ArticlePDF AvailableLiterature Review

Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of the role of puberty

Authors:

Abstract

Suicide is the second leading cause of death among adolescents. While clinicians and researchers have begun to recognize the importance of considering multidimensional factors in understanding risk for suicidal thoughts and behaviors (STBs) during this developmental period, the role of puberty has been largely ignored. In this review, we contend that the hormonal events that occur during puberty have significant effects on the organization and development of brain systems implicated in the regulation of social stressors, including amygdala, hippocampus, striatum, medial prefrontal cortex, orbitofrontal cortex, and anterior cingulate cortex. Guided by previous experimental work in adults, we also propose that the influence of pubertal hormones and social stressors on neural systems related to risk for STBs is especially critical to consider in adolescents with a neurobiological sensitivity to hormonal changes. Furthermore, facets of the pubertal transition, such as pubertal timing, warrant deeper investigation and may help us gain a more comprehensive understanding of sex differences in the neurobiological and psychosocial mechanisms underlying adolescent STBs. Ultimately, advancing our understanding of the pubertal processes that contribute to suicide risk will improve early detection and facilitate the development of more effective, sex-specific, psychiatric interventions for adolescents.
Molecular Psychiatry
https://doi.org/10.1038/s41380-021-01171-5
EXPERT REVIEW
Psychobiological risk factors for suicidal thoughts and behaviors
in adolescence: a consideration of the role of puberty
Tiffany C. Ho1,2 Anthony J. Gifuni1,3 Ian H. Gotlib1
Received: 17 April 2020 / Revised: 3 May 2021 / Accepted: 11 May 2021
© The Author(s) 2021. This article is published with open access
Abstract
Suicide is the second leading cause of death among adolescents. While clinicians and researchers have begun to recognize
the importance of considering multidimensional factors in understanding risk for suicidal thoughts and behaviors (STBs)
during this developmental period, the role of puberty has been largely ignored. In this review, we contend that the hormonal
events that occur during puberty have signicant effects on the organization and development of brain systems implicated in
the regulation of social stressors, including amygdala, hippocampus, striatum, medial prefrontal cortex, orbitofrontal cortex,
and anterior cingulate cortex. Guided by previous experimental work in adults, we also propose that the inuence of pubertal
hormones and social stressors on neural systems related to risk for STBs is especially critical to consider in adolescents with
a neurobiological sensitivity to hormonal changes. Furthermore, facets of the pubertal transition, such as pubertal timing,
warrant deeper investigation and may help us gain a more comprehensive understanding of sex differences in the
neurobiological and psychosocial mechanisms underlying adolescent STBs. Ultimately, advancing our understanding of the
pubertal processes that contribute to suicide risk will improve early detection and facilitate the development of more
effective, sex-specic, psychiatric interventions for adolescents.
Introduction
Suicide is the second leading cause of death among ado-
lescents ages 1024 years worldwide [1] and accounts for
17.7% of all deaths in youth ages 1524 years in the United
States [2]. Although suicide has a relatively low prevalence,
over the past 10 years suicide rates in the US have increased
by 76% in youth ages 1519 years and, more alarmingly, by
300% in youth ages 1014 years [3]. Despite the urgency of
suicide as a public health problem, research examining
predictors of suicidal thoughts and behaviors (STBs) has
had limited clinical impact, particularly in adolescents
[4,5]. Adolescents report a higher proportion of suicidal
ideation and attempts than do adults, making this a critical
developmental period during which to identify early risk
factors for STBs [6]. Indeed, the fact that adolescent sam-
ples represent only 20% of the literature on predictors of
STBs further underscores the need for more research with
this age group [5].
While clinicians and researchers recognize that predictors
of STBs are multidimensionalincluding genetic, epigenetic,
neurobiological, psychosocial, and environmental factors
the marked increase in the prevalence of STBs (and mental
health conditions more generally) during adolescence [7]
suggests that this developmental period, in which the con-
uence of these multidimensional inuences contributes to
elevated risk, is unique [8,9]. In this context, we contend that
one critical factor is puberty. The onset of puberty initiates a
neuroendocrine cascade that shapes the maturation of neural
circuits that underlie a range of socioemotional and cognitive
functioning, including emotion regulation and impulse con-
trol. Impaired emotion regulation and impulse control both
signicantly contribute to the emergence of mental disorders
in the face of environmental risk factors (e.g., life stress) that
inuence neurodevelopmental trajectories, result in the gen-
eration and experience of additional psychosocial stress, and,
thus, may represent an important pathway to developing
*Tiffany C. Ho
tiffany.ho@ucsf.edu
1Department of Psychology, Stanford University, Stanford, CA,
USA
2Department of Psychiatry and Weill Institute for Neuroscience,
University of California, San Francisco, San Francisco, CA, USA
3Psychiatry Department and Douglas Mental Health University
Institute, McGill University, Montréal, QC, Canada
1234567890();,:
1234567890();,:
STBs. Other notable aspects of development, such as pubertal
timing, and factors that may drive atypical pubertal timing
(e.g., exposure to early adversity), are also recognized as
important in affecting the development of adolescent-onset
psychiatric disorders, including STBs [10]. Indeed, atypical
pubertal timing results not only in corresponding changes in
neuroendocrine systems but also signicant shifts in social
identity and perceived status may explain sex differences in
the emergence and course of STBs (and related mental health
conditions that substantially increase the risk of STBs).
The goal of this review paper is to elucidate how pubertal
changes contribute to STBs in adolescence. While we do
not regard puberty-related processes as primary determi-
nants of STBs, we posit that exposure to sex hormones
during adolescence initiates a period of plasticity in neural
circuits that are sensitive to social context (including social
stressors that amplify emotion dysregulation and impulse
control in ways that increase risk for STBs) and that may be
target mechanisms for treatment of STBs, We further con-
tend that these processes are especially critical to consider
in adolescents who have existing vulnerabilities, including
neurobiological sensitivity to hormonal changes, exposure
to adverse psychosocial experiences during early develop-
ment, and underlying mental disorders. In the following
sections, we: (1) present an overarching framework and
highlight specic brain circuits involved in social cognition,
emotion regulation, and impulse control that are relevant to
understanding STBs in adolescence; (2) review basic
experimental ndings in adults that show the effect of
pubertal hormones on behaviors relevant to suicide risk and
survey the conicting correlational literature on hormones
with STBs in adolescents; (3) discuss the role of pubertal
timing and sex differences in these processes; and (4)
advance recommendations for future research in this area.
Puberty as a driver of neuroendocrine
mechanisms relevant for understanding
adolescent STBs
Contemporary theories of suicide have been informed pri-
marily by data from adults [11,12], it is unclear whether
these theoriesor specic components of associated mod-
elsextend to adolescents. Moreover, few theories of sui-
cidality explicitly integrate biological factors, including
endocrine or neural factors (with exceptions [1214]). Here,
we posit that adolescent STBs result from a pathological
response to stress during a time when the neurobiological
systems that regulate stress are recalibrating. There is
extensive evidence that STBs in adolescents are often pre-
ceded by life stressors, particularly stressors characterized
by interpersonal rejection, loss, or conict [1517]. Social
rejection in particular is a commonly experienced stressor
during adolescence due to unstable romantic and peer
relationships, and is a potent trigger for negative emotions
[18,19]. In this context, emotion-related impulsivitythe
tendency to react impulsively during experiences of
heighted affective states [20]may contribute to adolescent
risk for STBs. Indeed, both peer-related stressors [21,22]
and emotion-related impulsivity [23] have been shown
separately to predict STBs in adolescents.
We argue here that hormonal changes during puberty alter
the development of brain circuits implicated in the regulation
of social stressors. The brain is a target organ for all of the
pubertal hormones we review, and receptors for these hor-
mones are expressed abundantly in several key structures that
comprise brain networks governing social cognition, emotion
regulation, and impulse control generally (and social rejection
and emotion-related impulsivity specically) [24,25],
including the amygdala, hippocampus, striatum, anterior
cingulate cortex (ACC), medial prefrontal cortex (mPFC), and
orbitofrontal cortex (OFC). The amygdala, hippocampus, and
ACC comprise a network of regions implicated in detecting
social salience; the amygdala and mPFC comprise a key
emotion regulatory circuit; the striatum and frontal regions
including OFC comprise circuits underlying reward valuation
and impulse control. It is important to note that many of these
regions already exhibit sexual dimorphism during adoles-
cence, including larger amygdala and hippocampal volumes
in boys, greater variance in the hippocampus and striatum in
boys than in girls, sex differences in white matter organization
of callosal, cerebellar, and long-range association tracts (for a
review, see [26]), and sex differences in the developmental
trajectories of frontoparietal networks [27]. Moreover, several
of these regions have been highlighted in a recent review of
neuroimaging markers associated with STBs across the life-
span [28]; however, those authors did not consider the
importance of pubertal development in explaining the neu-
roendocrine basis of the emergence of STBs. We contend that
this is a critical factor to consider, given emerging evidence
that sex differences in these brain circuits during adolescence
appear to be explained, at least in part, by changes in pubertal
hormones [29,30]. Thus, we contend that the social land-
scape, and most notably environmental stressors, experienced
by boys and girls begins to differ during puberty, and that
these differences, in conjunction with sex-specic hormonal
effects on brain maturation, may explain important sex dif-
ferences in STBs that emerge by mid-adolescence.
