
  • 160.Haapakoski R, Mathieu J, Ebmeier KP, Alenius H, Kivimäki M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behav Immun 2015; 49: 206–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kraft AD, Harry GJ. Features of microglia and neuroinflammation relevant to environmental exposure and neurotoxicity. Int J Environ Res Public Health 2011; 8(7): 2980–3018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Hannestad J, DellaGioia N, Gallezot J-D, et al. The neuroinflammation marker translocator protein is not elevated in individuals with mild-to-moderate depression: a [(11)C]PBR28 PET study. Brain Behav Immun 2013; 33: 131–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Mineur YS, Obayemi A, Wigestrand MB, et al. Cholinergic signaling in the hippocampus regulates social stress resilience and anxiety-and depression-like behavior. Proc Natl Acad Sci U S A 2013; 110(9): 3573–3578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Caldarone BJ, Harrist A, Cleary MA, Beech RD, King SL, Picciotto MR. High-affinity nicotinic acetylcholine receptors are required for antidepressant effects of amitriptyline on behavior and hippocampal cell proliferation. Biol Psychiatry 2004; 56(9): 657–664. [DOI] [PubMed] [Google Scholar]
  • 165.Janowsky D, Davis J, El-Yousef MK, Sekerke HJ. A cholinergic-adrenergic hypothesis of mania and depression. Lancet 1972; 300(7778): 632–635. [DOI] [PubMed] [Google Scholar]
  • 166.Overstreet DH, Rezvani AH, Janowsky DS. Genetic animal models of depression and ethanol preference provide support for cholinergic and serotonergic involvement in depression and alcoholism. Biol Psychiatry 1992; 31(9): 919–936. [DOI] [PubMed] [Google Scholar]
  • 167.Dilsaver SC. Pathophysiology of “cholinoceptor supersensitivity” in affective disorders. Biol Psychiatry 1986; 21(8–9): 813–829. [DOI] [PubMed] [Google Scholar]
  • 168.Saricicek A, Esterlis I, Maloney KH, et al. Persistent β2*-nicotinic acetylcholinergic receptor dysfunction in major depressive disorder. Am J Psychiatry 2012; 169(8): 851–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Pandey GN, Pandey SC, Davis JM. Interaction of antidepressant drugs with phosphoinositide turnover in human platelets and rat brain. Clin Neuropharmacol 1992; 15(Suppl 1 Pt A): 202A–203A. [DOI] [PubMed] [Google Scholar]
  • 170.Nye JA, Purselle D, Plisson C, et al. Decreased brainstem and putamen SERT binding potential in depressed suicide attempters using [11C]-zient PET imaging. Depress Anxiety 2013; 30(10): 902–907. [DOI] [PubMed] [Google Scholar]
  • 171.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(10): 1048–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Leyton M, Paquette V, Gravel P, et al. α-[11 C] Methyl-l-tryptophan trapping in the orbital and ventral medial prefrontal cortex of suicide attempters. Eur Neuropsychopharmacol 2006; 16(3): 220–223. [DOI] [PubMed] [Google Scholar]
  • 173.Ellicott A, Hammen C, Gitlin M, Brown G, Jamison K. Life events and the course of bipolar disorder. Am J Psychiatry 1990; 147(9): 1194–1198. [DOI] [PubMed] [Google Scholar]
