PreprintPDF Available

Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo

Authors:

Abstract and Figures

Psilocybin is a serotonergic psychedelic with untapped therapeutic potential. There are hints that the use of psychedelics can produce neural adaptations, although the extent and time scale of the impact in a mammalian brain are unknown. In this study, we used chronic two-photon microscopy to image longitudinally the apical dendritic spines of layer 5 pyramidal neurons in the mouse medial frontal cortex. We found that a single dose of psilocybin led to ~10% increases in spine size and density, driven by an elevated spine formation rate. The structural remodeling occurred quickly within 24 hours and was persistent 1 month later. Psilocybin also ameliorated stress-related behavioral deficit and elevated excitatory neurotransmission. Overall, the results demonstrate that psilocybin-evoked synaptic rewiring in the cortex is fast and enduring, potentially providing a structural trace for long-term integration of experiences and lasting beneficial actions.
Content may be subject to copyright.
Title: Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo
Authors: Ling-Xiao Shao1, Clara Liao2, Ian Gregg1, Neil K. Savalia3, Kristina Delagarza1, Alex C. Kwan1,2,3,4
1Department of Psychiatry,
2Interdepartmental Neuroscience Program,
3Medical Scientist Training Program,
4Department of Neuroscience,
Yale University School of Medicine, New Haven, Connecticut, 06511, USA
Correspondence: alex.kwan@yale.edu
Keywords: dendrites; neural plasticity; structural remodeling; serotonergic psychedelic; hallucinogen;
antidepressant
Abstract
Psilocybin is a serotonergic psychedelic with untapped therapeutic potential. Here we chronically imaged
apical dendritic spines of layer 5 pyramidal neurons in mouse medial frontal cortex. We found that a single
dose of psilocybin led to ~10% increases in spine density and spine head width. Synaptic remodeling occurred
quickly within 24 hours and was persistent 1 month later. The results demonstrate structural plasticity that may
underpin psilocybin’s long-lasting beneficial actions.
Main Text
Serotonergic psychedelics are compounds that produce an atypical state of consciousness characterized by
altered perception, cognition, and mood. It has long been recognized that these compounds may have
therapeutic potential for neuropsychiatric disorders including depression, obsessive-compulsive disorder, and
addiction1. Among psychedelics, psilocybin is recently shown to relieve depression symptoms rapidly and with
sustained benefits for several months2, 3. The therapeutic effects may arise from the ability of psilocybin to
induce neural adaptations. The expressions of genes involved in synaptic plasticity are elevated after
administration of serotonergic psychedelics in rats4, 5. In neuronal cultures, bath application of serotonergic
psychedelics induces transient increases in spine size6 and proliferation of dendritic branches7. A recent study
showed that an analogue of ibogaine, a psychedelic with differing molecular targets from psilocybin, increases
spine formation rate in mice8. Thus, there is only limited evidence linking serotonergic psychedelics to
structural plasticity in vivo. Importantly, the time scale in which synaptic remodeling may occur in the
mammalian brain is unknown.
To test the potency and dose dependence of psilocybin in mice, we measured the head-twitch response, a
classic assay for characterizing psychedelic compounds in rodents. We observed that mice would exhibit high-
frequency headshakes intermittently after administration of psilocybin. We characterized 82 C57BL/6J mice
with 5 doses of psilocybin (0, 0.25, 0.5, 1, 2 mg/kg, i.p.; range = 7-10 per sex per dose). A sharp rise of elicited
head-twitch responses occurred at 1 mg/kg (Fig. 1a, b), consistent with a prior report9. Thus, we chose to use
1 mg/kgthe inflection point of the dose-dependence curveto assess psilocybin’s effect on structural
plasticity.
In the body, psilocybin is dephosphorylated to psilocin, an agonist of 5-HT2A receptors that are densely
expressed in apical dendrites of pyramidal neurons in the medial frontal cortex of primates and rodents10, 11.
We therefore hypothesize that psilocybin may modify the dendritic architecture in the medial frontal cortex. We
used chronic two-photon microscopy to track apical dendritic spines in the cingulate/premotor (Cg1/M2) region
of Thy1GFP mice, in which a sparse subset of layer 5 pyramidal neurons express GFP12 (Fig. 1c, d). We
imaged before and after administering psilocybin (1 mg/kg, i.p.) or saline at 2-day intervals and then again 1
month later for a total of 7 imaging sessions (Fig. 1e, f). In total, we tracked 1,820 dendritic spines on 161
branches from 12 animals. Spine morphology was analyzed blind to experimental conditions using
standardized procedures13. Our results indicate that a single dose of psilocybin induces a significant elevation
in spine density (+7±2% on Day 1, +12±3% on Day 7; main effect of treatment, P=0.011, mixed-effects model
to account for variations across dendrites and mice; Fig. 1g–i) and increase in the width of spine heads
(+11±2% on Day 1, and +5±1% on Day 7; main effect of treatment, P=0.013; Fig. 1j–l, Supplementary Fig.
1). Details for all statistical tests including sample sizes are provided in Supplementary Table 1.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
Figure 1: Psilocybin increases the density and size of dendritic spines in the mouse medial frontal cortex.
(a) Head-twitch responses as a function of dose, tested on 82 C57BL/6J mice. (b) Time course of head-twitch
responses after administrating psilocybin (1 mg/kg, i.p.), averaged from 2 males and 2 female C57BL/6J mice.
Line, moving average. (c) Imaging setup. (d) Fixed coronal section from Thy1GFP mice. (e) Timeline of experiment.
(f) Example field of view. (g) Effects of psilocybin or saline treatment on spine density, plotted as fold-change from
baseline value on Day -3. Mean ± SEM. (h, i) Similar to (g), plotted separately for females and males. (j – l) Similar
to (g i) for spine head width. Sample sizes and details of the ANOVA models are provided in Supplementary
Table 1.