Pubertal hormones and suicide-relevant
thoughts and behaviors
Puberty is composed of two phases: adrenarche and gona-
darche. During adrenarche, which typically begins around
ages 78 years, the adrenal glands produce increasing levels
T. C. Ho et al.
of the hormones dehydroepiandrosterone (DHEA) and tes-
tosterone [31,32]. Gonadarche is a longer process that typi-
cally takes 45 years, beginning around ages 910 and
occurring on average a year earlier in girls than in boys [33].
Gonadarche is triggered by the activation of hypothalamic-
pituitary-gonadal (HPG) axis, which leads to rising levels of
luteinizing hormone (LH) and follicle-stimulating hormone
(FSH). The release of LH and FSH initiates the development
of the gonads, which, in turn, leads to increases in sex
hormonesspecically, testosterone in boys and estradiol
(the predominant estrogen during adolescence) and proges-
terone in girlsand, by early to mid-adolescence, the
development of secondary sex characteristics and other phy-
sical changes [32]. Importantly, these hormones cross the
blood brain barrier, inuence brain development, and affect a
wide variety of signaling pathways (e.g., neurotransmitter
activity) that underlie mood and cognition [25]. Thus, puberty
involves transformation across virtually every psychobiolo-
gical domainendocrine, neural, physical, cognitive, and
socioemotionaland represents a vulnerable time during
which STBs may emerge.
Prevalence rates of STBs begin to increase after age 12 but
peak in later adolescence, suggesting that it is not simply the
rise in these hormones that accounts for the increase in STBs,
given that these endocrine changes begin much earlier than
age 12. Instead, the organizational effects of pubertal hor-
mones play a major role in risk for STBs in adolescence.
Organizational effects are those that occur during sensitive
periods of development that lay the foundation for sex-typical
brain and behavioral phenotypes and, thus, have an impact
even in the absence of circulating levels of the hormones. In
contrast, activational effects facilitate the expression of
behaviors under specic contexts but are temporary and occur
only when the hormones in question are present [34]. Evi-
dence for organizational effects of hormones during adoles-
cence comes from studies demonstrating that the same
developmental processes (e.g., neurogenesis, synaptic prun-
ing, dendritic branching, apoptosis) that occur during the
perinatal period as a result of surging levels of gonadal hor-
mones also occur during puberty [35,36]. These studies
suggest that puberty opens a sensitive window for experience-
dependent plasticity in neural circuits that underlie higher-
order processing of social stimuli, thereby rendering adoles-
cencea time of increased exposure to social stressorsa
vulnerable period for the onset of stress-related mental health
problems (Fig. 1). At the same time, it is possible that
ongoing uctuations in hormone levels are also related to
STB risk through their activational effects on neurotransmitter
systems, particularly in the context of responding to social
stressors, and that these effects are more relevant for adoles-
cents with a neurobiological sensitivity to hormonal changes.
Below, we review studies investigating ovarian (estradiol
and progesterone), testicular (testosterone), and adrenal
(DHEA) hormones in relation to STBs and, importantly,
their impact on the structural development and function of
specic brain circuits implicated in social cognition, emo-
tion regulation, and impulse control. A summary of this
section is outlined in Table 1and Fig. 1B. Before reviewing
the correlational literature in adolescents, within each
respective section, we rst review basic experimental stu-
dies that provide insight concerning the effects of estradiol,
progesterone, testosterone, and DHEA on human behaviors
relevant to suicide risk in adults in order to provide a more
reliable context for the actions of these hormones.
Finally, because ovarian hormones have been investi-
gated almost exclusively in females in this context and,
similarly, androgen hormones overwhelmingly in males,
our review of the association of these hormones with brain
and behavioral outcomes inherently raises issues of sex
differences or sex-specic effects in these processes;
we discuss these issues in more detail in a later section
(D. Consideration of sex differences).
Ovarian hormones (estradiol and progesterone)
Estradiol, progesterone, and their neurosteroid metabolites
all increase and begin to uctuate cyclically in girls during
puberty [34]. Consequently, the vast majority of research in
this area has focused on females (for reviews, see [14,37]).
To provide a context for understanding the effects of
estradiol and progesterone administration on behaviors
relevant to suicide risk, we will rst briey review studies
that controlled hormone conditions in women with affective
symptoms and premenstrual dysphoric disorder (PMDD),
for whom suicide risk is elevated compared to the general
population [38,39].
Decades of research have shown that there are no dif-
ferences between women with and without PMDD in
ovarian hormone levels or related neurosteroids (e.g., allo-
pregnanolone) but that suppressing ovarian hormones
reduces or eliminates symptoms of PMDD (for a review,
see [40]). A recent study that controlled for ovarian hor-
mone secretion and exposure in women with PMDD has
helped to clarify these two seemingly opposing ndings
[41]. In that study, women with PMDD who responded to a
gonadotropin-releasing hormone agonist treatment were
given placebo for one month before being administered
continuous estradiol/progesterone for three months;
researchers found that changes from low to high levels of
ovarian hormones, but not absolute levels of ovarian hor-
mones, were associated with increases in negative affect
[41]. Together, these data suggest that neurobiological
sensitivity to hormone changes is an important factor that
may explain certain clinical phenomena, such as PMDD and
suicide risk. Indeed, a recent review has covered this topic
extensively in adult women, demonstrating that cyclical
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
hormone changes may play an important role in acute risk
for daily suicidal ideation, planning, and intentin indivi-
duals with sensitivity to hormone changes [14]. We have
extended this theoretical model to adolescents, proposing
that in those who possess a neurobiological sensitivity to
hormonal changes, the normative uctuations during this
transitional period, coupled with adolescent-typical experi-
ences of greater exposure to life stressors, may exacerbate
the effects of these processes on emotion regulatory and
impulse control circuits. Indeed, results from studies that
have investigated suicide risk in adult women across the
menstrual cycle have been inconsistent. These studies have
typically assessed levels of estradiol and progesterone
during different phases of the menstrual cycle [4244], and
in women with low levels of estradiol and progesterone
(e.g., amenorrhea or menopause [45]). While some
investigators have reported that a higher risk of suicide
attempts and more severe suicidal thoughts and intentions
are associated with relatively low or declining levels of
estradiol and progesterone (i.e., during the early follicular/
menstrual or pre-menstrual phases), other researchers have
not found differences in estradiol or progesterone between
depressed women with and without STBs [46] or effects of
the menstrual cycle on suicide attempts in women with
PMDD [39].
The fact that there has been no evidence of differences in
ovarian hormone levels or in cyclical changes in female
adolescents with and without STBs is consistent with the
previously aforementioned studies in adult women with
reproductive mood disorders where the effect of menstrual
cycle and/or ovarian hormone levels on STBs in adult
women is absent. One interpretation is that it is the
Fig. 1 Summary of prevalence rates of suicidal ideation,
concentration of sex steroids, and brain volume as a function of
age and typical associations between pubertal hormones and brain
structures reported the extant literature. A) Graphical depictions of
prevalence rates of suicidal ideation, concentrations of sex steroids,
and brain volume as a function of age. Shaded region indicates
puberty. The schematized trajectories of gray matter volume adjusted
for total brain volume are based on data reported in [162]. B) Summary
of typical associations between pubertal hormones and brain structures
from both adolescent and adult samples. ACC anterior cingulate cor-
tex, AMYG amygdala, HPC hippocampus, MPFC medial prefrontal
cortex, OFC orbitofrontal cortex, STM striatum.
T. C. Ho et al.
Table 1 Summary of studies examining gonadal and adrenal hormones in relation to suicide attempts and suicidal ideation.
Publication Sample size and characteristics Age (years) Sex Psychiatric condition Suicide-related
outcome Study design and methods Findings Additional notes
Afzali et al. 2012 81 suicide attempters Mean =23.63
SD =8.41
Range =1555
F Assorted
(25 Past mental disorder, 22
Previous suicide attempt)
History of Suicide
Attempts Structured interview over
6 months after attempt Suicide attempts were not
associated with menstrual
cycle phase.
Patients with irregular
menstrual cycles were
excluded.
Baca-Garcia et al.