  • 174.Malkoff-Schwartz S, Frank E, Anderson B, et al. Social rhythm disruption and stressful life events in the onset of bipolar and unipolar episodes. Psychol Med 2000; 30(5): 1005–1016. [DOI] [PubMed] [Google Scholar]
  • 175.Cohen AN, Hammen C, Henry RM, Daley SE. Effects of stress and social support on recurrence in bipolar disorder. J Affect Disord 2004; 82(1): 143–147. [DOI] [PubMed] [Google Scholar]
  • 176.Bender RE, Alloy LB. Life stress and kindling in bipolar disorder: review of the evidence and integration with emerging biopsychosocial theories. Clin Psychol Rev 2011; 31(3): 383–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Horesh N, Apter A, Zalsman G. Timing, quantity and quality of stressful life events in childhood and preceding the first episode of bipolar disorder. J Affect Disord 2011; 134(1): 434–437. [DOI] [PubMed] [Google Scholar]
  • 178.Yeh YW, Ho PS, Chen CY, et al. Suicidal ideation modulates the reduction in serotonin transporter availability in male military conscripts with major depression: a 4-[18F]-ADAM PET study. World J Biol Psychiatry 2015; 16(7): 502–512. [DOI] [PubMed] [Google Scholar]
  • 179.Chou Y-H, Hsieh W-C, Chen L-C, Lirng J-F, Wang S-J. Association between the serotonin transporter and cytokines: implications for the pathophysiology of bipolar disorder. J Affect Disord 2016; 191: 29–35. [DOI] [PubMed] [Google Scholar]
  • 180.Chou Y-H, Lin C-L, Wang S-J, et al. Aggression in bipolar II disorder and its relation to the serotonin transporter. J Affect Disord 2013; 147(1): 59–63. [DOI] [PubMed] [Google Scholar]
  • 181.Chou Y-H, Wang S-J, Lirng J-F, et al. Impaired cognition in bipolar I disorder: the roles of the serotonin transporter and brain-derived neurotrophic factor. J Affect Disord 2012; 143(1): 131–137. [DOI] [PubMed] [Google Scholar]
  • 182.Chou YH, Wang SJ, Lin CL, Mao WC, Lee SM, Liao MH. Decreased brain serotonin transporter binding in the euthymic state of bipolar I but not bipolar II disorder: a SPECT study. Bipolar Disord 2010; 12(3): 312–318. [DOI] [PubMed] [Google Scholar]
  • 183.Hsieh W, Jou Y, Lin J, Wang S, Chou Y. The effect of cortisol and BDNF on serotonin transporter in bipolar I disorder. Paper presented at: 16 Annual conference in bipolar disorders, International Society for Bipolar Disorders, Wiley, 2014.
  • 184.Miller JM, Everett BA, Oquendo MA, Ogden RT, Mann JJ, Parsey RV. Positron emission tomography quantification of serotonin transporter binding in medication free bipolar disorder. Synapse 2016; 70(1): 24–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Yatham LN, Liddle PF, Erez J, et al. Brain serotonin-2 receptors in acute mania. Br J Psychiatry 2010; 196(1): 47–51. [DOI] [PubMed] [Google Scholar]
  • 186.Yatham LN, Liddle PF, Lam RW, et al. A positron emission tomography study of the effects of treatment with valproate on brain 5 HT2A receptors in acute mania. Bipolar Disord 2005; 7(s5): 53–57. [DOI] [PubMed] [Google Scholar]
  • 187.Sargent PA, Rabiner EA, Bhagwagar Z, et al. 5-HT 1A receptor binding in euthymic bipolar patients using positron emission tomography with [carbonyl-11 C] WAY-100635. J Affect Disord 2010; 123(1): 77–80. [DOI] [PubMed] [Google Scholar]
