Skip to main content
Full access
Editorials
Published Online: 1 December 2017

BDNF, 5-HT, and Anxiety: Identification of a Critical Periadolescent Developmental Period

Anxiety disorders are expressed at peak levels during childhood and adolescence, and this early incidence is highly predictive of adult anxiety disorders (1). In addition, treatment of children or adolescents with selective serotonin reuptake inhibitors (SSRIs), the first line of pharmacological intervention for anxiety and depression, has been associated with suicidality. This has led to preclinical studies demonstrating that SSRI administration during early postnatal and preadolescent periods can cause long-lasting anxiety behaviors, even into adulthood, in animal models (2). Mechanistic studies in adult animals have demonstrated a link between anxiety, 5-HT, and brain-derived neurotrophic factor (BDNF), a major neurotrophic factor that undergoes elevated expression during the periadolescent period (35). For example, SSRI administration increases the expression of BDNF in cortical and limbic brain regions, and BDNF is required for the anxiolytic and antidepressant actions of these agents. BDNF is also a critical neurotrophic factor for the development and function of the 5-HT neurotransmitter system, and mutant rodent models with reduced BDNF signaling display increased anxiety behaviors (6, 7).
A report by Dincheva et al. in this issue (8) extends this work by examining the effect of a common human single-nucleotide polymorphism (SNP), BDNF Val66Met, on the development of the 5-HT system and anxiety behavior and the long-lasting effects of chronic SSRI treatment during a critical adolescent period.
The BDNF Val66Met polymorphism (rs6265), found in approximately 25% of Caucasians and in larger proportions in Asian populations, blocks the processing and activity-dependent release of BDNF and has been linked to reduced hippocampal volume and executive function and increased susceptibility to anxiety and depressive behaviors (912). Previous work from the Lee lab at Weill Cornell Medicine has demonstrated that mice with a knock-in of the BDNF Met allele (BDNFMet/Met) reproduce the elevated anxiety behaviors observed in human carriers (6). In addition, the ability of fluoxetine to reduce anxiety behavior is blocked in BDNFMet/Met mice, consistent with reports that the actions of SSRI antidepressants require BDNF (3, 5).
The results of the Dincheva et al. study demonstrate that early adolescent BDNFMet/Met mice (at postnatal day 30, or P30) display normal behavior, even though anxiety behavior is increased in young adults (at P60), indicating that the impact of the BDNFMet/Met polymorphism and disrupted BDNF signaling occurs during the adolescent period. Anxiety behaviors are tested in standard models, including the elevated plus maze, in which anxiety is measured as reduced time in the closed versus open arms, and novelty-induced hypophagia as latency to feed in a novel, open environment. These studies were conducted in male mice, but a previous study from this group demonstrates that female BDNFMet/Met mice show a similar delayed onset of anxiety behavior. Further study by Dincheva et al. shows that fluoxetine administration during an early adolescent phase (P21–P42) rescues the anxiety behavior in the adult BDNFMet/Met mice (Figure 1); fluoxetine administration during later adolescence (P40–P61) or in adult mice (P60–P81) had no effect on behavior. Notably, in wild-type BDNFVal/Val mice, there was no effect of fluoxetine regardless of when in the course of development it was administered. These findings indicate that normal levels of anxiety are influenced by the status of the BDNF Val/Met allele and that fluoxetine administration during a critical developmental period can normalize this behavior in adult BDNFMet/Met mice. In contrast, deficits in fear conditioning observed in BDNFMet/Met mice are not normalized by SSRI administration during any of the developmental periods.
FIGURE 1. Development of 5-HT Projections and Anxiety Behavior in BDNFMet/Met Mice: Influence of Chronic Fluoxetine Treatmenta