2010a281 suicide attempters
176 healthy controls Mean =30.8
SD =8.8
Range =1892
F Assorted
(229 Mood disorder, 229
SUD, 275 Previous
psychiatric treatment)
Recent Suicide
Attempts and Recent
Suicidal Ideation
Blood sample within 24 h of
attempt: estradiol,
progesterone, LH, FSH
Suicide attempts were was
more likely during the
follicular phase.
Suicide intent severity was
elevated during low-estrogen/
low-progesterone states (pre-
menstrual phase, amenorreha,
menopause)
Buttereld
et al. 2005 130 inpatients Mean =49.4
SD =8.13 M PTSD Recent History of
Suicide Attempts (past
6 months) and
Suicidal Ideation
Blood: DHEA,
androstenedione,
testosterone, estradiol
Suicide attempters had higher
DHEA than nonattempters
Cayköylü et al.
2004a52 suicide attempters
50 healthy controls Mean =26.51
SD =7.82
Range =Not
Reported
F Assorted
(8 PMDD, 1 SCZ,
2 MDD, 1 OCD)
Recent Suicide
Attempts Blood sample within 12 h
of attempt: estradiol,
progesterone
Menstrual status determined
with self-report.
Suicide attempts were more
frequent during the
follicular phase.
Estradiol and progesterone
levels were not different in
suicide attempters compared to
healthy controls.
Patients attempting suicide
with OD or admitted to the
ICU were excluded.
Chatzittos
et al. 2013 28 suicide attempters
(10 female, 18 male)
19 healthy controls
(7 female, 12 male)
SA:
Mean =44
SD =14.6
Range =2666
HC:
Mean =30
SD =Not Reported
range =2348
Both Assorted
(14 Mood disorder,
4 Anxiety Disorder,
9 SUD, 19 PD)
History of Suicide
Attempts CSF: DHEA-S, DHEA,
cortisol, and 5-HIAA In males, suicide attempters
had higher CSF DHEA-S
levels compared to healthy
controls.
In females, no signicant
differences.
Exposure to early adversity
(e.g., interpersonal violence)
correlated negatively with
cortisol/DHEA-S ratio
Dogra et al. 2007a217 suicide decedents
237 non-suicide decedents 45% of suicide
dececents
ages 2130
Bimodal
distribution in the
non-suicide
decedents: 23%
ages 2025, 23%
ages 3035
Range =1145
F Not Reported Suicide Death Autopsy
Menstrual status determined
by visual examination of the
uterine cavity
54.46% of non-pregnant
women who died by suicide
were menstruating versus
6.75% in the non-suicide
decedent group
Fouriestié et al.
1986a108 suicide attempters Mean =25.3
SD =4.0
Range =Not
Reported
F Assorted
(9 with previous psychiatric
admission, 15 treated with
neuroleptic and/or
antidepressant medication,
9 treated with anxiolytics,
15 with previous suicide
attempts)
Recent Suicide
Attempts Blood sample within 12 h
of attempt: estradiol and
progesterone
Suicide attempts were more
likely to happen during phases
with low estradiol, during the
rst week of the menstrual
cycle (42%) and after the
fourth week (12%).
Frequency of suicide attempts
did not vary signicantly
during the menstrual cycle in
OC users.
Patients admitted to the ICU
were excluded.
Gustavsson
et al. 2003 43 suicide attempters Mean=38.0
SD =12.0
Range =Not
Reported
M Assorted
(14 SUD, 9 DDNOS, 10
MDD, 4 Dysthymia,
9 Adjustment disorder,
4 Anxiety disorder, 2
Psychosis)
Recent Suicide
Attempts CSF in days (557 days,
mean =16) following
suicide attempt: testosterone
Suicide attempters with
depressive disorders showed
higher CSF testosterone than
those with other psychiatric
diagnoses.
CSF testosterone positively
correlated with irritability
and negatively correlated
with social desirability.
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
Table 1 (continued)
Publication Sample size and characteristics Age (years) Sex Psychiatric condition Suicide-related
outcome Study design and methods Findings Additional notes
Papadopoulou et al.
2018a70 suicide attempters Mean =35.5
SD =8.9
Range =1852
F Assorted
(28 MDD, 13 BD, 14
Psychosis, 15 PD or
adjustment disorder)
Recent Suicide
Attempts Blood sample within 72 h of
suicide attempt or within 48
h after transfer to the ICU:
progesterone, LH, FSH
Menstrual status determined
with progesterone levels, LH
and FSH were used to rule
out menopausal status
Suicide attempts were more
frequent in the last 4 days of
days of luteal phase and during
the 4 days of menses.
No effect of menstrual status
on lethality (violent vs non-
violent mode of attempt) or
psychiatric diagnosis.
Markianos et al.
2009a15 suicide attempters
(intentional jumps)
18 accident victims
(falling from a height)
40 healthy controls
SA:
Mean =39.9,
SD =14.3,
Range =2262
Non-SA: Mean =
37.6 SD =15.2
Range =2066
HC:
Mean =31.6
SD =9.0
Range =2559
M Assorted
(10 SCZ, 5 MDD) History of Suicide
Attempts Blood: testosterone,
LH, FSH Suicide attempters had lower
levels of testosterone (trending,
p=0.065) and LH compared
to accident victims.
Both suicide attempters and
accident victims had lower
levels of testosterone and LH
compared to HC.
Martin et al. 1997b81 female and 79 male
adolescents Mean =16.0
SD =1.0
Range =1519
Both Not Reported History of Suicide
Attempts and Suicidal
Ideation
Blood: progesterone In males, progesterone was
higher in those with past
suicide attempts and with
suicide ideation.
In females, progesterone levels
negatively correlated with past
suicide attempts and disclosed
suicide ideation
SUD excluded from
analyses.
Roland et al. 1986 39 suicide decedents
48 non-suicide (sudden death)
decedents
SA:
Mean =39.1
SD =18.3
Range =1576
HC:
Mean =51.5
SD =13.8
Range =1279
M Not Reported Suicide Death Autopsy
Blood: testosterone Suicide decedents showed
higher levels of testosterone
compared to non-suicide
decedents.
Sher et al. 2012 67 patients with bipolar
disorders and at least one past
suicide attempt (51 female,
16 male)
Mean =34.5
SD =9.9
Range =1875
Both Bipolar Disorder History of Suicide
Attempts Blood: testosterone Testosterone levels positively
correlated with the number of
past suicide attempts, while
controlling for sex.
Testosterone levels were also
positively correlated with
number of manic episodes,
while controlling for sex.
Sher et al. 2014 51 patients with bipolar
disorder and at least one past
suicide attempt
Mean =33.2
SD =9.6 F Bipolar Disorder History of and
Prospective Suicide
Attempts (prospective
follow-up for up to
2.5 years)
Blood: testosterone At baseline, testosterone levels
positively correlated with the
number of suicide attempts and
past major depressive episodes.
Higher testosterone levels
predicted suicide attempts in
the follow-up period.
Sher et al. 2018a17 combat veterans with post-
deployment suicide attempt (0
female, 17 male) and 17 non-
suicidal combat veterans
(2 female, 15 male)
SA:
Mean =37.5
SD =11.6
Non-SA: Mean =
35.7 SD =10.8
Both PTSD History of Suicides
Attempt and Suicidal
Ideation
Blood: DHEA, DHEA-S Suicide attempters had lower
levels of DHEA and DHEA-S
compared with nonattempters.
Suicidal ideation negatively
correlated with DHEA and
DHEA-S levels across all
participants.
Suicidal ideation negatively
correlated with DHEA-S levels
in nonattempters.
DHEA/DHEA-S ratios
positively correlate
with adolescent and adult
aggresion scores in suicide
attempters.
T. C. Ho et al.
Table 1 (continued)
Publication Sample size and characteristics Age (years) Sex Psychiatric condition Suicide-related
outcome Study design and methods Findings Additional notes
Stefansson
et al. 2016 28 suicide attempters
(10 female, 18 male)
19 healthy controls
(7 female, 12 male)
SA:
Mean =44.0
SD =14.6
Range =2366
HC:
Mean =30.0
SD =Not Reported
Range =2348
Both Assorted (MDD,
PTSD, SUD) Recent Suicide
Attempts and
Prospective Suicide
Death (prospective 21-
year follow-up)
CSF and blood in days
(mean =8.6, range =
217 days) following suicide
attempt: testosterone, cortisol
In males, CSF and blood
testosterone levels were higher
in suicide attempters compared
to healthy controls.
In females, no differences.
In males, CSF testosterone/
cortisol ratio positively
correlated with impulsivity
and aggressiveness in the
suicide attempters.