  • 188.Nugent AC, Bain EE, Carlson PJ, et al. Reduced post-synaptic serotonin type 1A receptor binding in bipolar depression. Eur Neuropsychopharmacol 2013; 23(8): 822–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Amsterdam JD, Newberg AB. A preliminary study of dopamine transporter binding in bipolar and unipolar depressed patients and healthy controls. Neuropsychobiology 2007; 55(3–4): 167–170. [DOI] [PubMed] [Google Scholar]
  • 190.Chang TT, Yeh TL, Chiu NT, et al. Higher striatal dopamine transporters in euthymic patients with bipolar disorder: a SPECT study with [99mTc] TRODAT 1. Bipolar Disord 2010; 12(1): 102–106. [DOI] [PubMed] [Google Scholar]
  • 191.Yatham LN, Liddle PF, Lam RW, et al. PET study of the effects of valproate on dopamine D2 receptors in neuroleptic-and mood-stabilizer-naive patients with nonpsychotic mania. Am J Psychiatry 2002; 159(10): 1718–1723. [DOI] [PubMed] [Google Scholar]
  • 192.Hannestad JO, Cosgrove KP, DellaGioia NF, et al. Changes in the cholinergic system between bipolar depression and euthymia as measured with [123 I] 5IA single photon emission computed tomography. Biol Psychiatry 2013; 74(10): 768–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Cannon DM, Carson RE, Nugent AC, et al. Reduced muscarinic type 2 receptor binding in subjects with bipolar disorder. Arch Gen Psychiatry 2006; 63(7): 741–747. [DOI] [PubMed] [Google Scholar]
  • 194.Cannon DM, Klaver J, Gandhi SK, et al. Genetic variation in cholinergic muscarinic-2 receptor gene modulates M2 receptor binding in vivo and accounts for reduced binding in bipolar disorder. Mol Psychiatry 2011; 16(4): 407–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Prange AJ, Wilson IC, Lynn CW, Alltop LB, Stikeleather RA. L-tryptophan in mania: contribution to a permissive hypothesis of affective disorders. Arch Gen Psychiatry 1974; 30(1): 56–62. [DOI] [PubMed] [Google Scholar]
  • 196.Gao J, Jia M, Qiao D, et al. TPH2 gene polymorphisms and bipolar disorder: a meta-analysis. Am J Med Genet B Neuropsychiatr Genet 2016; 171B(2): 145–152. [DOI] [PubMed] [Google Scholar]
  • 197.Chou P-H, Tseng W-J, Chen L-M, Lin C-C, Lan T-H, Chan C-H. Late onset bipolar disorder: a case report and review of the literature. J Clin Gerontol Geriatr 2015; 6(1): 27–29. [Google Scholar]
  • 198.Fountoulakis KN, Kelsoe JR, Akiskal H. Receptor targets for antidepressant therapy in bipolar disorder: an overview. J Affect Disord 2012; 138(3): 222–238. [DOI] [PubMed] [Google Scholar]
  • 199.Mamo D, Kapur S, Shammi C, et al. A PET study of dopamine D2 and serotonin 5-HT2 receptor occupancy in patients with schizophrenia treated with therapeutic doses of ziprasidone. Am J Psychiatry 2004; 161(5): 818–825. [DOI] [PubMed] [Google Scholar]
  • 200.Kishi T, Yoshimura R, Fukuo Y, et al. The serotonin 1A receptor gene confer susceptibility to mood disorders: results from an extended meta-analysis of patients with major depression and bipolar disorder. Eur Arch Psychiatry Clin Neurosci 2013; 263(2): 105–118. [DOI] [PubMed] [Google Scholar]
  • 201.López-Figueroa AL, Norton CS, López-Figueroa MO, et al. Serotonin 5-HT 1A, 5-HT 1B, and 5-HT 2A receptor mRNA expression in subjects with major depression, bipolar disorder, and schizophrenia. Biol Psychiatry 2004; 55(3): 225–233. [DOI] [PubMed] [Google Scholar]