Increased spine density could be due to higher formation rate, lower elimination rate, or both. To distinguish
between the possibilities, we leveraged the longitudinal data set to determine the turnover rates of dendritic
spines. In females, the spine formation increased by 8±2% after psilocybin (7±1% on Day -1, 15±2% on Day 1;
Fig. 2a, b). Likewise, the spine formation rate was higher by 4±2% in males after psilocybin (6±1% on Day -1,
10±2% on Day 1). By contrast, there was no change in the elimination rate of spines (Fig. 2c). A key question
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
was whether the new spines formed after psilocybin administration would persist, because nascent dendritic
spines can take 4 days to mature into functional synapses14. For this reason, we imaged 34 days after
psilocybin administration and observed that a fraction of the psilocybin-induced new spines remained stable
(Fig. 2d). Altogether, these results demonstrate that a single dose of psilocybin induces rapid and long-lasting
dendritic remodeling in layer 5 pyramidal neurons in the mouse medial frontal cortex.
Figure 2. Psilocybin elevates the formation rate of dendritic spines. (a) Example field of view. Purple
arrowhead, stable spine. Green arrowhead, new spine. (b) Effects of psilocybin or saline treatment on the
formation rates of dendritic spines for female and male mice, plotted as difference from baseline value on Day -1.
Mean ± SEM. (c) Similar to (b) for elimination rates. (d) Fraction of spines newly formed on Day 1 that remained
stable on Day 7 and Day 34 for female and male mice. Filled circles, individual dendritic segments. Sample sizes
and details of the ANOVA models are provided in Supplementary Table 1.
To further support the conclusions, we tried to replicate the findings in another cohort of animals using a
different approach. We administered Thy1GFP mice with psilocybin (1 mg/kg, i.p.) or saline, sacrificed them 24
hours later, and imaged coronal brain sections using confocal microscopy. We expanded analyses to 6 areas
of the brain, including 2 zones that encompass apical and basal dendrites and 3 regions of the frontal cortex:
Cg1/M2, prelimbic/infralimbic (PrL/IL), and primary motor cortex (M1) (Fig. 3a–c). The results, consisting of
23,226 dendritic spines counted on 1,885 branches from 12 animals, reaffirmed the ability of psilocybin to
promote the growth of new dendritic spines in Cg1/M2 in female mice (spine density: 0.46±0.02 versus 0.50
±0.01 μm-1; Fig. 3d). Effects of psilocybin on spine density were more pronounced in female animals
(treatment x sex, P = 0.013, two-way ANOVA; Fig. 3d), echoing the trend observed in the two-photon imaging
data (Fig. 1h–i, k–l). We did not detect differences in spine protrusion length and spine head width (Fig. 3e, f),
which may be due to the across-subjects design, as it has less power than the within-subjects design of the
chronic imaging experiment. There were select morphological differences in PrL/IL and M1 (Fig. 3g–n;
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
Supplementary Fig. 2). Psilocybin also had significant impact on basal dendrites in Cg1/M2 (Fig. 3o–r).
Overall, the two sets of data converge to indicate that psilocybin promotes the rapid growth of new dendritic
spines in layer 5 pyramidal neurons in the medial frontal cortex.
Figure 3. Region-specific effects of psilocybin. (a) Stitched confocal image of a coronal brain section from a
Thy1GFP mouse. (b) Magnified images showing apical and basal dendritic segments. (c) Images of apical dendrites
in Cg1/M2. (d) Effects of psilocybin and saline on spine density for apical dendrites in Cg1/M2. Open circles,
individual dendritic segments. Gray line, mean ± SEM. (e) Similar to (d) for spine protrusion length. (f) Similar to
(d) for spine head width. (g – j) Similar to (c – f) for PrL/IL. (k – n) Similar to (c – f) for M1. (o – r) Similar to (c – f)
for basal dendrites in Cg1/M2. Sample sizes and details of the ANOVA models are provided in Supplementary
Table 1.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
The regional specificity of the psilocybin-induced structural plasticity is notable, because prior studies indicated
that the cingulate cortex is essential for head-twitch responses in rodents15. The colocalization of head-twitch
responses (which reflect hallucinogenic potency) and dendritic plasticity (which is typically involved in
therapeutic effects) in the same brain region adds to the debate over whether the hallucinogenic effects of
serotonergic psychedelics are dissociable from the therapeutic effects16, 17. By demonstrating that psilocybin
can promote structural plasticity in the mammalian brain in vivo, our study suggests that dendritic remodeling
may be a mechanism shared by compounds with rapid antidepressant effects. For instance, ketamine similarly
increases spine density by elevating spine formation rate in the medial frontal cortex18, 19. However, still
unknown is how drugs with disparate molecular targets may yield comparable circuit-level modifications20.
Elucidating the mechanisms will be crucial towards unraveling the neurobiology of rapid-acting
antidepressants.
Methods
Animals
All experiments were performed on males and females. Thy1GFP (Tg(Thy1-EGFP)MJrs/J, Stock No.007788)
transgenic mice and C57BL/6J (Stock No. 000664) mice were obtained from Jackson Laboratory. For head-
twitch response, 6 to 10-week-old C57BL/6J mice were used. For two-photon imaging, Thy1GFP mice
underwent surgery when they were 6 to 8-week-old and then were used for imaging ~2 weeks later. For
confocal imaging, 8 to 12-week-old Thy1GFP mice were used. Mice were group housed (2 5 mice per cage)
under controlled temperature in a 12hr lightdark cycle (7:00 AM to 7:00 PM) with free access to food and
water. Animal care and experimental procedures were approved by the Institutional Animal Care & Use
Committee (IACUC) at Yale University.
Head-twitch response
Head-twitch response was evaluated using 40 male and 42 female C57BL/6J mice. Upon arrival, animals
habituated at the housing facility for >2 weeks before behavioral testing. Behavioral testing took place between
10:00 AM and 4:00 PM. Animals were weighed and injected intraperitoneally with saline or psilocybin (0.25,
0.5, 1, or 2 mg/kg). We measured head-twitch response in groups of two animals: after injections, the two
animals were immediately placed into separate chambers, made by inserting a plastic divider to halve an open-
field-activity box (12” W x 6” H x 10” D). The box was within a sound attenuating cubicle with a built-in near-
infrared light source and a white light source (interior: 28” W x 34” H x 22” D, Med Associates Inc.). Videos
were recorded by a high-speed (213 fps), near-infrared camera (Genie Nano M1280, Teledyne Dalsa)
mounted overhead above the open-field-activity box. Typical recordings were 30 minutes long and, for a
subset of mice (2 males and 2 females), extended to >150 minutes. Between each measurement, the open-
field activity box was thoroughly cleaned with 70% ethanol. The videos were scored for head twitches by an
experienced observer blind to the experimental conditions.