No differences associated
with MDD, PD, or SUD
Tripodianakis
et al. 2006 80 suicide attempters
(29 with schizophrenia)
56 healthy controls
29 nonattempters with
schizophrenia
SA:
Mean =34.4
SD =12.6
HC:
Mean =35.3
SD =8.7
M Schizophrenia History of Suicide
Attempts Blood: testosterone,
LH, FSH Suicide attempters had lower
blood testosterone compared to
healthy controls.
Attempters with schizophrenia
had lower levels of testosterone
compared to nonattempters
with schizophrenia.
Attempters who used a
violent method had lower
testosterone levels than non-
violent attempters.
Zhang et al. 2015 245 suicide attempters
(172 female, 73 male)
245 healthy controls
(172 female, 73 male)
SA:
Mean =42.9
SD =Not Reported
Range =1650
HC:
Mean =37
SD =Not Reported
Range =1453
Both Not Reported History of Suicide
Attempts Blood: testosterone In males, testosterone was
higher in male suicide
attempters compared to healthy
controls.
In females, no signicant
differences.
Ffemale, FH follicular hormone, DDNOS depressive disorder not otherwise specied, ICU intensive care unit, LH lutenizing hormone, Mmale, MDD major depressive disorder, OC oral
contraceptive, OD overdose, OCD obsessive-compulsive disorder, PTSD post-traumatic stress disorder, SA suicide attempt, SCZ Schizophrenia, SD standard deviation, SUD substance use
disorder.
aLow-quality study due to limited sample size and/or limitations in study design (e.g., single-timepoint cross-sectional associations between ovarian hormones and suicidal thoughts and
behaviors).
bAdolescent sample.
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
neurobiological sensitivity to changes in estradiol and
progesteronerather than between-person differences in
levels of these hormonesthat represents a key trait-like
source of variance for understanding suicide risk in the
context of puberty, a time when the stability of these hor-
mone dynamics is still in ux [47]. Intensive (e.g., daily
samples) longitudinal studies conducted in adult women
have also provided little evidence that hormone levels at a
single point in time are correlated with levels within the
same cycle and also for subsequent cycles, particularly for
estradiol [48,49]. It is also important to note that both
between-person and within-person variability of ovarian
hormone levels are affected by multiple confounds that
investigators ought to consider, including, but not limited,
to cycle length [50], diurnal effects [51], cycle phase
[48,49], effects of study participation [48], anovulation
[52], culture and/or diet [53], and personal and family
medical history (e.g., polycystic ovary syndrome, breast
cancer [48,54]). Nevertheless, dense-sampling studies have
demonstrated robust between-person effects of menstrual
phase, such that progesterone is reliably higher in the luteal
phase relative to the follicular phase and, albeit to a lesser
extent, estradiol is higher in the follicular phase relative to
the luteal phase [48,49]. We note in Table 1which studies
employ non-experimental cross-sectional designs and that
attempt to relate ovarian hormones without consideration of
these relevant factors (e.g., cycle phase).
Given these limitations in measuring ovarian hormones,
it is not surprising that little is known about the effects of
estradiol and progesterone on the human brain in general,
much less on the circuits we have hypothesized are impli-
cated in STB risk. Nonetheless, there is evidence that
estrogen receptors are expressed strongly in brain regions
involved in social cognition broadly and social rejection
specically, including the amygdala, hippocampus, ACC,
and vmPFC [5558]. Several studies have also found that
women with comparably higher levels of estradiol (endo-
genous or synthetic) show greater amygdala-based resting-
state functional connectivity and activation in ACC and
vmPFC, which are key regions involved in processing
salient information [5961]. Similarly, longitudinal studies
with naturally cycling women have documented larger
hippocampal gray matter volumes during periods of rela-
tively high estradiol (late follicular/preovulatory phases)
than of relatively low estradiol early follicular/premenstrual
phases [62,63].
There is also evidence from functional magnetic reso-
nance imaging (fMRI) studies wherein groups of women
during different phases of their menstrual cycle are com-
pared that show that relatively higher levels of estradiol
(pre-ovulatory phase) are associated with greater activation
of the hippocampus during both cognitive tasks [64] and
affective stress tests [65], and with stronger functional
connectivity of the hippocampus with brain regions
involved in processing salient information [63]. Similarly,
higher levels of progesterone (luteal phase) have also been
found to be associated with activation of the striatum and
PFC during cognitive processing [64]. Finally, results from
fMRI studies utilizing dense-sampling designs of naturally
cycling women have reported intriguing, albeit inconsistent,
results. One study found no effects of estradiol across the
menstrual cycle on intrinsic connectivity patterns [66],
whereas another found that higher levels of estradiol drove
stronger subsequent connectivity within attention networks
(specically among brain circuits that are implicated in
internally focused attentional states and externally focused
attentional states) [67]. In contrast, one study found that
progesterone mediated patterns of positive functional con-
nectivity between the hippocampus and PFC [66] whereas
another study found that higher levels of progesterone was
associated with lower connectivity across all networks [67].
It is clear that additional research is needed to clarify the
magnitude and directionality of the effects of estradiol and
progesterone on patterns of brain connectivity, particularly
in adolescents, and their effects on longer-term neurode-
velopmental trajectories. It is also critical that researchers
work to characterize the extent to which these hormones
effect brain circuits specic to STBs, or alternatively,
whether they are implicated in mental illness risk more
broadly. Nevertheless, the studies we have reviewed pro-
vide initial evidence that brain circuits that support the
regulation of affectively salient stimuli are sensitive to the
effects of estradiol and progesterone; importantly, these
same circuits have also been shown to be affected in adults
and adolescents with STBs [28,68].
Additional research is also needed to elucidate the pre-
cise neural mechanisms by which estradiol may impact the
development of brain circuits, including its effects on neu-
rotransmitters. Estradiol has also been found to alter ser-
otonin transmission, binding, and metabolism by increasing
the production of tryptophan and inhibiting the expression
of the serotonin reuptake transporter gene (for a review, see
[69]). Serotoninergic abnormalities in the number of ser-
otonergic neurons and in serotonin transportation, receptor
binding, and levels have all been found in victims of suicide
[70,71] (for a review, see [72]). Serotonin has been shown
to be associated with cortisol reactivity to stressors [73]
and positively associated with greater 5-HT1A receptor
bindingwhich could contribute to lower serotonin sig-
naling by inhibiting further serotonin release into the
synapsein depressed patients who died by suicide versus
both depressed and psychiatrically healthy individuals who
did not die by suicide [74]. Similarly, allopregnanolone, a
progesterone metabolite, binds to GABAAreceptors, which
mediates the majority of inhibitory signaling in the brain
[75] and plays an important role in downregulating the
T. C. Ho et al.
HPA-axis in response to acute stressors [76]. Several stu-
dies have found that stress exposure alters the availability of
GABAAreceptors as well as their composition and sensi-
tivity to neurosteroid regulation, which, in turn, can inu-
ence subsequent responses to stress [7779]. Emerging data
has also implicated GABA dysfunction in STBs, whereby
depressed patients who died by suicide showed a higher
expression of genes that encode for proteins involved in
GABAergic synaptic transmission in the ACC and a lower
expression of these genes in the dorsolateral PFC than did
both depressed and psychiatrically healthy individuals who
did not die by suicide [80]. Future work is needed to
explicitly investigate the extent to which patterns of struc-
tural or functional connectivity of circuits involving the
amygdala, hippocampus, ACC, and PFC that are develop-
ing in response to puberty-related changes in estradiol and
progesterone (and related neurosteroids) exhibit corre-
sponding changes in neurotransmitter systems that support
socioemotional processes relevant to STB risk.
Testosterone
Unlike ovarian hormones, which have been studied pri-
marily in female adolescents, investigators have found in
both sexes that levels of androgens are associated with
appetitive behaviors, aggression, competition, and other
related social behaviors that are relevant to STB risk
[81,82]. In female adolescents, the sex steroid with the
most androgenic activity is DHEA, which is produced by
the adrenal cortex (discussed separately in the following
section); in males adolescents, it is testosterone [31,32].
In contrast to the data published thus far on ovarian
hormones, there is evidence of strong within-person relia-
bility and stability in testosterone levels in both men and
women. Indeed, some researchers contend that levels of
testosterone are trait-like [83]. Perhaps not surprising, tes-
tosterone has been linked more strongly with STB risk, and
specically with suicide attempts, than has any other pub-
ertal hormone (see Table 1). However, the empirical nd-
ings thus far have been mixed (for opposing reviews, see
[84,85]). Several researchers have theorized that testoster-
one is linked to suicidal behaviors through its modulation of
emotion-related impulsivity and impulsive aggression,
which are considered to be among the most robust predis-
posing traits to suicidal behaviors in youth [86]. In drawing
from the literature of studies in which testosterone is
administered and behaviors are subsequently assessed, as
well as studies that link changes in social interactions with
changes in endogenous testosterone, there appear to be
reliable effects of testosterone on socially motivated beha-
viors, including exerting dominance and displays of
aggression (either physically or non-physically) and other
social status seeking behaviors (for a review, see [87]).