  • 202.Lan MJ, Hesselgrave N, Ciarleglio A, et al. Higher pretreatment 5 HT1A receptor binding potential in bipolar disorder depression is associated with treatment remission: a naturalistic treatment pilot PET study. Synapse 2013; 67(11): 773–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Berk M, Dodd S, Kauer-Sant’Anna M, et al. Dopamine dysregulation syndrome: implications for a dopamine hypothesis of bipolar disorder. Acta Psychiatrica Scandinavica 2007; 116(s434): 41–49. [DOI] [PubMed] [Google Scholar]
  • 204.El-Mallakh RS, Wyatt RJ. The Na, K-ATPase hypothesis for bipolar illness. Biol Psychiatry 1995; 37(4): 235–244. [DOI] [PubMed] [Google Scholar]
  • 205.Peet M. Induction of mania with selective serotonin re-uptake inhibitors and tricyclic antidepressants. Br J Psychiatry 1994; 164(4): 549–550. [DOI] [PubMed] [Google Scholar]
  • 206.Bottlender R, Rudolf D, Strauss A, Möller H-J. Antidepressant-associated maniform states in acute treatment of patients with bipolar-I depression. Eur Arch Psychiatry Clin Neurosci 1998; 248(6): 296–300. [DOI] [PubMed] [Google Scholar]
  • 207.Zhan L, Kerr J, Lafuente MJ, et al. Altered expression and coregulation of dopamine signalling genes in schizophrenia and bipolar disorder. Neuropathol Appl Neurobiol 2011; 37(2): 206–219. [DOI] [PubMed] [Google Scholar]
  • 208.Anand A, Barkay G, Dzemidzic M, et al. Striatal dopamine transporter availability in unmedicated bipolar disorder. Bipolar Disord 2011; 13(4): 406–413. [DOI] [PubMed] [Google Scholar]
  • 209.Greenwood T, Schork NJ, Eskin E, Kelsoe JR. Identification of additional variants within the human dopamine transporter gene provides further evidence for an association with bipolar disorder in two independent samples. Mol Psychiatry 2006; 11(2): 125–133. [DOI] [PubMed] [Google Scholar]
  • 210.Greenwood TA, Alexander M, Keck PE, et al. Evidence for linkage disequilibrium between the dopamine transporter and bipolar disorder. Am J Med Genet 2001; 105(2): 145–151. [DOI] [PubMed] [Google Scholar]
  • 211.Anderson G, Maes M. Bipolar disorder: role of immune-inflammatory cytokines, oxidative and nitrosative stress and tryptophan catabolites. Curr Psychiatry Rep 2015; 17(2): 1–9. [DOI] [PubMed] [Google Scholar]
  • 212.Rosenblat JD, McIntyre RS. Bipolar disorder and inflammation. Psychiatr Clin North Am 2016; 39(1): 125–137. [DOI] [PubMed] [Google Scholar]
  • 213.Rao JS, Harry GJ, Rapoport SI, Kim H-W. Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol Psychiatry 2010; 15(4): 384–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Haarman BC, Riemersma-Van der Lek RF, de Groot JC, et al. Neuroinflammation in bipolar disorder—a [(11)C]-(R)-PK11195 positron emission tomography study. Brain Behav Immun 2014; 40: 219–225. [DOI] [PubMed] [Google Scholar]
  • 215.Post R, Ketter T, Denicoff K, et al. The place of anticonvulsant therapy in bipolar illness. Psychopharmacology 1996; 128(2): 115–129. [DOI] [PubMed] [Google Scholar]
  • 216.Association AP. DSM 5. Washington, DC: American Psychiatric Association; 2013.