a The BDNF Met polymorphism decreases the processing and activity-dependent release of BDNF; the effects of this polymorphism on 5-HT and anxiety behavior were examined in the Dincheva et al. study (8). At an early periadolescent time point, postnatal day 21 (P21) levels of 5-HT fiber density in the prefrontal cortex (PFC) and hippocampus (HP) were normal, but by P42, 5-HT fiber density was retarded, indicating that BDNF plays a permissive role for the development of the 5-HT system. The reduction of 5-HT fiber density as well as S100B were long lasting, as these effects were still observed at P70 and were associated with increased anxiety behavior in adult mice. Chronic administration of fluoxetine during a critical periadolescent period (P21–P42, but not P40–P61 or P60–P81) restored 5-HT fiber growth S100B to normal levels by P70, and these effects were associated with normal behavior. These effects could occur during the adolescent period when fluoxetine was administered (P21–P42), although this is unknown, as levels of 5-HT fiber density and S100B were not determined at P42 or other early time points.
Mechanistic studies of the ameliorative effects of fluoxetine showed that the development of 5-HT fiber projections to the prefrontal cortex is decreased between P21 and P42 in BDNFMet/Met compared with wild-type mice. Surprisingly, administration of fluoxetine during the critical P21–P42 adolescent period, but not during the P60–P81 period, normalized 5-HT fiber density in the prefrontal cortex and ventral hippocampus of adult BDNFMet/Met mice (Figure 1); there was no effect of fluoxetine on fiber density at any time tested in BDNFVal/Val mice. Next, the mechanism by which SSRI treatment leads to increased 5-HT fiber density was examined. Since BDNFMet/Met mice fail to show an increase in BDNF levels with SSRI administration, the authors examined another factor, S100B, which is produced by astrocytes and is known to have neurotrophic effects on 5-HT neurons. The results demonstrate that levels of S100B are decreased in the dorsal raphe of BDNFMet/Met compared with BDNFVal/Val adult mice; notably, fluoxetine administration during the critical P21–P42 period, but not during other periods, also rescued this S100B deficit (Figure 1).
The results of this study demonstrate several important findings. First, the BDNF Met polymorphism and the consequential reduction in functional release of BDNF results in increased anxiety behavior, decreased 5-HT fiber density, and reduced S100B levels in adult mice and indicate that early adolescence is a critical period for the neurotrophic actions of BDNF. Second, administration of fluoxetine to increase 5-HT tone during this early critical periadolescent period results in a long-lasting rescue of anxiety behavior, as well as deficits in 5-HT fiber density and S100B, in the BDNFMet/Met mice. Notably, fluoxetine had no effect in wild-type mice, consistent with previous reports, although the literature is mixed. Additional studies will be required to replicate these findings and to further characterize the critical period during which fluoxetine produces beneficial long-term effects, as opposed to increased anxiety caused by earlier postnatal SSRI administration. Further studies are also needed to determine the causal relationships between anxiety, 5-HT fiber density, and S100B. For example, is S100B required for fluoxetine rescue of anxiety behavior and 5-HT fiber density in BDNFMet/Met mice? A related question is whether a decrease in S100B during the critical periadolescent period is sufficient to cause anxiety behavior and to decrease 5-HT fiber density in wild-type mice. BDNF influences many neurotransmitter, neuropeptide, and intracellular signaling systems, so there could be other neurochemical deficits that contribute to anxiety behavior and 5-HT fiber density in the BDNFMet/Met mice. It will also be interesting to determine whether fluoxetine increases 5-HT fiber density and S100B immediately after the critical periadolescent treatment period or whether this effect is delayed.
The results of the Dincheva et al. study have important clinical implications, demonstrating that it is essential to optimize the timing of pharmacological interventions to disentangle the potential beneficial versus deleterious effects of SSRI treatment during childhood and adolescent periods. Toward this goal, further studies will be required to define the critical periadolescent period for intervention in primate models. This is particularly relevant for children or adolescent patients who carry the BDNF Met allele, but it could also generalize to anxiety disorders caused by other genetic polymorphisms or environmental conditions (e.g., trauma or stress). Characterization of the discrete critical periods for the anxiogenic versus palliative effects of SSRIs and interactions with BDNF as well as other polymorphisms and environmental factors could lead to pharmacological and genetic interventions to treat anxiety as well as mood disorders in a personalized, patient-specific manner.

