ALL Metrics
-
Views
-
Downloads
Get PDF
Get XML
Cite
Export
Track
Review

Serotonin receptors in depression: from A to B

[version 1; peer review: 3 approved]
PUBLISHED 09 Feb 2017
Author details Author details
OPEN PEER REVIEW
REVIEWER STATUS

Abstract

The role of serotonin in major depressive disorder (MDD) is the focus of accumulating clinical and preclinical research. The results of these studies reflect the complexity of serotonin signaling through many receptors, in a large number of brain regions, and throughout the lifespan. The role of the serotonin transporter in MDD has been highlighted in gene by environment association studies as well as its role as a critical player in the mechanism of the most effective antidepressant treatments – selective serotonin reuptake inhibitors. While the majority of the 15 known receptors for serotonin have been implicated in depression or depressive-like behavior, the serotonin 1A (5-HT1A) and 1B (5-HT1B) receptors are among the most studied. Human brain imaging and genetic studies point to the involvement of 5-HT1A and 5-HT1B receptors in MDD and the response to antidepressant treatment. In rodents, the availability of tissue-specific and inducible knockout mouse lines has made possible the identification of the involvement of 5-HT1A and 5-HT1B receptors throughout development and in a cell-type specific manner. This, and other preclinical pharmacology work, shows that autoreceptor and heteroreceptor populations of these receptors have divergent roles in modulating depression-related behavior as well as responses to antidepressants and also have different functions during early postnatal development compared to during adulthood.

Keywords

serotonin, MDD, major depressive disorder, serotonin receptor, 5-HT1A, 5-HT1B, 5-HTT, selective serotonin reuptake inhibitors, antidepressant,

Introduction

The serotonin hypothesis of depression has dominated the field of depression for over four decades1. This theory is centered on the idea that reduced serotonin signaling is a risk factor in the etiology and/or pathophysiology of major depressive disorder (MDD)2. However, the most robust body of evidence for the role of serotonin in depression is the efficacy of increasing extracellular serotonin for the treatment of depression. The discovery that the efficacy of tricyclic antidepressants (TCAs) and monoamine oxidase inhibitor (MAOI) antidepressants was largely due to their serotonergic actions, which prompted the use of serotonin selective reuptake inhibitors (SSRIs), the first among them fluoxetine, to treat depression36. These drugs act at the serotonin transporter (5-HTT, also known as SERT) and cause increases in extracellular serotonin, which is the purported mechanism of action68. Many subsequent drugs inhibiting serotonin reuptake have shown behavioral efficacy as antidepressant drugs, suggesting that increasing synaptic serotonin levels may lead to the treatment of depression6,9.

Despite the relative success in treating depression by increasing extracellular serotonin, there is a lack of strong evidence supporting a direct correlation between low serotonin signaling and depression. While some studies report an association between levels of platelet serotonin and depression, this has not been a consistent finding in large sample sets, and it is also unclear how platelet levels are related to brain levels of serotonin10,11. Additionally, few studies report direct correlations between cerebrospinal fluid 5-hydroxyindoleacetic acid (5-HIAA), a serotonin metabolite, and depression12,13. Low levels of tryptophan have been consistently linked to depression; however, these effects could be independent of serotonin14,15. The lack of consistent clear-cut abnormalities in global measures of serotonin signaling isn’t surprising if one considers the complexity of the receptors at which serotonin binds, the intricate neuroanatomical circuitry of the serotonin system, and the developmental role serotonin plays as a neurotrophic factor1618. Many recent studies have focused on understanding the mechanisms through which serotonin affects depression by studying the impact of 5-HTT and the 15 known receptors through gene-association studies, human brain imaging, and pharmacological and genetic mouse models19.

The success in treating depression by targeting the transporter with SSRIs prompted investigations into whether variability in 5-HTT expression levels could be involved in the etiology of depression. A highly cited study showed that there is an association between a polymorphism in the serotonin transporter (5-HTTLPR) and susceptibility to developing depression20. This and other studies have shown that the short “s” allele, which results in lower levels of 5-HTT expression (at least in vitro) and therefore increased extracellular 5-HT, is associated with a higher risk of depression when combined with stressful life events21,22. This discovery would be unexpected if developmental considerations were not considered. Although inhibiting the function of the transporter during adulthood decreases depressive symptoms as in the case of SSRIs, reduced expression of 5-HTT during development may increase depressive behavior in adulthood. A human functional magnetic resonance imaging (fMRI) study supports this, showing that short allele carriers show morphological and functional alterations in limbic circuits23. Additionally, mice lacking 5-HTT throughout life display increased depressive-like behaviors, and pharmacological blockade of 5-HTT in mice exclusively during early postnatal development resulted in increased adult depressive behavior24. These results highlight the differences in developmental versus adult effects of altered serotonin neurotransmission on depression.