  • 217.Davidson LM, Baum A. Chronic stress and posttraumatic stress disorders. J Consult Clin Psychol 1986; 54(3): 303. [DOI] [PubMed] [Google Scholar]
  • 218.Murrough JW, Huang Y, Hu J, et al. Reduced amygdala serotonin transporter binding in posttraumatic stress disorder. Biol Psychiatry 2011; 70(11): 1033–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Frick A, Ahs F, Linnman C, et al. Increased neurokinin-1 receptor availability in the amygdala in social anxiety disorder: a positron emission tomography study with [11C] GR205171. Transl Psychiatry 2015; 5: e597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Frick A, Åhs F, Palmquist ÅM, et al. Overlapping expression of serotonin transporters and neurokinin-1 receptors in posttraumatic stress disorder: a multi-tracer PET study. Mol Psychiatry 2015; 21(10): 1400–1407. [DOI] [PubMed] [Google Scholar]
  • 221.Bonne O, Bain E, Neumeister A, et al. No change in serotonin type 1A receptor binding in patients with posttraumatic stress disorder. Am J Psychiatry 2005; 162(2): 383–385. [DOI] [PubMed] [Google Scholar]
  • 222.Sullivan GM, Ogden RT, Huang Y-Y, Oquendo MA, Mann JJ, Parsey RV. Higher in VIVO Serotonin 1A binding in posttraumatic stress disorder: a pet study with [11C] way 100635. Depress Anxiety 2013; 30(3): 197–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Hoexter MQ, Fadel G, Felício AC, et al. Higher striatal dopamine transporter density in PTSD: an in vivo SPECT study with [99mTc] TRODAT-1. Psychopharmacology 2012; 224(2): 337–345. [DOI] [PubMed] [Google Scholar]
  • 224.Bremner JD, Innis RB, Southwick SM, Staib L, Zoghbi S, Charney DS. Decreased benzodiazepine receptor binding in prefrontal cortex in combat-related posttraumatic stress disorder. Am J Psychiatry 2000; 157(7): 1120–1126. [DOI] [PubMed] [Google Scholar]
  • 225.Geuze E, Van Berckel B, Lammertsma A, et al. Reduced GABAA benzodiazepine receptor binding in veterans with post-traumatic stress disorder. Mol Psychiatry 2008; 13(1): 74–83. [DOI] [PubMed] [Google Scholar]
  • 226.Neumeister A, Normandin MD, Pietrzak RH, et al. Elevated brain cannabinoid CB1 receptor availability in post-traumatic stress disorder: a positron emission tomography study. Mol Psychiatry 2013; 18(9): 1034–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Pietrzak RH, Huang Y, Corsi-Travali S, et al. Cannabinoid type 1 receptor availability in the amygdala mediates threat processing in trauma survivors. Neuropsychopharmacology 2014; 39(11): 2519–2528. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 228.Redpath HL, Cooper D, Lawrie SM. Imaging symptoms and syndromes: similarities and differences between schizophrenia and bipolar disorder. Biol Psychiatry 2013; 73: 495–496. [DOI] [PubMed] [Google Scholar]
  • 229.Czermak C, Staley JK, Kasserman S, et al. β2 Nicotinic acetylcholine receptor availability in post-traumatic stress disorder. Int J Neuropsychopharmacol 2008; 11(3): 419–424. [DOI] [PubMed] [Google Scholar]
  • 230.Krystal JH, Neumeister A. Noradrenergic and serotonergic mechanisms in the neurobiology of posttraumatic stress disorder and resilience. Brain Res 2009; 1293: 13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Bruening S, Oh E, Hetzenauer A, et al. The anxiety like phenotype of 5 HT1A receptor null mice is associated with genetic background specific perturbations in the prefrontal cortex GABA–glutamate system. J Neurochem 2006; 99(3): 892–899. [DOI] [PubMed] [Google Scholar]
  • 232.Amato JL, Bankson MG, Yamamoto BK. Prior exposure to chronic stress and MDMA potentiates mesoaccumbens dopamine release mediated by the 5-HT1B receptor. Neuropsychopharmacology 2007; 32(4): 946–954. [DOI] [PubMed] [Google Scholar]
  • 233.Mitsushima D, Yamada K, Takase K, Funabashi T, Kimura F. Sex differences in the basolateral amygdala: the extracellular levels of serotonin and dopamine, and their responses to restraint stress in rats. Eur J Neurosci 2006; 24(11): 3245–3254. [DOI] [PubMed] [Google Scholar]