Surgery
Prior to surgery, the mouse was injected with carprofen (5 mg/kg, s.c.; 024751, Henry Schein Animal Health,)
and dexamethasone (3 mg/kg, i.m.; 002459, Henry Schein Animal Health). During surgery, the mouse was
anesthetized with isoflurane (3 4% for induction and 1 1.5% for the remainder of surgery) and fixed in a
stereotaxic apparatus (David Kopf Instruments). The body of the mouse rested on a water-circulating heating
pad (Stryker Corp) set to 38 °C. Petrolatum ophthalmic ointment (Dechra) was used to cover the animal’s
eyes. The hair on the head was shaved, and the scalp was wiped and disinfected with ethanol pad and
betadine. An incision was made to remove the skin and the connective tissue above the skull was removed.
Subsequently, a dental drill was used to make a ~3-mm-diameter circular craniotomy above the right medial
frontal cortex (center position: +1.5 mm anterior-posterior, AP; +0.4 mm medial-lateral, ML; relative to bregma).
Artificial cerebrospinal fluid (ACSF, containing (in mM): 135 NaCl, 5 HEPES, 5 KCl, 1.8 CaCl2, 1 MgCl2; pH
7.3) was used to irrigate the exposed dura above brain. A two-layer glass window was made from two round 3-
mm-diameter, #1 thickness glass coverslip (64-0720 (CS-3R), Warner Instruments), bonded by UV-curing
optical adhesive (NOA 61, Norland Products). The glass window was carefully placed over the craniotomy and,
while maintaining a slight pressure, adhesive (Henkel Loctite 454) was used to secure the glass window to the
surrounding skull. A stainless steel headplate was affixed on the skull with C&B Metabond (Parkell) centered
on the glass window. Carprofen (5 mg/kg, s.c.) was given to the mouse immediately after surgery and on each
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
of the following 3 days. The mouse would recover for at least 10 days after the surgery before the start of
imaging experiments.
Two-photon imaging
The two-photon microscope (Movable Objective Microscope, Sutter Instrument) was controlled by ScanImage
2020 software21. The laser excitation was provided by a tunable Ti:Sapphire femtosecond laser (Chameleon
Ultra II, Coherent) and focused onto the mouse brain with a water-immersion 20X objective (XLUMPLFLN,
20x/0.95 N.A., Olympus). The laser power measured at the objective was ≤ 40 mW. To image GFP-expressing
dendrites, the laser excitation wavelength was set at 920 nm, and a 475 550 nm bandpass filter was used to
collect the fluorescence emission. During an imaging session, the mouse was head fixed and anesthetized
with 1 1.5% isoflurane. Body temperature was controlled using a heating pad and DC Temperature Controller
(40-90-8D, FHC) with rectal thermistor probe feedback. Each imaging session did not exceed 2 hours. We
imaged apical tuft dendrites at 0 200 µm below the dura. To target Cg1/M2 region, we imaged within 0 400
µm of the midline as demarcated by the sagittal sinus. Multiple fields of view were imaged in the same mouse.
For each field of view, 10 40-µm-thick image stacks were collected at 1 µm steps and at 1024 × 1024 pixels
at 0.11 µm per pixel resolution.
For longitudinal imaging, we would return to the same fields of view across imaging sessions by locating and
triangulating from a landmark on the left edge of the glass window. Each mouse was imaged on days -3, -1, 1,
3, 5 and 7 relative to the day of treatment. A subset of mice (2 males and 2 females) was imaged additionally
on day 34. On the day of treatment (day 0), there was no imaging, and the mouse was injected with either
psilocybin (1 mg/kg, i.p.) or saline (10 mL/kg, i.p.). After injection, the mouse was placed in a clean cage under
normal room lighting to visually inspect for head-twitch responses for 10 minutes, before returning the mouse
to its home cage.
Confocal imaging
Each mouse was injected with either psilocybin (1 mg/kg, i.p.) or saline (10 mL/kg, i.p.). At 24 hr after injection,
the mouse was deeply anesthetized with isoflurane and transcardially perfused with phosphate buffered saline
(PBS, P4417, Sigma-Aldrich) followed by paraformaldehyde (PFA, 4% in PBS). The brains were fixed in 4%
PFA for 24 hours at 4 °C, and then 50-µm-thick coronal brain slices were sectioned using a vibratome
(VT1000S, Leica) and placed on slides with coverslip with mounting medium. The brain slices were imaged
with a confocal microscope (LSM 880, Zeiss) equipped with a Plan-Apochromat 63x/1.40 N.A. oil objective for
dendritic spine imaging and a Plan-Apochromat 20x/0.8 N.A. objective for stitching images of an entire brain
slice.
Analysis of the imaging data
Analyses of the two-photon and confocal imaging data were mostly similar, with an additional pre-processing
step for motion correction of the two-photon imaging data using the StackReg plug-in22 in ImageJ23. Structural
parameters such as spine head width and spine protrusion length were quantified based on a standardized
protocol13, 18. Briefly, if a protrusion extended for >0.4 µm from the dendritic shaft, a dendritic spine was
counted. The head width of a dendritic spine was measured as the width at the widest part of the head of the
spine. The protrusion length of a dendritic spine referred to the distance from its root at the shaft to the tip of
the head. The line segment tool in ImageJ was used to measure the distances. Change in spine density, spine
head width and spine protrusion length across imaging sessions were shown as fold-change from the value
measured on the first imaging session (day -3) for each dendritic segment. The spine formation rate was
calculated as the number of dendritic spines newly formed between two consecutive imaging sessions divided
by the total number of dendritic spines observed in the first imaging session. The spine elimination rate was
calculated as the number of dendritic spines lost between two consecutive imaging sessions divided by the
total number of dendritic spines observed in the first imaging session. To assess the long-term dynamics of the
spine formation and elimination rates across imaging sessions, we calculated the difference from the baseline
rate, which was the spine formation or elimination rate of the same dendritic segment before psilocybin and
saline injection (i.e., from day -3 to day -1). To quantify the persistence of newly formed spines, we calculated
the number of dendritic spines newly formed on day 1 that are still present on day 7 and day 34, and divided by
the total number of newly formed dendritic spines on day 1.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
Statistics
Sample sizes and statistical analyses for each experiment are listed in Supplementary Table 1. Sample sizes
were selected based on previous experiments reported in related publications18, 24. Animals were randomly
assigned in the saline and psilocybin groups. No animals were excluded from data analysis. GraphPad Prism 8
and R were used for statistical analysis. In the figures, data are presented as the mean ± SEM per dendritic
branch.