In addition to the serotonin transporter, the majority of the 15 serotonin receptors have been implicated in the modulation of depression, depressive-like behaviors, or the response to anti-depressant treatment19. There are numerous pre-clinical studies which have investigated the role of serotonin receptors using pharmacological manipulations and genetic knockout (KO) models in rodents (Table 1). Given the breadth of this literature, this review will focus on two receptors that are among the most extensively studied for their role in modulating depression, the 5-HT1A and 5-HT1B receptor subtypes. In addition, attention will be paid to population-dependent and development-dependent effects of serotonin signaling at these receptors and will draw from both rodent and human studies.

Table 1. Preclinical evidence supporting the role for serotonin receptors in depression.

ReceptorPubMed
Hits*
Pharmacological studies on
depression
Genetic effects on
depression
Other behavioral
phenotypes
5-HT2A588Antagonists have
antidepressant-like effects
and potentiate the effects of
SSRIs133,134
No known effect of 5-HT2A
KO on depressive-like
behavior135
Agonists are hallucinogenic;
antagonists are
antipsychotic and anxiolytic;
KO mouse has reduced
anxiety-like behavior135137
5-HT2B52Agonists have
antidepressant-like effects138
Required for behavioral
effects of SSRIs138,139
KO mouse shows increased
impulsivity140
5-HT2C282Antagonists have
antidepressant-like effects;
agonists have pro-depressive
effects141,142
No known effect of 5-HT2C
KO on depressive-like
behavior
Antagonists have anxiolytic
effects; agonists decrease
impulsivity and motivation for
drug and food consumption;
KO mouse has reduced
anxiety-like behavior143145
5-HT3A252Antagonist has
antidepressant-like effects146
5-HT3 required for
exercise-induced
antidepressant effects; KO
has antidepressant-like
phenotype147,148
Antagonists are anxiolytic149
5-HT481Agonists have rapid
antidepressant-like
effects150,151
KO has attenuated
responses to stress152
Agonists are anxiolytic;
agonists improve cognitive
performance and reduce
feeding151,153
5-HT5A5UnknownUnknownKO mice display increased
exploratory behavior154
5-HT662Agonists produce
antidepressant-like effects
and antagonists block the
effects of SSRIs155,156
UnknownAntagonists enhance
cognitive performance;
blockade of signaling is
anxiogenic157,158
5-HT7137Antagonists have
antidepressant-like effects159
KOs have an
antidepressant-like
phenotype159
Antagonists have
pro-cognitive effects160

*Number of PubMed hits based on the search terms including “depression” and the receptor as of August 25, 2016.

N.B. 5-HT1D, 1E, 1F, 3B, and 5B are not included in the chart owing to a lack of published research concerning the role of these receptors in behavior.

5-HT, serotonin; KO, knockout; SSRI, selective serotonin reuptake inhibitor.

The 5-HT1A and 5-HT1B receptors are both inhibitory Gi/o-coupled seven transmembrane receptors that are located throughout the brain2527. A major difference between these two receptors is their subcellular distribution28. 5-HT1A receptors are somatodendritic, while 5-HT1B receptors are located on axon terminals27,29. This difference is also reflected in their mechanisms of inhibitory action (Figure 1). Activation of either receptor causes decreased neurotransmitter release; however, 5-HT1A receptor activation causes hyperpolarization, leading to decreased firing, while 5-HT1B receptors inhibit voltage-gated calcium channels in the presynaptic terminal3032. Another mechanism for 5-HT1B receptor-mediated inhibition is via effects on 5-HTT, and activation of the 5-HT1B receptor increases serotonin reuptake33,34.

d15ad5ca-d168-42ee-a467-64137248cbe1_figure1.gif

Figure 1. Schematic illustrating the inhibitory effects of serotonin (5-hydroxytryptamine, 5-HT) 1A (5-HT1A) (red) and 5-HT1B (blue) receptors on the normal firing and neurotransmitter release of a neuron (top).