While testosterone exert complex effects on interpersonal
behavior, longitudinal studies show that the puberty-related
increases in testosterone are not accompanied by a con-
current rise in aggressive behaviors [83,88,89]. Additional
studies in animals as well as in humans [90] suggest that
testosterone levels correlate more closely with social dom-
inance, rather than aggressive behaviors. Hence, testoster-
one may be an important moderator of the behavioral
response to events associated with loss of social status [91],
which are known precipitants of STBs. In the context of
adolescent STB risk, it may very well be that testosterone is
a driver of heightened sensitivity to social context, which
can lead to signicant emotion dysregulation and impulse
control and, in turn, elevated STB risk.
Consistent with these points is evidence that elevated
testosterone has also been found in male adults to be
associated with psychological states and individual traits
associated with suicide risk, including depression severity,
irritability, and impulsive aggression [92,93]. Researchers
have found higher levels of testosterone in both male and
female suicide attempters than in their same-sex non-
attempting counterparts [94], and in post-mortem samples
of suicide completers compared to individuals who died
from other causes [95]. In a recent study, male, but not
female, young adults who attempted suicide had higher
levels of testosterone than did age- and sex-matched healthy
volunteers [92]. Thus, there studies do not preclude the
possibility that testosterone levels are higher in individuals
with a mental disorder than they are in healthy persons. One
study found in women with bipolar disorder with a history
of suicide attempt that higher testosterone levels predicted
subsequent suicide attempts [96]. Interestingly, in another
sample of men and women with bipolar disorder, higher
levels of testosterone were associated with number of sui-
cide attempts only after controlling for sex [97]. In contrast,
other studies have found lower levels of testosterone both in
male suicide attempters relative to psychiatric healthy
controls [98] and in patients who were hospitalized due to
accidents [99]. One possible discrepancy for these ndings
involves the measurement of testosterone in plasma versus
cerebrospinal uid; studies assaying plasma testosterone
have typically reported inverse associations with suicide
attempts. Nevertheless, it is clear that adequate clinical
controls (attempters or completers relative to nonattempters
who otherwise have similar clinical histories) must be
included in research in order to clarify the results of these
studies.
As is the case with research on ovarian hormones and
STBs, no studies have examined the psychobiological
mechanisms and specic brain circuits through which tes-
tosterone may drive risk for STBs in either adolescents or
adults. Testosterone inuences many of the same neuro-
transmitters as does estradiol [25]; moreover, testosterone
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
can be converted into estradiol via aromization [100],
adding additional complexity in these processes should be
addressed in future work. That said, one pathway through
which testosterone may affect mood and cognition is
through altering dopaminergic neurotransmission: several
studies have found evidence of differences between suicide
attempters and completers in dopamine receptor density,
transporter binding capacity, and metabolism [101103],
primarily in the striatum. In fact, testosterone receptors are
widely expressed in dopaminergic neurons of the striatum
and portions of the PFC, including vmPFC and OFC [104].
Research with both macaques and humans has documented
changes in the mechanisms that underlie dopamine signal-
ing during adolescence, which may be explained, in part, by
effects of testosterone (and estrogen; for reviews, see
[105,106]). In particular, testosterone has been shown to
increase basal dopamine levels and decrease the number of
enzymes that break down dopamine in the striatum and PFC
[105]. Consequently, it is possible that adolescent-typical
increases in testosterone during puberty, especially in boys,
contribute to elevated STB risk through enhanced dopami-
nergic transmission. This is consistent with previous reports
that, compared to depressed patients who did not die by
suicide, depressed patients who die by suicide have lower
growth hormone responses to apomorphine [102,107],
which is an indication of higher dopamine transporter
binding [108]. Nonetheless, it is important to note that some
studies provide evidence of lower dopamine (measured by
lower levels of homovanillic acid and total dopamine in
urine) in patients who attempted suicide versus those who
did not, see [103], or of no signicant associations between
dopamine levels/receptor binding and STBs [109].
With respect to brain development, higher levels of
pubertal testosterone have been associated with white
matter organization in tracts implicated in social cognition
and emotion regulation, including the uncinate fasciculus
(which connects the amygdala and vmPFC) and corpus
callosum in both boys [110,111] and girls [29,112].
Several researchers have documented other effects of tes-
tosterone on adolescent and young adult brain and behavior.
Human neuroimaging studies using fMRI have shown that
in healthy young men, activation in the amygdalaa brain
structure that is rich in androgen receptors [63] and is
affected by circulating androgens [64]to fearful and angry
faces co-varies positively with individual differences in
serum testosterone concentrations [65,66] (but see [67] for
opposing results). In an experimental study, administration
of testosterone was associated with increased amygdala
reactivity to threat-related stimuli in young women [113].
These results are consistent with a longitudinal study in
adolescents that found that increased levels of pubertal
testosterone disrupted typical coupling between the amyg-
dala and OFC, leading to increased amygdala reactivity to
threat-related stimuli in both sexes [114]. Finally, whereas
in boys higher testosterone levels are associated with lower
activation in the striatum and PFC during processing of
emotional conict, in girls higher testosterone levels are
associated with lower activation in the PCC/precuneus
[115]. Other investigators have shown in both boys and
girls that higher levels of testosterone are associated with
higher striatal activation during reward consumption [116],
and with higher striatal activation when adolescents select
smaller yet more immediate rewards [117]. In an observa-
tional longitudinal neuroimaging study in male and female
adolescents between the ages of 827 years, activation in
the striatum (nucleus accumbens) to rewarding stimuli
peaked during adolescence, and was associated with
accompanying changes in testosterone levels [118]. In a
recent study, in which salivary testosterone was measured in
adolescents before and after a neuroimaging scan, acute
increases in testosterone were associated with smaller dif-
ferences in activation between reward cues signaling reward
or non-reward outcomes for a given trial in vmPFC and
posterior cingulate cortex (PCC) in both sexes [119]. Thus,
by altering neural processing of both negative (e.g., threa-
tening) and positive (e.g., rewarding) valenced stimuli,
testosterone may be facilitating the expression of adolescent
sensation-seeking and risk-taking behavior [118] through
dopaminergic transmission among these subcortical and
prefrontal circuits. Certainly, these processes may not be
specic to STBs and may represent a more general diathesis
for mental disorders that are characterized by sensation-
seeking and risky behaviors; however, future research is
needed to test this possibility more explicitly.
In sum, studies to date have demonstrated that higher
levels of testosterone are associated with aspects of emotion-
related impulsivity in males with a history of suicidal
behaviors (e.g., attempts). Higher levels of testosterone are
also associated with steeper temporal discounting of rewards
driven by patterns of activation in striatal, vmPFC, and OFC
in both sexes; sex differences in the effect of testosterone on
adolescent brain circuits appear to be most prominent in
emotion-related contexts. Indeed, in adults, both endogen-
enous levels [120] and exogenous administration [121]of
testosterone enhance risk taking. The predisposition to take
risky decision has been shown to be a critical risk factor for
suicidal behavior in adults [122] and, to a lesser extent given
the sparse literature on this topic, in adolescents [123].
Hence, functional alterations in decision-making systems
induced by puberty-related rises in testosterone levels, or
their context-dependent uctuations, might be related to
suicidal risk through an increase probability of perpetrating
highly risky and self-destructive behaviors in the face of
overwhelming stress (particularly social stressors involving
loss of desired status). Future work is needed to test expli-
citly whether higher levels of testosterone exacerbate affect
T. C. Ho et al.
striatal and PFC systems underlying maladaptive responses
to social stressors that, in turn, lead to heightened risk
for STBs.
DHEA
DHEA (and its sulfate, DHEA-S) is the most abundant
steroid hormone in humans and is a precursor to sex ster-
oids. Researchers have not yet examined the relation
between DHEA (or DHEA-S) and STBs in adolescents;
moreover, and the few studies that have been conducted
with adults differ in their measurement of DHEA. One
study showed that male suicide attempters had higher levels
of DHEA-S than did healthy controls, and that exposure to
extreme social threat (i.e., interpersonal violence) as a child
was negatively correlated with the ratio of cortisol/DHEA-S
[124]. Among adults diagnosed with PTSD, those who had
attempted suicide had signicantly higher levels of DHEA
than did those without a history of attempt [125]. Another
study found that, in combat veterans, the ratio of DHEA/
DHEA-S was positively correlated with total adolescent
aggression scores, total adulthood aggression scores, and
lifetime aggression scores in those who had attempted sui-
cide but not in nonattempters [126].