For in vivo two-photon imaging, dendritic spine scoring was performed while blind to treatment and time.
Longitudinal measurements of dendrite structure were analyzed with mixed effects models for repeated
measures using the lme4 package in R. Linear mixed effects models were preferred to the commonly used
repeated measures analysis of variance (ANOVA) due to fewer assumptions being made about the underlying
data (e.g., balanced sampling, compound symmetry). Separate mixed effects models were created for each of
five dependent variables: fold-change in spine density, fold-change in average spine head width, fold-change
in average spine protrusion length, spine formation rate, and spine elimination rate. Each model included fixed
effects for treatment (psilocybin vs. saline), sex (female vs. male), and time (Day 1, 3, 5, and 7) as factors,
including all second and higher-order interactions between terms. Importantly, variation within mouse and
dendrite across days was accounted by including random effects terms for dendrites nested by mice. Visual
inspection of residual plots revealed no deviations from homoscedasticity or normality. P-values were
calculated by likelihood ratio tests of the full model with the effect in question against the model without the
effect in question. Post hoc t-tests were used to contrast psilocybin and saline groups per day, with and without
splitting the sample by sex, applying Bonferroni correction for multiple comparisons. Spine persistence from
two-photon imaging was analyzed with separate repeated measures ANOVAs for male and female mice, using
fixed effects of treatment (psilocybin vs. saline), time (day 7 vs. day 34), and their interaction as independent
predictors within dendrite.
For confocal imaging data, stricter blinding procedures involved one person performing imaging, another
person scrambling the image file names, and a third person performing dendritic structural measurements blind
to sex, treatment, and brain region. Data were unblinded after all of the measurements were completed. For
each brain region in the confocal dataset (Cg1/M2, PrL/IL, and M1), separate two-way ANOVAs were
constructed for apical and basal dendrites using spine density, spine head width, or spine protrusion length as
the dependent variable. Treatment (psilocybin vs. saline), sex (female vs. male), and their interaction were
included as independent predictors. Post hoc t-tests were used to contrast psilocybin and saline groups within
sex, applying Bonferroni correction for multiple comparisons.
Data availability
The data that support the findings of this study will be made publicly available at https://github.com/Kwan-Lab.
Code availability
The code used to analyze the data in this study will be made publicly available at https://github.com/Kwan-Lab.
Acknowledgements
We thank B. Kelmendi and C. Pittenger for help on obtaining psilocybin, Usona Institute for providing
psilocybin, J. Taylor for use of open-field activity boxes, H. Atilgan and H. Ortega for assistance on setting up
video recording, and A. Halberstadt for advice on scoring head-twitch responses. This work was supported by
the Yale Center for Psychedelic Science, NIH/NINDS training grant T32NS041228 (C.L.), and NIH/NIGMS
Medical Scientist Training grant T32GM007205 (N.K.S.). We thank the Yale Center for Advanced Light
Microscopy Facility for their assistance with confocal imaging, supported in part via NIH grant S10OD023598.
Author contributions
L.X.S. and A.C.K. designed the research. L.X.S. performed the two-photon and confocal imaging experiments,
and analyzed the two-photon imaging data. N.K.S. blinded and I.G. analyzed the confocal imaging data. C.L.
performed and analyzed the behavioral experiments. I.G. and K.D. assisted with analyzing the behavioral data.
N.K.S. assisted with the statistical analyses. L.X.S. and A.C.K. wrote the paper, with input from all other
authors.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
Competing interests
A.C.K. received psilocybin from the investigational drug supply program at Usona Institute. The authors
declare no other competing interests.
References
1. Vollenwe ider, F.X . & Preller, K.H. Psychedelic drugs : neurobiology an d pote ntial for treatment of
psychiatric disorders. Nat Rev Neurosci 21, 611-624 (2020).
2. Davis, A.K., et al. Effects of Psilocybin-Assisted Therapy on Major Depressive Disorder: A Randomized
Clinical Trial. JAMA Psychiatry (2020).
3. Carhart-Harris, R.L., et al. Psilocybin with psychological support for treatment-resistant depression: an
open-label feasibility study. The Lancet Psychiatry 3, 619-627 (2016).
4. Nichols, C.D. & Sanders-Bush, E. A single dose of lysergic acid diethylamide influences gene
expression patterns within the mammalian brain. Neuropsychopharmacology 26, 634-642 (2002).
5. Vaidya, V.A. , Marek, G.J., Aghajanian, G.K. & Duman, R.S. 5-HT2A receptor-mediated regulation of
brain-derived neurotrophic factor mRNA in the hippocampus and the neocortex. J Neurosci 17, 2785-2795
(1997).
6. Jones, K.A., et al. Rapid modulation of spine morphology by the 5-HT2A serotonin receptor through
kalirin-7 signaling. Proc Natl Acad Sci U S A 106, 19575-19580 (2009).
7. Ly, C., et al. Psychedelics Promote Structural and Functional Neural Plasticity. Cell Rep 23, 3170-3182
(2018).
8. Cameron, L.P., et al. A non-hallucinogenic psychedelic analogue with therapeutic potential. Nature
(2020).
9. Halberstadt, A.L., Koedood, L., Powell, S.B. & Geyer, M.A. Differential contributions of serotonin
receptors to the behavioral effects of indoleamine hallucinogens in mice. J Psychopharmacol 25, 1548-1561
(2011).
10. Willins, D.L., Deutch, A.Y. & Roth, B.L. Serotonin 5-HT2A receptors are expressed on pyramidal cells
and interneurons in the rat cortex. Synapse 27, 79-82 (1997).
11. Jakab, R.L. & Goldman-Rakic, P.S. 5-Hydroxytryptamine2A serotonin receptors in the primate cerebral
cortex: possible site of action of hallucinogenic and antipsychotic drugs in pyramidal cell apical dendrites. Proc
Natl Acad Sci U S A 95, 735-740 (1998).