  • 234.Gressier F, Calati R, Balestri M, et al. The 5 HTTLPR polymorphism and posttraumatic stress disorder: a meta analysis. J Trauma Stress 2013; 26(6): 645–653. [DOI] [PubMed] [Google Scholar]
  • 235.Klemenhagen KC, Gordon JA, David DJ, Hen R, Gross CT. Increased fear response to contextual cues in mice lacking the 5-HT1A receptor. Neuropsychopharmacology 2006; 31(1): 101–111. [DOI] [PubMed] [Google Scholar]
  • 236.Bolaños-Jiménez F, de Castro RM, Seguin L, et al. Effects of stress on the functional properties of pre-and postsynaptic 5-HT 1B receptors in the rat brain. Eur J Pharmacol 1995; 294(2): 531–540. [DOI] [PubMed] [Google Scholar]
  • 237.Charney DS, Deutch AY, Krystal JH, Southwick SM, Davis M. Psychobiologic mechanisms of posttraumatic stress disorder. Arch Gen Psychiatry 1993; 50(4): 294–305. [DOI] [PubMed] [Google Scholar]
  • 238.Segman R, Cooper-Kazaz R, Macciardi F, et al. Association between the dopamine transporter gene and posttraumatic stress disorder. Mol Psychiatry 2002; 7(8): 903. [DOI] [PubMed] [Google Scholar]
  • 239.Weizman R, Laor N, Karp L, et al. Alteration of platelet benzodiazepine receptors by stress of war. Am J Psychiatry 1994; 151(5): 766–767. [DOI] [PubMed] [Google Scholar]
  • 240.Hill MN, Gorzalka BB. The endocannabinoid system and the treatment of mood and anxiety disorders. CNS Neurol Disord Drug Targets 2009; 8(6): 451–458. [DOI] [PubMed] [Google Scholar]
  • 241.Anasetti C, Martin PJ, Storb R, et al. Treatment of acute graft-versus-host disease with a nonmitogenic anti-CD3 monoclonal antibody. Transplantation 1992; 54(5): 844–851. [DOI] [PubMed] [Google Scholar]
  • 242.Görena MZ, Cabadakb H. Noradrenaline and post-traumatic stress disorder. In Colin R Martin, Victor R Preedy, & Vinood B Patel, eds. Comprehensive Guide to Post-Traumatic Stress Disorder. Springer International Publishing; 2015. https://www.ncbi.nlm.nih.gov/pubmed/?term=Pietrzak+RH%2C+Gallezot+J-D%2C+Ding+Y-S%2C+Henry+S%2C+Potenza+MN%2C+Southwick+SM%2C+Krystal+JH%2C+Carson+RE%2C+Neumeister+A.+Association+of+posttraumatic+stress+disorder+with+reduced+in+vivo+norepinephrine+transporter+availability+in+the+locus+coeruleus. Accessed May 18, 2017.
  • 243.Pietrzak RH, Gallezot J-D, Ding Y-S, et al. Association of posttraumatic stress disorder with reduced in vivo norepinephrine transporter availability in the locus coeruleus. JAMA Psychiatry 2013; 70(11): 1199–1205. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 244.Levin ED, McClernon FJ, Rezvani AH. Nicotinic effects on cognitive function: behavioral characterization, pharmacological specification, and anatomic localization. Psychopharmacology 2006; 184(3–4): 523–539. [DOI] [PubMed] [Google Scholar]
  • 245.Averill LA, Purohit P, Averill CL, Boesl MA, Krystal JH, Abdallah CG. Glutamate dysregulation and glutamatergic therapeutics for PTSD: evidence from human studies. Neurosci Lett 2016; 169: 147–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Ring HA. The value of positron emission tomography in psychopharmacology. Human Psychopharmacology: Clinical and Experimental 1995; 10(2): 79–87. [Google Scholar]
  • 247.Parsey RV, Mann JJ. Applications of positron emission tomography in psychiatry. Semin Nucl Med 2003; 33(2): 129–135. [DOI] [PubMed] [Google Scholar]
  • 248.Cuthbert BN. The RDoC framework: facilitating transition from ICD/DSM to dimensional approaches that integrate neuroscience and psychopathology. World Psychiatry 2014; 13(1): 28–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Pietrzak RH, Naganawa M, Huang Y, et al. Association of in vivo κ-opioid receptor availability and the transdiagnostic dimensional expression of trauma-related psychopathology. JAMA Psychiatry 2014; 71(11): 1262–1270. [DOI] [PubMed] [Google Scholar]

Articles from Chronic Stress are provided here courtesy of SAGE Publications

RESOURCES