Overall, these studies suggest that higher levels of DHEA
or DHEA-S may be associated with risk for STBs. Indeed,
DHEA modulates neurotransmitter systems implicated in
suicidal thoughts and mood disturbances, including acting as
an antagonist for GABA receptors, with genes widely
expressed in hippocampus, amygdala, and striatum [127].
There is also intriguing evidence that psychiatric conditions
characterized by excessive stress and elevated suicide risk
lead to a downregulation of neurosteroid biosynthesis
including the conversion of DHEA to GABAergic metabo-
lites, such as allopregnanolone [128]and changes in
GABAAreceptor subunit composition [129]. Thus, STB risk
may be associated with higher levels of DHEA due to an
insufcient ability to metabolize DHEA. With respect to the
effects of DHEA on the adolescent brain, however, there
have been a small number of studies. Thus, far investigators
have found that higher levels of DHEA are associated with
larger hippocampal volumes in both male and female ado-
lescents [130] and with lower white matter organization
across a broad set of white matter tracts [110]. Other studies
have found that higher DHEA levels are associated with
reduced cingulate activation and greater functional con-
nectivity between the amygdala with ACC and other regions
involved in processing salient information in adolescents
during the processing of social stimuli (e.g., viewing fearful
faces) [131]. Moreover, higher DHEA levels are associated
in girls with lower activation in cingulate regions implicated
in processing salience information and with greater exter-
nalizing problems [132], but in boys with stronger functional
connectivity between the amygdala and inferior frontal gyrus
(the opposite pattern was found in girls) and with higher
anxiety symptoms [131].
Despite the heterogeneity in clinical, developmental, and
demographic features across the studies reviewed in this
section, there appear to be consistent associations between
higher levels of DHEA and both STBs and altered structure
and function of brain circuits underlying emotion regula-
tion. In adults with clinical disorders, higher levels of
DHEA (either alone or relative to cortisol) appear to be
associated with STBs. In psychiatrically healthy adoles-
cents, higher levels of DHEA are associated with reduced
downregulation of affective signals in the amygdala from
emotion regulatory cortical regions, including the ACC and
portions of the PFC. Thus, higher levels of DHEA may
disrupt the development of emotion regulatory brain circuits
across adolescence in ways that increase risk for STBs
when individuals are exposed to interpersonal stressors
(particularly those characterized by threat). However, it is
worth noting one study of psychiatrically healthy adults in
which administration of exogenous DHEA reduced activa-
tion in the amygdala and hippocampus, increased activation
in the vmPFC, and led to stronger connectivity between the
amygdala and hippocampus during an emotion reappraisal
task; moreover, decreased activation in the hippocampus
during the task was associated with lower negative affect,
suggesting that higher levels of DHEA improve negative
mood by downregulating affective signals in the hippo-
campus [133]. Additional research is needed to examine
whether these associations hold in clinical samples of
adolescents and whether (or how) they are related to risk
for STBs.
Consideration of sex differences
Across most countries, being female increases the risk of
suicidal thoughts and related behaviors [5]. Despite this
higher prevalence of STBs in women, men are more likely to
die by suicide [134], leading to the gender paradox of sui-
cide.It is notable that the sex difference in suicide deaths
increases dramatically in adolescence [135], suggesting that
puberty plays an important role in explaining this difference
(although this sex difference is now narrowing among ado-
lescents [136], indicating that other factors, including envir-
onmental or cultural inuences, are also likely). Whereas past-
year ideation, plans, and attempts tend to peak during mid-
adolescence (~16 years) in girls, these rates increase steadily
throughout mid- to late adolescence in boys [137,138].
As in adults [5,139], there are sex differences in suicide-
related outcomes in adolescents that are mediated in part by
differences in lethality and mental illness. In a psycholo-
gical autopsy study, adolescent male suicide victims were
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
more likely to use lethal methods and had a higher pre-
valence of conduct disorder than did female victims [140].
A recent meta-analysis of sex-specic suicide risk in ado-
lescents (ages 1226 years) found distinct clinical and
environmental risk factors for suicide attempt in male and
female adolescents [139]. Thus, in addition to the hormonal
differences that underlie sex-specic neurodevelopmental
changes, different clinical conditions (externalizing dis-
orders in boys versus mood disorders in girls) and social
stressors (peer inuence in boys versus direct trauma/vic-
timization from romantic relationships in girls) may further
explain or moderate these pathways to risk for STBs.
Because of the sparse literature, however, it is not clear
whether sex-specic risk factors are present before puberty,
whether puberty affects neuroendocrine systems in sex-
specic ways to increase risk for STBs, or whether boys and
girls are exposed to differential environmental risk factors
as a result of going through puberty. Future research should
attempt to clarify the extent to which pubertal processes
play a central role in STBs or whether they are implicated in
mental illness more generally.
While adrenal and gonadal hormones and physical
maturation are important indicators of pubertal develop-
ment, other aspects of puberty may be relevant in the
context of adolescent risk for STBs. Pubertal timingthe
age at which individuals mature relative to their peers
[141]has been linked to individual differences in mental
disorders in adolescence [142,143]. Moreover, the timing
of pubertal onset may have a different impact on devel-
oping neuroendocrine function depending on whether it
occurs earlier or later in a given individual [34,139].
Given that girls typically enter puberty earlier than do
boys, considering the role of pubertal timing may also
elucidate sex differences in risk for STBas well
as mental illness more generallyand accompanying
endocrine and neural processes. A growing number of
studies are reporting that early menarche is associated
with elevated risk for suicidal ideation in adolescent girls
[144146]. In a recent longitudinal study of a large birth
cohort, earlier age of menarche was associated with
increased suicidal behaviors at 16 and 21 years of age
[147], suggesting that earlier puberty has an enduring
effect on STB risk throughout adolescence and young
adulthood. In addition, considerable evidence suggests
that pubertal timing is inuenced by early life adversity
[148], which itself is a robust predictor of STBs [149].
Thus, early puberty resulting from early adversity may be
a mechanism by which suicide risk is instantiated or
exacerbated in vulnerable adolescents. Finally, it is
important to acknowledge that sexual orientation and
identication as a sexual minority are increasingly being
recognized as risk factors for STBs [150]; more research
in this area is critically needed.
Future directions
No research has yet examined whether and how pubertal
hormones affect neurodevelopmental trajectories of brain
circuits that mediate social cognition and emotion-related
impulsivity explain risk for STBs during adolescence.
Studies are needed to clarify the role of pubertal timing and
the multidimensional mechanismsbiological, social, cul-
turalby which puberty-related processes inuence risk for
STBs. An important next step for the eld is to rst
establish reliable associations between pubertal hormones
and adolescent brain structure and function and to then map
those associations onto neural circuits underlying STBs in
adolescents. Other critical knowledge gaps include disen-
tangling neuroendocrine mechanisms that are more closely
linked to suicidal thoughts versus attempts (and other self-
harming behaviors) and that facilitate the transition from
ideation to action [151].
Although we focused here on the effects of pubertal
hormones, we should acknowledge that there is consider-
able evidence that HPA-axis dysfunction is associated with
STBs and self-harming behaviors in adolescents
[13,21,152,153]. Puberty is also the time when HPG-axis
regulation of the HPA axis develops [154157]. Given that
the expected suppression of the HPA axis by the HPG axis
[158] is disrupted in individuals at risk for STBs (due to
such factors altering developmental changes in stress
response and regulatory systems, including experiences of
adversity and exposure to trauma during early life [154]), it
is critical that researchers explicitly examine the role of
puberty in this context.
It is also important that researchers in this area consider
moving away from a nomothetic framework for the pre-
diction of STBs and instead adopting idiographic or person-
centered models. Puberty is a highly individualized process;
adolescents differ markedly in their hormone levels, their
neurobiological sensitivity to typical endocrine changes
[14], their pubertal timing, their neurodevelopmental tra-
jectories, and their psychological responses to maturation.
Certainly, measuring these variables at the individual level
is challenging and will have to take into account population
norms on several dimensions (e.g., ethnicity, socio-
demographic factors). Absent such data, standardization
within age bands or residualized scores (e.g., regressing
pubertal stage on age to obtain a measurement of pubertal
timing) can be used to capture individual variability (for a
treatment of these issues, see [159]). Another solution is to
use dense samplingapproaches that leverage repeated
assessments (e.g., self-reported mood states, saliva samples,
neurobiological measurements) from the same individual in
order to capture intra-individual variation in these processes
[13], which we argue is critical for understanding the con-
tribution of ovarian hormones in risk for STBs in adolescent
T. C. Ho et al.
females. Dense-sampling has the additional advantage of
being able to identify proximal factors that lead to suicidal
states and other related high-risk events, which are often
transient and rarely captured in the laboratory or clinic
[160].