12. Feng, G., et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of
GFP. Neuron 28, 41-51 (2000).
13. Holtmaat, A., et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial
window. Nat Protoc 4, 1128-1144 (2 0 0 9).
14. Knott, G.W., Holtmaat, A., Wilbrecht, L., Welker, E. & Svoboda, K. Spine growth precedes synapse
formation in the adult neocortex in vivo. Nat Neurosci 9, 1117-112 4 (2006).
15. Canal, C.E. & Morgan, D. Head-twitch response in rodents induced by the hallucinogen 2,5-dimethoxy-
4-iodoamphetamine: a comprehensive history, a re-evaluation of mechanisms, and its utility as a model. Drug
Test An al 4, 556-576 (2012).
16. Olson, D.E. The Subjective Effects of Psychedelics May Not Be Necessary for Their Enduring
Therapeutic Effects. ACS Pharmacology & Translational Science (2020).
17. Yaden, D. B . & Griffit h s , R.R. The S u bjecti v e Effects of P sychede l i cs Ar e N ecessar y for Their E n during
Therapeutic Effects. ACS Pharmacology & Translational Science (2020).
18. Phoumthipphavong, V., Barthas, F., Hassett, S. & Kwan, A.C. Longitudinal Effects of Ketamine on
Dendritic Architecture In Vivo in the Mouse Medial Frontal Cortex. eNeuro 3, ENEURO.0133-0115.2016
(2016).
19. Moda-Sava, R.N., et al. Sustained rescue of prefrontal circuit dysfunction by antidepressant-induced
spine formation. Science 364 (2019).
20. Savalia, N.K., Shao, L.-X. & Kwan, A.C. A Dendrite-Focused Framework for Understanding the Actions
of Ketamine and Psychedelics. Trends i n Neurosci e n ces, in press (2020).
21. Pologruto, T.A., Sabatini, B.L. & Svoboda, K. ScanImage: flexible software for operating laser scanning
microscopes. Biomed Eng Online 2, 13 (2003).
22. Thevenaz, P., Ruttimann, U.E. & Unser, M. A pyramid approach to subpixel registration based on
intensity. IEEE Trans Image Process 7, 27-41 (1998).
23. Schneider, C.A., Rasband, W.S. & Eliceiri, K.W. NIH Image to ImageJ: 25 years of image analysis.
Nature Methods 9, 671-675 (2012).
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
24. Grutzendler, J., Kasthuri, N. & Gan, W.B. Long-term dendritic spine stability in the adult cortex. Nature
420, 812-816 (2002).
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
Supplementary Figures
Supplementary Figure 1: Psilocybin increases spine protrusion length in Cg1/M2. (a) Effects of psilocybin or
saline treatment on spine density in Thy1GFP mice, plotted as fold-change from baseline value on Day -3. (b, c)
Similar to (a), plotted separately for females and males. (j l) Similar to (g i) for spine head width. Mean ± SEM.
Sample sizes and statistical analyses are provided in Supplementary Table 1.
Supplementary Figure 2. Effects of psilocybin on basal dendrites in PrL/IL and M1. (a) Images of basal
dendritic segments in PrL/IL from coronal brain sections from a Thy1GFP mouse. (b) Effects of psilocybin and saline
on spine density for basal dendrites in PrL/IL. Open circles, individual dendritic segments. (c) Similar to (b) for
spine protrusion length. (d) Similar to (b) for spine head width. (e – h) Similar to (a – d) for M1. Mean, SEM.
Sample sizes and details of the ANOVA models are provided in Supplementary Table 1.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 17, 2021. ; https://doi.org/10.1101/2021.02.17.431629doi: bioRxiv preprint
... Ketamine increases BDNF protein translation (135,145) and its antidepressant effects are absent when administered to inducible BDNF knockout mice (135) or homozygous mice harboring the BDNF Val66Met mutation (136). Ketamine and psychedelics modulate cortical neuron function by increasing dendritic spine and synapse density in the PFC (129,(146)(147)(148)(149); however, ketamine's effects on structural plasticity appear to last for approximately a week (150) while psilocybin's effects seem to be more durable lasting for at least a month (151,152). Though the primary molecular targets of ketamine and serotonergic psychedelics are distinct, their downstream pharmacology overlaps, requiring AMPA receptor, TrkB, and mTOR activation to elicit changes in neuronal structure and function (135-137, 148, 153, 154). Moreover, their effects seem to be C max driven, as very short stimulation periods (15 min−1 h) are sufficient to induce sustained changes in cortical neuron structure (153). ...
... This correlates well with the effects of the drug on dendritic spine density (129,150). In contrast, the mood-elevating properties of psilocybin seem to last significantly longer (40)(41)(42)151), as do its effects on neuronal structure (152). Currently, it is unclear exactly how long the effects of non-hallucinogenic psychoplastogens will last following a single administration, and head-to-head comparisons with ketamine and psilocybin are warranted to help establish optimal dosing frequency. ...
Article
Full-text available
Psychedelics have inspired new hope for treating brain disorders, as they seem to be unlike any treatments currently available. Not only do they produce sustained therapeutic effects following a single administration, they also appear to have broad therapeutic potential, demonstrating efficacy for treating depression, post-traumatic stress disorder (PTSD), anxiety disorders, substance abuse disorder, and alcohol use disorder, among others. Psychedelics belong to a more general class of compounds known as psychoplastogens, which robustly promote structural and functional neural plasticity in key circuits relevant to brain health. Here we discuss the importance of structural plasticity in the treatment of neuropsychiatric diseases, as well as the evidence demonstrating that psychedelics are among the most effective chemical modulators of neural plasticity studied to date. Furthermore, we provide a theoretical framework with the potential to explain why psychedelic compounds produce long-lasting therapeutic effects across a wide range of brain disorders. Despite their promise as broadly efficacious neurotherapeutics, there are several issues associated with psychedelic-based medicines that drastically limit their clinical scalability. We discuss these challenges and how they might be overcome through the development of non-hallucinogenic psychoplastogens. The clinical use of psychedelics and other psychoplastogenic compounds marks a paradigm shift in neuropsychiatry toward therapeutic approaches relying on the selective modulation of neural circuits with small molecule drugs. Psychoplastogen research brings us one step closer to actually curing mental illness by rectifying the underlying pathophysiology of disorders like depression, moving beyond simply treating disease symptoms. However, determining how to most effectively deploy psychoplastogenic medicines at scale will be an important consideration as the field moves forward.