Activation of 5-HT1A receptors results in decreased firing (middle), while activation of 5-HT1B receptors causes decreased neurotransmitter release through actions in the presynaptic terminal (bottom).

Both 5-HT1A and 5-HT1B receptors act as autoreceptors located on serotonin neurons and also have heteroreceptor populations located on non-serotonin receptors (Figure 2). Although the mRNA in the raphe (corresponding to autoreceptors) is comparable between the two receptors, their heteroreceptors have distinct patterns of expression35. 5-HT1A receptors are enriched in the hippocampus and cortex, while 5-HT1B receptors are highly expressed in the basal ganglia36,37. These differences in mechanism of action and localization may play a role in the different functional effects of these receptors.

d15ad5ca-d168-42ee-a467-64137248cbe1_figure2.gif

Figure 2. Diagram summarizing the roles of autoreceptor and heteroreceptor populations of serotonin (5-hydroxytryptamine, 5-HT) 1A (5-HT1A) and 5-HT1B receptors on behavior during development and adulthood.

5-HTT, serotonin transporter.

While this review focuses on the contribution of 5-HT1A and 5-HT1B receptors in depression and depressive-like behaviors, these receptors also modulate other psychiatric-relevant phenotypes. For example, alterations in 5-HT1A receptor expression influence anxiety behavior, and 5-HT1B receptor signaling affects reward- and impulsivity-related phenotypes. These receptor-based differences in serotonergic regulation of emotional behavior, which segment into endophenotypes, could contribute to the heterogeneity of symptoms found in MDD38. Understanding the neural circuits that subserve these receptor-based and endophenotype-based differences can help clarify the often confusing and sometimes contradictory findings from various preclinical approaches. From a behavioral perspective, these phenotypes can be segmented through formal unsupervised factor analyses to better divide depressive behaviors into meaningful endophenotypes. Then predictors of the different endophenotypes could be tested by including genetic or pharmacological manipulations.

5-HT1A and depression

Of the 15 known serotonin receptors, the 5-HT1A receptor is the most studied for its role in depression39. Quantification of 5-HT1A receptor levels in humans from post mortem and positron emission tomography (PET) imaging studies reveals an increased level of 5-HT1A receptors in patients diagnosed with MDD4042. Gene association studies have linked a polymorphism in the 5-HT1A regulatory region (rs6295; G-1019C) with receptor levels in the brain and also to increased risk for depression4347. The GG genotype at this single nucleotide polymorphism (SNP) is associated with altered levels of 5-HT1A receptor expression and reduced responsiveness to antidepressant treatment43,48. Additionally, clinical studies have revealed antidepressant effects of buspirone and other 5-HT1A receptor agonists49,50.

Rodent models have also shown that 5-HT1A receptor agonists, such as 8-hydroxy-2-(di-n-propylamino) tetralin (8-OH-DPAT), can have acute antidepressant-like effects5153. These effects are blocked by 5-HT1A receptor antagonists, suggesting that the antidepressant-like response is specific to 5-HT1A receptor signaling54. 5-HT1A heteroreceptors, expressed throughout the limbic system, are the likely site of action for these acute 5-HT1A receptor-mediated effects50,55. On the other hand, 5-HT1A autoreceptors work in opposition to the heteroreceptors, leading to pro-depressive effects. Specifically, activation results in hyperpolarization and reduced firing of raphe neurons, leading to diminished serotonin release in projection regions56. Therefore, stimulation of 5-HT1A autoreceptors from increased extracellular serotonin following SSRI treatment is thought to oppose SSRI actions by downregulating serotonin neuron activity57. Over the first few weeks of treatment, these receptors desensitize, which may underlie the delayed behavioral efficacy of SSRIs58. Therefore, blocking 5-HT1A autoreceptor activation has been introduced as an adjunctive therapy to SSRIs. 5-HT1A receptor partial agonists such as pindolol, and more recently vilazodone, have been shown to be an effective adjunctive therapy to SSRIs in clinical studies5962. The development of new agonists that preferentially target subpopulations of 5-HT1A receptors, for example autoreceptors versus heteroreceptors, potentially through biased agonism, may be useful tools for the treatment of MDD63.