References

1.
Lee FS, Heimer H, Giedd JN, et al: Adolescent mental health: opportunity and obligation. Science 2014; 346:547–549
2.
Gingrich JA, Malm H, Ansorge MS, et al: New insights into how serotonin selective reuptake inhibitors shape the developing brain. Birth Defects Res 2017; 109:924–932
3.
Duman RS, Monteggia LM: A neurotrophic model for stress-related mood disorders. Biol Psychiatry 2006; 59:1116–1127
4.
Hill JL, Martinowich K: Activity-dependent signaling: influence on plasticity in circuits controlling fear-related behavior. Curr Opin Neurobiol 2016; 36:59–65
5.
Castrén E, Kojima M: Brain-derived neurotrophic factor in mood disorders and antidepressant treatments. Neurobiol Dis 2017; 97(Pt B):119–126
6.
Chen ZY, Jing D, Bath KG, et al: Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science 2006; 314:140–143
7.
Glatt CE, Lee FS: Common polymorphisms in the age of Research Domain Criteria (RDoC): integration and translation. Biol Psychiatry 2016; 79:25–31
8.
Dincheva I, Yang J, Li A, et al: Effect of early-life fluoxetine on anxiety-like behaviors in BDNF Val66Met mice. Am J Psychiatry 2017; 174:1203–1213
9.
Gatt JM, Nemeroff CB, Dobson-Stone C, et al: Interactions between BDNF Val66Met polymorphism and early life stress predict brain and arousal pathways to syndromal depression and anxiety. Mol Psychiatry 2009; 14:681–695
10.
Soliman F, Glatt CE, Bath KG, et al: A genetic variant BDNF polymorphism alters extinction learning in both mouse and human. Science 2010; 327:863–866
11.
Verhagen M, van der Meij A, van Deurzen PA, et al: Meta-analysis of the BDNF Val66Met polymorphism in major depressive disorder: effects of gender and ethnicity. Mol Psychiatry 2010; 15:260–271
12.
Wei SM, Eisenberg DP, Nabel KG, et al: Brain-derived neurotrophic factor Val66Met polymorphism affects the relationship between an anxiety-related personality trait and resting regional cerebral blood flow. Cereb Cortex 2017; 27:2175–2182

Information & Authors

Information

Published In

Go to American Journal of Psychiatry
Go to American Journal of Psychiatry
American Journal of Psychiatry
Pages: 1137 - 1139
PubMed: 29191031

History

Accepted: October 2017
Published online: 1 December 2017
Published in print: December 01, 2017

Keywords

  1. Antidepressants
  2. Anxiety Disorder-Generalized
  3. Development
  4. BDNF

Authors

Affiliations

Ronald S. Duman, Ph.D. [email protected]
From the Departments of Psychiatry and Neuroscience, Yale University School of Medicine, New Haven, Conn.

Notes

Address correspondence to Dr. Duman ([email protected]).

Funding Information

Dr. Duman has received consulting fees from Janssen and Taisho and research funding from Allergan, Janssen, Naurex, Navitor, Relmada, and Taisho. Dr. Freedman has reviewed this editorial and found no evidence of influence from these relationships.

Metrics & Citations

Metrics

Citations

Export Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format
Citation style
Style
Copy to clipboard

There are no citations for this item

View Options

View options

PDF/ePub

View PDF/ePub

Get Access

Login options

Already a subscriber? Access your subscription through your login credentials or your institution for full access to this article.

Personal login Institutional Login Open Athens login
Purchase Options

Purchase this article to access the full text.

PPV Articles - American Journal of Psychiatry

PPV Articles - American Journal of Psychiatry

Not a subscriber?

Subscribe Now / Learn More

PsychiatryOnline subscription options offer access to the DSM-5-TR® library, books, journals, CME, and patient resources. This all-in-one virtual library provides psychiatrists and mental health professionals with key resources for diagnosis, treatment, research, and professional development.

Need more help? PsychiatryOnline Customer Service may be reached by emailing [email protected] or by calling 800-368-5777 (in the U.S.) or 703-907-7322 (outside the U.S.).

Media

Figures

Other

Tables

Share

Share

Share article link

Share