... The same treatment evoked chromatin changes that persisted long after the DOI treatment, suggesting that therapeutic responses to hallucinogen treatment may arise through persistent and epigenetic changes in synaptic architecture. Others have recently used chronic two-photon microscopy to monitor synaptic spine density in mouse cortex [110]. In female mice, a single dose of psilocybin (1 mg/kg, i.p.) evoked a 20% increase spine density in layer V of the frontal cortex that persisted for up to 34 days, along with a sharp increase in spine dimension that peaked at day one and declined over the following week; these changes were scarcely evident in male mice. ...
Article
Full-text available
Hallucinogens are a loosely defined group of compounds including LSD, N,N-dimethyltryptamines, mescaline, psilocybin/psilocin, and 2,5-dimethoxy-4-methamphetamine (DOM), which can evoke intense visual and emotional experiences. We are witnessing a renaissance of research interest in hallucinogens, driven by increasing awareness of their psychotherapeutic potential. As such, we now present a narrative review of the literature on hallucinogen binding in vitro and ex vivo, and the various molecular imaging studies with positron emission tomography (PET) or single photon emission computer tomography (SPECT). In general, molecular imaging can depict the uptake and binding distribution of labelled hallucinogenic compounds or their congeners in the brain, as was shown in an early PET study with N1-([11C]-methyl)-2-bromo-LSD ([11C]-MBL); displacement with the non-radioactive competitor ketanserin confirmed that the majority of [11C]-MBL specific binding was to serotonin 5-HT2A receptors. However, interactions at serotonin 5HT1A and other classes of receptors and pleotropic effects on second messenger pathways may contribute to the particular experiential phenomenologies of LSD and other hallucinogenic compounds. Other salient aspects of hallucinogen action include permeability to the blood–brain barrier, the rates of metabolism and elimination, and the formation of active metabolites. Despite the maturation of radiochemistry and molecular imaging in recent years, there has been only a handful of PET or SPECT studies of radiolabeled hallucinogens, most recently using the 5-HT2A/2C agonist N-(2[11CH3O]-methoxybenzyl)-2,5-dimethoxy- 4-bromophenethylamine ([11C]Cimbi-36). In addition to PET studies of target engagement at neuroreceptors and transporters, there is a small number of studies on the effects of hallucinogenic compounds on cerebral perfusion ([15O]-water) or metabolism ([18F]-fluorodeoxyglucose/FDG). There remains considerable scope for basic imaging research on the sites of interaction of hallucinogens and their cerebrometabolic effects; we expect that hybrid imaging with PET in conjunction with functional magnetic resonance imaging (fMRI) should provide especially useful for the next phase of this research.
Article
Full-text available
Psilocybin is a naturally occurring psychedelic compound with profound perception-, emotion- and cognition-altering properties and great potential for treating brain disorders. However, the neural mechanisms mediating its effects require in-depth investigation as there is still much to learn about how psychedelic drugs produce their profound and long-lasting effects. In this review, we outline the current understanding of the neurophysiology of psilocybin's psychoactive properties, highlighting the need for additional preclinical studies to determine its effect on neural network dynamics. We first describe how psilocybin's effect on brain regions associated with the default-mode network (DMN), particularly the prefrontal cortex and hippocampus, likely plays a key role in mediating its consciousness-altering properties. We then outline the specific receptor and cell types involved and discuss contradictory evidence from neuroimaging studies regarding psilocybin's net effect on activity within these regions. We go on to argue that in vivo electrophysiology is ideally suited to provide a more holistic, neural network analysis approach to understand psilocybin's mode of action. Thus, we integrate information about the neural bases for oscillatory activity generation with the accumulating evidence about psychedelic drug effects on neural synchrony within DMN-associated areas. This approach will help to generate important questions for future preclinical and clinical studies. Answers to these questions are vital for determining the neural mechanisms mediating psilocybin's psychotherapeutic potential, which promises to improve outcomes for patients with severe depression and other difficulty to treat conditions.
Article
Full-text available
The high symptomatic and biological heterogeneity of major depressive disorder (MDD) makes it very difficult to find broadly efficacious treatments that work against all symptoms. Concentrating on single core symptoms that are biologically well understood might consist of a more viable approach. The Research Domain Criteria (RDoC) framework is a trans-diagnostic dimensional approach that focuses on symptoms and their underlying neurobiology. Evidence is accumulating that psychedelics may possess antidepressant activity, and this can potentially be explained through a multi-level (psychobiological, circuitry, (sub)cellular and molecular) analysis of the cognitive systems RDoC domain. Cognitive deficits, such as negative emotional processing and negativity bias, often lead to depressive rumination. Psychedelics can increase long-term cognitive flexibility, leading to normalization of negativity bias and reduction in rumination. We propose a theoretical model that explains how psychedelics can reduce the negativity bias in depressed patients. At the psychobiological level, we hypothesize that the negativity bias in MDD is due to impaired pattern separation and that psychedelics such as psilocybin help in depression because they enhance pattern separation and hence reduce negativity bias. Pattern separation is a mnemonic process that relies on adult hippocampal neurogenesis, where similar inputs are made more distinct, which is essential for optimal encoding of contextual information. Impairment in this process may underlie the negative cognitive bias in MDD by, for example, increased pattern separation of cues with a negative valence that can lead to excessive deliberation on aversive outcomes. On the (sub) cellular level, psychedelics stimulate hippocampal neurogenesis as well as synaptogenesis, spinogenesis and dendritogenesis in the prefrontal cortex. Together, these effects help restoring resilience to chronic stress and lead to modulation of the major connectivity hubs of the prefrontal cortex, hippocampus, and amygdala. Based on these observations, we propose a new translational framework to guide the development of a novel generation of therapeutics to treat the cognitive symptoms in MDD.