Differences in receptor levels have also been modeled in mice by using genetic loss-of-function models and have allowed causal links between receptor expression levels and depressive-like behavior. 5-HT1A receptor KO mice have an anti-depressive phenotype64,65. Tissue-specific KOs have been especially valuable for the dissection of this phenotype and have allowed investigations into the distinct roles of different populations of receptors66. The absence of heteroreceptors results in increased depressive-like behavior- as measured in the forced swim test. This mouse model also allowed for temporal control of receptor expression, which revealed a developmental sensitive period for the effect of heteroreceptors on depressive-like behavior. Specifically, knockdown of 5-HT1A heteroreceptors in adulthood was not sufficient to produce the depressive-like behavior. On the other hand, reduction of autoreceptors in adulthood increased mobility in the forced swim test, suggesting an “anti-depressed” phenotype.

Preclinical studies have also confirmed a causal role for alterations in 5-HT1A receptor expression in antidepressant efficacy. 5-HT1A receptor KO mice do not show a behavioral response to fluoxetine67. As expected from the pharmacology work, this effect is not mediated by autoreceptors, since reduced expression of 5-HT1A autoreceptors actually increases the speed and efficacy of SSRI response, requiring only 8 days of fluoxetine treatment to show a behavioral antidepressant-like response68. Recent data show that 5-HT1A heteroreceptors are critical for an effective behavioral response to an SSRI in mice69. Genetic or viral deletion of 5-HT1A receptors specifically in the dentate gyrus of the hippocampus reduced the behavioral response to fluoxetine. Furthermore, expression of 5-HT1A receptors only in the dentate gyrus was sufficient for normal antidepressant-like responses. These results importantly demonstrate a mechanism for 5-HT1A-mediated antidepressant effects localized in the mature granule cells of the dentate gyrus.

5-HT1A and other psychiatric-relevant phenotypes

Anxiety behavior is also modulated strongly by the 5-HT1A receptor, and, among depressed patients, almost half have a comorbid anxiety disorder70. In preclinical studies, 5-HT1A receptor agonists have anxiolytic effects, and 5-HT1A receptor KO mice display increased anxiety-like behavior64,65,71,72. The effect has a developmental sensitive period, since early developmental but not adult rescue of the receptor was sufficient to restore the normal phenotype in the KO73. Consistent with this, early postnatal blockade of 5-HT1A receptors, through genetic or pharmacological methods, also leads to increased anxiety74,75. Recent work has shown that the sensitive period is peri-pubertal, and tissue-specific KO mice point to a role for autoreceptors during this period of development66,76,77.

Other psychiatric disorders have also been linked to the 5-HT1A receptor, including bipolar disorder and post-traumatic stress disorder78,79. Additionally, the SNP rs6295 found in the premotor region that is associated with risk for depression is also linked with psychiatric hospitalization, a history of substance abuse, and prior suicide attempts43. Consistent with the studies in depression, the G allele is associated with reduced expression of the 5-HT1A receptor in the prefrontal cortex and an increased risk for psychiatric outcomes. Interestingly, the effects on receptor expression were also seen in the brain during early human embryonic development, suggesting its potential importance in mediating developmental contributions to adult depression. Finally, there were associations with childhood maltreatment with trends towards significant genotype by environment interactions40.

5-HT1B and depression

While the 5-HT1B receptor is best known for its role in regulating aggressive and impulsive behavior, it also plays an important role in modulating depression. Activation of the 5-HT1B receptor decreases serotonin levels in the brain through effects on release, synthesis, and reuptake33,80,81. In humans, reduced 5-HT1B receptor function is associated with MDD82. Additionally, patients with MDD are less responsive to 5-HT1B receptor agonists, suggesting reduced expression or desensitization83,84. This is consistent with clinical studies showing that 5-HT1B receptor agonists produce antidepressant effects in humans8587. This has also been shown in mice, with specific agonists resulting in antidepressant-like behavior88,89. However, genetic KO of the receptor also results in antidepressant-like behavior, suggesting that this is possibly caused by compensatory effects9094.

Both autoreceptor and heteroreceptor populations of 5-HT1B receptors have been implicated in depressive-like behaviors using rodent models. However, since 5-HT1B receptors are located on presynaptic terminals, heteroreceptors and autoreceptors have overlapping localization95. This rules out brain imaging and pharmacological manipulations in preclinical models as tools to differentiate the role of the two populations of receptors. Therefore, it has been only the recent availability of a tissue-specific genetic mouse model that has allowed the dissection of the role of 5-HT1B receptors in the regulation of behavior96.