Other important study design considerations include larger
sample sizes of both sexes in order to detect sex differences
more reliably. As we alluded to in the earlier sections of our
review, there are other important confounds to consider that
will affect the reliability and validity of estimating the effects
of within-person changes in sex hormones on brain and
behavior. These confounds include genetic factors, environ-
mental inuences, medical conditions, and cycle length (given
that the range of menstrual cycles and periods of anovulation
are longer in adolescents than in adults, particularly in the rst
few years post-menarche [161]). Finally, understanding the
normative associations between changes in hormones with the
structural and functional development of the brain is a critical
next step that must be established before understanding how
these alterations are altered in individuals at risk for STBs.
Gaining a more comprehensive understanding of neurobio-
logical trajectories and the mechanisms of neuroplasticity in
adolescents as a consequence of puberty will also contribute
to our knowledge of sex-specic psychiatric interventions and
may help to address the heterogeneity of risk factors identied
to date [5]. For instance, the granularity of data obtained from
dense-sampling methods will facilitate the identication of
individuals with hormonal sensitivity to the typical uctua-
tions occurring during puberty, as well as characterize within-
person pubertal processes associated with responses to social
stressors and poorer impulse control during heightened
emotional conict. With this information, researchers and
clinicians will be able to stratify individuals on the basis of
neuroendocrine risk (e.g., those with hormone sensitivity) and
also to identify points in time (e.g., dramatic increases in
stress perception or depressive symptoms, unusual variability
in DHEA) during which an individual might benetfrom
immediate intervention.
Summary and conclusions
Globally, suicide has surpassed all physical diseases as a
cause of death in adolescents. Puberty drives psychobiolo-
gical changes in adolescence that have not been examined
explicitly in relation to suicide risk. In this review, we
conceptualized suicidal thoughts and behaviors in adoles-
cents as resulting from pathological responses to social
stressors at a time when stress-regulatory systems are still
maturing. We argued that alterations in brain circuits
comprised of connections among the amygdala, hippo-
campus, striatum, ACC, vmPFC, and OFCthat underlie
social cognition, emotion regulation, and impulse control
and are shaped by puberty-related changes in sex hormones.
We propose that alterations in these circuits may partially
explain the ways in which changes in sex hormones are
linked with increased suicidal thoughts and behaviors dur-
ing adolescence. However, we also highlighted critical
moderators to be considered in this model, including a
neurobiological sensitivity to uctuations in ovarian hor-
mones, exposure to early adversity, and underlying mental
disorders (Fig. 2). To date, these specic hypotheses have
not been tested. Although there is emerging research iden-
tifying shared and unique neural circuits that underlie sui-
cidal thoughts and behaviors [68], it is paramount that that
these associations be examined in prospective studies. It
will also be important to use both experimental designs and
large-scale longitudinal studies to elucidate the extent to
which pubertal hormones affect the acute functioning of
these circuits and drive the development of these circuits
over the course of adolescence and young adulthood.
Moreover, we expect that although pubertal hormones are
not a primary driver of suicide risk, they may play an
outsized role in individuals with a neurobiological sensi-
tivity to hormonal uctuations. Therefore, it is critical that
we identify key moderators of the paths in our model, which
we hypothesize includes neurobiological sensitivity to
hormonal uctuations, experiences of adversity and life
Fig. 2 Conceptual model linking aspects of the pubertal transition
with risk for suicidal thoughts and behaviors. Experiences of early
adversity affect the programming and development of endocrine and
neural systems which undergo signicant maturation during puberty.
Puberty-related changes in ovarian, gonadal, and other related hor-
mones shape the neural circuits underlying social cognition, emotion
regulation, and impulse control (which include structures such as the
amygdala, hippocampus, striatum, anterior cingulate cortex, and por-
tions of prefrontal cortex). Alterations in these circuits may partially
explain the ways in which changes in sex hormones are linked with the
emergence of suicidal thoughts and behaviors during adolescence.
Moderators of these processes, including a neurobiological sensitivity
to ovarian hormones, experience of ongoing life stressors, and
underlying mental disorders, are highlighted in red.
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
stress that inuence neurodevelopmental trajectories
(which, in turn, may result in generating and experiencing
additional stress), and underlying mental disorders. In
addition to conducting longitudinal studies with larger
sample sizes, we suggest that researchers also use dense-
sampling methods to identify individuals according to these
stratication parameters (e.g., hormonal sensitivity), as well
as points in time at which individuals may be at risk and
could benet from more immediate intervention. In con-
clusion, we want to emphasize that increasing our under-
standing of pubertal science across endocrine, neural, and
psychosocial domains will yield signicant insights con-
cerning how best to reduce the frequency of suicide-related
deaths during the vulnerable period of adolescence.
Acknowledgements This work was supported by the National Institute
of Mental Health (R37MH101495 to IHG, K01MH117442 to TCH),
the Fonds de Recherche du QuébecSanté (FRQS/MSSS Resident
Physician Health Research Career Training Program to AJG), Preci-
sion Health and Integrated Diagnostics Center at Stanford (PHIND to
IHG and TCH), and the Ray and Dagmar Dolby Family Fund (to
TCH). The content is solely the responsibility of the authors and does
not necessarily represent the ofcial views of the National Institutes of
Health. All authors report no biomedical conicts of interest. The
funding agencies played no role in the design and conduct of the study;
collection, management, analysis, and interpretation of the data; and
preparation, review, or approval of the manuscript.
Compliance with ethical standards
Conict of interest The authors declare no competing interests.
Publishers note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as
long as you give appropriate credit to the original author(s) and the
source, provide a link to the Creative Commons license, and indicate if
changes were made. The images or other third party material in this
article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not
included in the articles Creative Commons license and your intended
use is not permitted by statutory regulation or exceeds the permitted
use, you will need to obtain permission directly from the copyright
holder. To view a copy of this license, visit http://creativecommons.
org/licenses/by/4.0/.
References
1. Mokdad AH, Forouzanfar MH, Daoud F, Mokdad AA, El
Bcheraoui C, Moradi-Lakeh M, et al. Global burden of diseases,
injuries, and risk factors for young peoples health during
19902013: a systematic analysis for the Global Burden of
Disease Study 2013. Lancet. 2016;387:2383401.
2. Murphy SL, Mathews TJ, Martin JA, Minkovitz CS, Strobino
DM Annual Summary of Vital Statistics: 20132014. Pediatrics
[Internet]. 2017 [cited 2020 Apr 10];139. Available from: https://
pediatrics.aappublications.org/content/139/6/e20163239
3. Curtin SC, Heron M. Death rates due to suicide and homicide
among persons aged 1024: United States, 20002017. NCHS
Data Brief. 2019;352:18.
4. Carter G, Milner A, McGill K, Pirkis J, Kapur N, Spittal MJ.
Predicting suicidal behaviours using clinical instruments: Sys-
tematic review and meta-analysis of positive predictive values
for risk scales. Br J Psychiatry. 2017;210:38795.
5. Franklin JC, Ribeiro JD, Fox KR, Bentley KH, Kleiman EM,
Huang X, et al. Risk factors for suicidal thoughts and behaviors: a
meta-analysis of 50 years of research. Psychol Bull. 2017;
143:187232.
6. Cha CB, Franz PJ, Guzmán EM, Glenn CR, Kleiman EM, Nock
MK. Annual research review: suicide among youth epide-
miology, (potential) etiology, and treatment. J Child Psychol
Psychiatry. 2018;59:46082.
7. Glenn CR, Kleiman EM, Kellerman J, Pollak O, Cha CB,
Esposito EC, et al. Annual research review: a meta-analytic
review of worldwide suicide rates in adolescents. J Child Psychol
Psychiatry. 2020;61:294308.
8. Nelson EE, Leibenluft E, McClure EB, Pine DS. The social re-
orientation of adolescence: a neuroscience perspective on the
process and its relation to psychopathology. Psychol Med.
2005;35:16374.
9. Paus T, Keshavan M, Giedd JN. Why do many psychiatric
disorders emerge during adolescence? Nat Rev Neurosci. 2008;
9:94757.
10. Rudolph KD Puberty as a Developmental Context of Risk for
Psychopathology. In: Lewis M, Rudolph KD, editors. Handbook
of Developmental Psychopathology. Boston, MA: Springer US;
2014. p. 33154.
11. OConnor RC, Nock MK. The psychology of suicidal behaviour.
Lancet Psychiatry. 2014;1:7385.
12. Lutz P-E, Mechawar N, Turecki G. Neuropathology of suicide:
recent ndings and future directions. Mol Psychiatry.
2017;22:1395412.
13. Miller AB, Prinstein MJ. Adolescent suicide as a failure
of acute stress-response systems. Annu Rev Clin Psychol.
2019;07:42550.
14. Owens SA, Eisenlohr-Moul T. Suicide risk and the menstrual
cycle: a review of candidate rdoc mechanisms. Curr Psychiatry
Rep. 2018;20:106.