Article
Full-text available
Classic psychedelics produce altered states of consciousness that individuals often interpret as meaningful experiences. Across a number of human studies, when the participant-rated intensity of the overall drug effects are statistically controlled for, certain subjective effects predict therapeutic and other desirable outcomes. Underlying neurobiological mechanisms are likely necessary but not sufficient to confer full and enduring beneficial effects. We propose that the subjective effects of psychedelics are necessary for their enduring beneficial effects and that these subjective effects account for the majority of their benefit.
Article
Full-text available
The psychedelic alkaloid ibogaine has anti-addictive properties in both humans and animals¹. Unlike most medications for the treatment of substance use disorders, anecdotal reports suggest that ibogaine has the potential to treat addiction to various substances, including opiates, alcohol and psychostimulants. The effects of ibogaine—like those of other psychedelic compounds—are long-lasting², which has been attributed to its ability to modify addiction-related neural circuitry through the activation of neurotrophic factor signalling3,4. However, several safety concerns have hindered the clinical development of ibogaine, including its toxicity, hallucinogenic potential and tendency to induce cardiac arrhythmias. Here we apply the principles of function-oriented synthesis to identify the key structural elements of the potential therapeutic pharmacophore of ibogaine, and we use this information to engineer tabernanthalog—a water-soluble, non-hallucinogenic, non-toxic analogue of ibogaine that can be prepared in a single step. In rodents, tabernanthalog was found to promote structural neural plasticity, reduce alcohol- and heroin-seeking behaviour, and produce antidepressant-like effects. This work demonstrates that, through careful chemical design, it is possible to modify a psychedelic compound to produce a safer, non-hallucinogenic variant that has therapeutic potential.
Article
Full-text available
Importance: Major depressive disorder (MDD) is a substantial public health burden, but current treatments have limited effectiveness and adherence. Recent evidence suggests that 1 or 2 administrations of psilocybin with psychological support produces antidepressant effects in patients with cancer and in those with treatment-resistant depression. Objective: To investigate the effect of psilocybin therapy in patients with MDD. Design, Setting, and Participants: This randomized, waiting list–controlled clinical trial was conducted at the Center for Psychedelic and Consciousness Research at Johns Hopkins Bayview Medical Center in Baltimore, Maryland. Adults aged 21 to 75 years with an MDD diagnosis, not currently using antidepressant medications, and without histories of psychotic disorder, serious suicide attempt, or hospitalization were eligible to participate. Enrollment occurred between August 2017 and April 2019, and the 4-week primary outcome assessments were completed in July 2019. A total of 27 participants were randomized to an immediate treatment condition group (n = 15) or delayed treatment condition group (waiting list control condition; n = 12). Data analysis was conducted from July 1, 2019, to July 31, 2020, and included participants who completed the intervention (evaluable population). Interventions: Two psilocybin sessions (session 1: 20 mg/70 kg; session 2: 30 mg/70 kg) were given (administered in opaque gelatin capsules with approximately 100 mL of water) in the context of supportive psychotherapy (approximately 11 hours). Participants were randomized to begin treatment immediately or after an 8-week delay. Main Outcomes and Measures: The primary outcome, depression severity was assessed with the GRID-Hamilton Depression Rating Scale (GRID-HAMD) scores at baseline (score of ≥17 required for enrollment) and weeks 5 and 8 after enrollment for the delayed treatment group, which corresponded to weeks 1 and 4 after the intervention for the immediate treatment group. Secondary outcomes included the Quick Inventory of Depressive Symptomatology-Self Rated (QIDS-SR). Results: Of the randomized participants, 24 of 27 (89%) completed the intervention and the week 1 and week 4 postsession assessments. This population had a mean (SD) age of 39.8 (12.2) years, was composed of 16 women (67%), and had a mean (SD) baseline GRID-HAMD score of 22.8 (3.9). The mean (SD) GRID-HAMD scores at weeks 1 and 4 (8.0 [7.1] and 8.5 [5.7]) in the immediate treatment group were statistically significantly lower than the scores at the comparable time points of weeks 5 and 8 (23.8 [5.4] and 23.5 [6.0]) in the delayed treatment group. The effect sizes were large at week 5 (Cohen d = 2.2; 95% CI, 1.4-3.0; P < .001) and week 8 (Cohen d = 2.6; 95% CI, 1.7-3.6; P < .001). The QIDS-SR documented a rapid decrease in mean (SD) depression score from baseline to day 1 after session 1 (16.7 [3.5] vs 6.3 [4.4]; Cohen d = 3.0; 95% CI, 1.9-4.0; P < .001), which remained statistically significantly reduced through the week 4 follow-up (6.0 [5.7]; Cohen d = 3.1; 95% CI, 1.9-4.2; P < .001). In the overall sample, 16 participants (67%) at week 1 and 17 (71%) at week 4 had a clinically significant response to the intervention (≥50% reduction in GRID-HAMD score), and 14 participants (58%) at week 1 and 13 participants (54%) at week 4 were in remission (≤7 GRID-HAMD score). Conclusions and Relevance: Findings suggest that psilocybin with therapy is efficacious in treating MDD, thus extending the results of previous studies of this intervention in patients with cancer and depression and of a nonrandomized study in patients with treatment-resistant depression. Trial Registration: ClinicalTrials.gov Identifier: NCT03181529
Article
Full-text available
Renewed interest in the use of psychedelics in the treatment of psychiatric disorders warrants a better understanding of the neurobiological mechanisms underlying the effects of these substances. After a hiatus of about 50 years, state-of-the art studies have recently begun to close important knowledge gaps by elucidating the mechanisms of action of psychedelics with regard to their effects on receptor subsystems, systems-level brain activity and connectivity, and cognitive and emotional processing. In addition, functional studies have shown that changes in self-experience, emotional processing and social cognition may contribute to the potential therapeutic effects of psychedelics. These discoveries provide a scientific road map for the investigation and application of psychedelic substances in psychiatry.
Article
Full-text available
Atrophy of neurons in the prefrontal cortex (PFC) plays a key role in the pathophysiology of depression and related disorders. The ability to promote both structural and functional plasticity in the PFC has been hypothesized to underlie the fast-acting antidepressant properties of the dissociative anesthetic ketamine. Here, we report that, like ketamine, serotonergic psychedelics are capable of robustly increasing neuritogenesis and/or spinogenesis both in vitro and in vivo. These changes in neuronal structure are accompanied by increased synapse number and function, as measured by fluorescence microscopy and electrophysiology. The structural changes induced by psychedelics appear to result from stimulation of the TrkB, mTOR, and 5-HT2A signaling pathways and could possibly explain the clinical effectiveness of these compounds. Our results underscore the therapeutic potential of psychedelics and, importantly, identify several lead scaffolds for medicinal chemistry efforts focused on developing plasticity-promoting compounds as safe, effective, and fast-acting treatments for depression and related disorders.