Our recent studies show that selective ablation of 5-HT1B autoreceptors results in decreased depressive-like behaviors in mice97. These mice also show increased elevations in serotonin levels compared to controls following SSRI administration, suggesting a potential mechanism of action for the behavioral effects. Specifically, removing the terminal auto-inhibition may result in increased serotonin in projection regions that are relevant to depressive behavior. Furthermore, we also showed that the impact of 5-HT1B autoreceptors on behavior was not due to developmental expression, since the phenotype was not recapitulated in a mouse with developmental knockdown. These data are consistent with other evidence suggesting a pro-depressive role for the activation of 5-HT1B autoreceptors98,99. For example, 5-HT1B mRNA is elevated in the raphe of rats following stress and in models of depression such as learned helplessness, and viral overexpression of 5-HT1B receptors in the raphe results in depressive-like behavior following stress100. In rats, reductions in 5-HT1B receptor mRNA in the raphe are seen following SSRI treatment in post mortem brains101,102. This effect isn’t seen in other brain regions such as the cortex, hippocampus, or striatum, suggesting that this effect is specific to autoreceptors. Additionally, another study showed that 5-HT1B autoreceptors may desensitize following SSRI treatment, similar to 5-HT1A autoreceptors103. Finally, a recent PET study in humans reported that following effective cognitive behavioral therapy for depression, 5-HT1B receptor binding was reduced in the brainstem104.

There is evidence which suggests an opposing role for 5-HT1B heteroreceptors in depressive behaviors. Activation of 5-HT1B heteroreceptors in a rodent serotonin depletion model (to remove the contribution of autoreceptors) results in an antidepressant-like effect105. Additionally, reduced expression of 5-HT1B heteroreceptors in the ventral striatum is associated with depression in humans82. Finally, 5-HT1B receptors located in the ventral striatum have been suggested to interact with p11 (a 5-HT1B receptor-binding protein) to affect depression-related behaviors106,107.

5-HT1B and other psychiatric-related phenotypes

Reward dysfunction is a major symptom of MDD which is mediated, in part, by altered signaling in the mesolimbic reward system108112. 5-HT1B receptors have been implicated in the neural basis of dysregulated reward sensitivity in a number of human studies and preclinical models113116, and both 5-HT1B receptor protein and mRNA are located within the mesolimbic pathway in the nucleus accumbens (NAc) and ventral tegmental area (VTA)95. Additionally, activation of 5-HT1B receptors in the VTA increases dopamine levels in the NAc, potentially via effects on GABAergic signaling in the VTA117.

Many studies linking the receptor to functional deficits in reward processing have focused on addiction. Polymorphisms in the 5-HT1B receptor gene have also been associated with drug and alcohol abuse118120. Additionally, a PET imaging study revealed increased 5-HT1B receptor binding in pathological gamblers, who have known deficits in reward sensitivity, and gambling disorder is highly comorbid with depression and alcohol and substance use disorders116,121. Another PET imaging study shows that there is reduced 5-HT1B receptor binding in cocaine-dependent participants compared to in healthy controls122. In preclinical models, 5-HT1B receptor KO mice are more motivated to self-administer cocaine123. Consistent with this, 5-HT1B receptor agonists attenuate the motivation for cocaine but paradoxically increase the rewarding effects of cocaine124. These effects are mediated by 5-HT1B receptor expression on medium spiny neurons in the NAc, likely through their projections to the VTA125,126. Additionally, 5-HT1B receptors are required for the rewarding properties of social interaction, supporting an impact on general reward systems114.

5-HT1B receptors are also implicated in impulsive aggression. In humans, polymorphisms in the gene encoding 5-HT1B receptors have been associated with aggression, suicide, and disorders that include impulsivity as a core phenotype, including attention deficit hyperactivity disorder and substance use disorder115,120,127. In mice, 5-HT1B receptor KOs are highly aggressive in tests of male and female aggression and also display increased impulsivity128130. Additionally, 5-HT1B receptor agonists are known as “serenics” because they decrease aggression131. While the aggressive and impulsive phenotype was originally thought to be modulated by the same underlying circuits, our recent work shows that distinct populations of 5-HT1B receptors modulate aggression and impulsivity96. Furthermore, developmental expression of the 5-HT1B receptor influences aggression, while adult expression modulates impulsive behavior.