Article
Full-text available
Background: Psilocybin is a serotonin receptor agonist that occurs naturally in some mushroom species. Recent studies have assessed the therapeutic potential of psilocybin for various conditions, including end-of-life anxiety, obsessive-compulsive disorder, and smoking and alcohol dependence, with promising preliminary results. Here, we aimed to investigate the feasibility, safety, and efficacy of psilocybin in patients with unipolar treatment-resistant depression. Methods: In this open-label feasibility trial, 12 patients (six men, six women) with moderate-to-severe, unipolar, treatment-resistant major depression received two oral doses of psilocybin (10 mg and 25 mg, 7 days apart) in a supportive setting. There was no control group. Psychological support was provided before, during, and after each session. The primary outcome measure for feasibility was patient-reported intensity of psilocybin's effects. Patients were monitored for adverse reactions during the dosing sessions and subsequent clinic and remote follow-up. Depressive symptoms were assessed with standard assessments from 1 week to 3 months after treatment, with the 16-item Quick Inventory of Depressive Symptoms (QIDS) serving as the primary efficacy outcome. This trial is registered with ISRCTN, number ISRCTN14426797. Findings: Psilocybin's acute psychedelic effects typically became detectable 30-60 min after dosing, peaked 2-3 h after dosing, and subsided to negligible levels at least 6 h after dosing. Mean self-rated intensity (on a 0-1 scale) was 0·51 (SD 0·36) for the low-dose session and 0·75 (SD 0·27) for the high-dose session. Psilocybin was well tolerated by all of the patients, and no serious or unexpected adverse events occurred. The adverse reactions we noted were transient anxiety during drug onset (all patients), transient confusion or thought disorder (nine patients), mild and transient nausea (four patients), and transient headache (four patients). Relative to baseline, depressive symptoms were markedly reduced 1 week (mean QIDS difference -11·8, 95% CI -9·15 to -14·35, p=0·002, Hedges' g=3·1) and 3 months (-9·2, 95% CI -5·69 to -12·71, p=0·003, Hedges' g=2) after high-dose treatment. Marked and sustained improvements in anxiety and anhedonia were also noted. Interpretation: This study provides preliminary support for the safety and efficacy of psilocybin for treatment-resistant depression and motivates further trials, with more rigorous designs, to better examine the therapeutic potential of this approach. Funding: Medical Research Council.
Article
Full-text available
A single subanesthetic dose of ketamine, an NMDA receptor antagonist, leads to fast-acting antidepressant effects. In rodent models, systemic ketamine is associated with higher dendritic spine density in the prefrontal cortex, reflecting structural remodeling that may underlie the behavioral changes. However, turnover of dendritic spines is a dynamic process in vivo, and the longitudinal effects of ketamine on structural plasticity remain unclear. The purpose of the current study is to use subcellular resolution optical imaging to determine the time course of dendritic alterations in vivo following systemic ketamine administration in mice. We used two-photon microscopy to visualize repeatedly the same set of dendritic branches in the mouse medial frontal cortex (MFC) before and after a single injection of ketamine or saline. Compared to controls, ketamine-injected mice had higher dendritic spine density in MFC for up to 2 weeks. This prolonged increase in spine density was driven by an elevated spine formation rate, and not by changes in the spine elimination rate. A fraction of the new spines following ketamine injection was persistent, which is indicative of functional synapses. In a few cases, we also observed retraction of distal apical tuft branches on the day immediately after ketamine administration. These results indicate that following systemic ketamine administration, certain dendritic inputs in MFC are removed immediately, while others are added gradually. These dynamic structural modifications are consistent with a model of ketamine action in which the net effect is a rebalancing of synaptic inputs received by frontal cortical neurons.
Article
Pilot studies have hinted that serotonergic psychedelics such as psilocybin may relieve depression, and could possibly do so by promoting neural plasticity. Intriguingly, another psychotomimetic compound, ketamine, is a fast-acting antidepressant and induces synapse formation. The similarities in behavioral and neural effects have been puzzling because the compounds target distinct molecular receptors in the brain. In this opinion article, we develop a conceptual framework that suggests the actions of ketamine and serotonergic psychedelics may converge at the dendrites, to both enhance and suppress membrane excitability. We speculate that mismatches in the opposing actions on dendritic excitability may relate to these compounds’ cell-type and region selectivity, their moderate range of effects and toxicity, and their plasticity-promoting capacities.
Article
Psychedelics represent one of the most promising classes of experimental medicines for the treatment of neuropsychiatric disorders due to their ability to promote neural plasticity and produce both rapid and sustained therapeutic effects following a single administration. Conventional wisdom holds that peak mystical experiences induced by psychedelics are a critical component of their therapeutic mechanisms of action, though evidence supporting that claim is largely correlational. Here, I present data suggesting that the subjective effects induced by psychedelics may not be necessary to produce long-lasting changes in mood and behavior. Understanding the role of subjective effects in the therapeutic mechanisms of psychedelics will have important implications for both basic neuroscience and for increasing patient access to the next generation of medicines developed as a result of psychedelic research.
Article
Why is ketamine an antidepressant? A better understanding of the mechanisms underlying the action of antidepressants is urgently needed. Moda-Sava et al. explored a possible mode of action for the drug ketamine, which has recently been shown to help patients recover from depression (see the Perspective by Beyeler). Ketamine rescued behavior in mice that was associated with depression-like phenotypes by selectively reversing stress-induced spine loss and restoring coordinated multicellular ensemble activity in prefrontal microcircuits. The initial induction of ketamine's antidepressant effect on mouse behavior occurred independently of effects on spine formation. Instead, synaptogenesis in the prefrontal region played a critical role in nourishing these effects over time. Interventions aimed at enhancing the survival of restored synapses may thus be useful for sustaining the behavioral effects of fast-acting antidepressants. Science , this issue p. eaat8078 ; see also p. 129