Conclusion

There is a considerable body of research that implicates serotonin in the modulation of depression and depression-related behaviors. The preclinical work delineating the effects of signaling through the 5-HT1A and 5-HT1B receptors has been made possible because of careful pharmacological studies as well as the development of transgenic mouse models that have allowed for tissue-specific and inducible knockdown. These studies have highlighted the complexity of serotonin receptors, showing that their role varies through the lifespan and by cell-type population. Additionally, the availability of specific radioligands for PET imaging of these receptors has allowed for the translation of findings from preclinical work to humans. The large number of studies concerning the role of these receptors is partially due to the fact that the 5-HT1 receptor subtypes were some of the first discovered, and it may be only a matter of time before the roles of more newly discovered receptors are clarified17.

Despite the amassing of evidence of serotonin receptor-specific involvement in depression, the primary pharmaceutical treatment strategy for depression remains the inhibition of serotonin reuptake. The lack of new treatment options is surprising given the need for them, since current SSRI treatments are ineffective in one-third of patients132. Additionally, the majority of patients, as seen in the STAR*D study, don’t respond to administration of the first SSRI treatment, requiring multi-step treatment plans that take months132. Furthermore, the considerable differences in treatment outcome also emphasize the heterogeneity of the depressed patient population. A better understanding of receptor signaling and neural circuit mechanisms by which serotonin affects depression may inform the development of novel, more targeted drugs that influence specific receptors, signaling cascades, or time periods. Also, personalized treatment plans could be developed based on symptoms, biomarkers, or pathophysiological presentation.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 09 Feb 2017
Comment
Author details Author details
Competing interests
Grant information
Copyright
Download
 
Export To
metrics
Views Downloads
F1000Research - -
PubMed Central
Data from PMC are received and updated monthly.
- -
Citations
CITE
how to cite this article
Nautiyal KM and Hen R. Serotonin receptors in depression: from A to B [version 1; peer review: 3 approved] F1000Research 2017, 6(F1000 Faculty Rev):123 (https://doi.org/10.12688/f1000research.9736.1)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
track
receive updates on this article
Track an article to receive email alerts on any updates to this article.

Open Peer Review

Current Reviewer Status: ?
Key to Reviewer Statuses VIEW
ApprovedThe paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approvedFundamental flaws in the paper seriously undermine the findings and conclusions
Version 1
VERSION 1
PUBLISHED 09 Feb 2017
Views
0
Cite
Reviewer Report 09 Feb 2017
Irwin Lucki, Department of Psychiatry, Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA 
Approved
VIEWS 0
I confirm that I have read this submission and believe that I have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Lucki I. Reviewer Report For: Serotonin receptors in depression: from A to B [version 1; peer review: 3 approved]. F1000Research 2017, 6(F1000 Faculty Rev):123 (https://doi.org/10.5256/f1000research.10497.r19617)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
Views
0
Cite
Reviewer Report 09 Feb 2017
Randy Blakely, Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA 
Approved
VIEWS 0
I confirm that I have read this submission and believe that I have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Blakely R. Reviewer Report For: Serotonin receptors in depression: from A to B [version 1; peer review: 3 approved]. F1000Research 2017, 6(F1000 Faculty Rev):123 (https://doi.org/10.5256/f1000research.10497.r19618)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
Views
0
Cite
Reviewer Report 09 Feb 2017
John Neumaier, Department of pharmacology, University of Washington, Seattle, WA, USA 
Approved
VIEWS 0
I confirm that I have read this submission and believe that I have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Neumaier J. Reviewer Report For: Serotonin receptors in depression: from A to B [version 1; peer review: 3 approved]. F1000Research 2017, 6(F1000 Faculty Rev):123 (https://doi.org/10.5256/f1000research.10497.r19619)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 09 Feb 2017
Comment
Alongside their report, reviewers assign a status to the article:
Approved - the paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations - A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approved - fundamental flaws in the paper seriously undermine the findings and conclusions
Sign In
If you've forgotten your password, please enter your email address below and we'll send you instructions on how to reset your password.

The email address should be the one you originally registered with F1000.

Email address not valid, please try again

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Google account password, please click here.

You registered with F1000 via Facebook, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Facebook account password, please click here.

Code not correct, please try again
Email us for further assistance.
Server error, please try again.