15. King CA, Merchant CR. Social and interpersonal factors relating
to adolescent suicidality: a review of the literature. Arch Suicide
Res. 2008;12:18196.
16. Liu RT, Miller I. Life events and suicidal ideation and behavior:
a systematic review. Clin Psychol Rev. 2014;34:18192.
17. Stewart JG, Shields GS, Esposito EC, Cosby EA, Allen NB,
Slavich GM, et al. Life stress and suicide in adolescents. J
Abnorm Child Psychol. 2019;47:170722.
18. Platt B, Cohen Kadosh K, Lau JYF. The role of peer rejection in
adolescent depression. Depress Anxiety. 2013;30:80921.
19. Prinstein MJ, Giletta M Peer relations and developmental psy-
chopathology. In: Developmental psychopathology: theory and
method, Vol 1, 3rd ed. Hoboken, NJ, US: John Wiley & Sons
Inc; 2016. p. 52779.
20. Whiteside SP, Lynam DR, Miller JD, Reynolds SK. Validation
of the UPPS impulsive behaviour scale: a four-factor model of
impulsivity. Eur J Personal. 2005;19:55974.
21. Eisenlohr-Moul TA, Miller AB, Giletta M, Hastings PD,
Rudolph KD, Nock MK, et al. HPA axis response and psycho-
social stress as interactive predictors of suicidal ideation and
behavior in adolescent females: a multilevel diathesis-stress
framework. Neuropsychopharmacology 2018;43:256471.
T. C. Ho et al.
22. van Geel M, Vedder P, Tanilon J. Relationship between peer
victimization, cyberbullying, and suicide in children and ado-
lescents: a meta-analysis. JAMA Pediatr. 2014;168:43542.
23. Auerbach RP, Stewart JG, Johnson SL. Impulsivity and
suicidality in adolescent inpatients. J Abnorm Child Psychol.
2017;45:91103.
24. Mahfouz A, Lelieveldt BPF, Grefhorst A, van Weert LTCM,
Mol IM, Sips HCM. et al. Genome-wide coexpression of steroid
receptors in the mouse brain: identifying signaling pathways and
functionally coordinated regions. Proc Natl Acad Sci USA.
2016;113:273843.
25. McEwen BS, Milner TA. Understanding the broad inuence of
sex hormones and sex differences in the brain. J Neurosci Res.
2017;95:2439.
26. Kaczkurkin AN, Raznahan A, Satterthwaite TD. Sex differences
in the developing brain: insights from multimodal neuroimaging.
Neuropsychopharmacology 2019;44:7185.
27. Scheinost D, Finn ES, Tokoglu F, Shen X, Papademetris X,
Hampson M, et al. Sex differences in normal age trajectories of
functional brain networks. Hum Brain Mapp. 2015;36:152435.
28. Schmaal L, van Harmelen A-L, Chatzi V, Lippard ETC, Toen-
ders YJ, Averill LA, et al. Imaging suicidal thoughts and beha-
viors: a comprehensive review of 2 decades of neuroimaging
studies. Mol Psychiatry. 2020;25:40827.
29. Ho TC, Colich NL, Sisk LM, Oskirko K, Jo B, Gotlib IH. Sex
differences in the effects of gonadal hormones on white matter
microstructure development in adolescence. Dev Cogn Neurosci.
2020;42:100773.
30. Vijayakumar N, Op de Macks Z, Shirtcliff EA, Pfeifer JH.
Puberty and the human brain: Insights into adolescent develop-
ment. Neurosci Biobehav Rev. 2018;92:41736.
31. Reiter EO, Fuldauer VG, Root AW. Secretion of the adrenal
androgen, dehydroepiandrosterone sulfate, during normal infancy,
childhood, and adolescence, in sick infants, and in children with
endocrinologic abnormalities. J Pediatr. 1977;90:76670.
32. Søeborg T, Frederiksen H, Mouritsen A, Johannsen TH, Main
KM, Jørgensen N. et al. Sex, age, pubertal development and use
of oral contraceptives in relation to serum concentrations of
DHEA, DHEAS, 17α-hydroxyprogesterone, Δ4-androstene-
dione, testosterone and their ratios in children, adolescents and
young adults. Clin Chim Acta. 2014;437:613.
33. Dorn LD, Dahl RE, Woodward HR, Biro F. Dening the
boundaries of early adolescence: A users guide to assessing
pubertal status and pubertal timing in research with adolescents.
Appl Dev Sci. 2006;10:3056.
34. Schulz KM, Sisk CL. The organizing actions of adolescent
gonadal steroid hormones on brain and behavioral development.
Neurosci Biobehav Rev. 2016;70:14858.
35. Piekarski DJ, Johnson CM, Boivin JR, Thomas AW, Lin WC,
Delevich K, et al. Does puberty mark a transition in sensitive
periods for plasticity in the associative neocortex? Brain Res.
2017;1654:12344.
36. Juraska JM, Willing J. Pubertal onset as a critical transition for
neural development and cognition. Brain Res. 2017;1654:8794.
37. Saunders KEA, Hawton K. Suicidal behaviour and the menstrual
cycle. Psychol Med. 2006;36:90112.
38. Osborn E, Brooks J, OBrien PMS, Wittkowski A Suicidality in
women with premenstrual dysphoric disorder: a systematic lit-
erature review. Arch Womens Ment Health [Internet]. 2020 Sep
16 [cited 2021 Mar 13]; Available from: https://doi.org/10.1007/
s00737-020-01054-8
39. Shams-Alizadeh N, MarouA, Rashidi M, Roshani D, Farha-
difar F, Khazaie H. Premenstrual dysphoric disorder and suicide
attempts as a correlation among women in reproductive age.
Asian J Psychiatry. 2018;31:636.
40. Wei S-M, Schiller CE, Schmidt PJ, Rubinow DR. The role of
ovarian steroids in affective disorders. Curr Opin Behav
Sci.2018;23:10312.
41. Schmidt PJ, Martinez PE, Nieman LK, Koziol DE, Thompson KD,
Schenkel L, et al. Exposure to a change in ovarian steroid levels
but not continuous stable levels triggers PMDD symptoms fol-
lowing ovarian suppression. Am J Psychiatry. 2017;174:9809.
42. Dogra TD, Leenaars AA, Raintji R, Lalwani S, Girdhar S,
Wenckstern S, et al. Menstruation and suicide: an exploratory
study. Psychol Rep. 2007;101:4304.
43. Zengin Y, Çalık I, Büyükcam F, Şen J, Akpınar Ş, Erdem A,
et al. The relationship between suicide attempts and menstrual
cycles in the emergency department and the sociodemographic
and clinical characteristics of these patients. Eurasia J Emerg
Med. 2015;14:11822.
44. Afzali S, Taheri SK, Jamilian M, Eslambolchi P. The relation-
ship between menstrual cycle phases and suicide attempts in
suicidal women admitted to the poisoning ward of Farshchian
Hospital, Hamedan, Iran. Iran J Toxicol. 2012;5:5314.
45. Baca-Garcia E, Diaz-Sastre C, Ceverino A, Perez-Rodriguez
MM, Navarro-Jimenez R, Lopez-Castroman J, et al. Suicide
attempts among women during low estradiol/low progesterone
states. J Psychiatr Res. 2010;44:20914.
46. Kim B, Kang E-S, Fava M, Mischoulon D, Soskin D, Yu B-H,
et al. Follicle-stimulating hormone (FSH), current suicidal
ideation and attempt in female patients with major depressive
disorder. Psychiatry Res. 2013;210:9516.
47. Sun BZ, Kangarloo T, Adams JM, Sluss PM, Welt CK, Chandler
DW, et al. Healthy post-menarchal adolescent girls demonstrate
multi-level reproductive axis immaturity. J Clin Endocrinol
Metab. 2018;104:61323.
48. Chatterton RT, Mateo ET, Hou N, Rademaker AW, Acharya S,
Jordan VC, et al. Characteristics of salivary proles of oestradiol
and progesterone in premenopausal women. J Endocrinol.
2005;186:7784.
49. Gann PH, Giovanazzi S, Van Horn L, Branning A, Chatterton
RT.Saliva as a medium for investigating intra- and inter-
individual differences in sex hormone levels in premenopausal
women.Cancer Epidemiol Biomark Prev. 2001;10:5964.
50. Landgren B-M, Undén A-L, Diczfalusy E. Hormonal prole of
the cycle in 68 normally menstruating women. Eur J Endocrinol.
1980;94:8998.
51. Shirtcliff EA, Dahl RE, Pollak SD. Pubertal development: cor-
respondence between hormonal and physical development. Child
Dev. 2009;80:32737.
52. Hambridge HL, Mumford SL, Mattison DR, Ye A, Pollack AZ,
Bloom MS, et al. The inuence of sporadic anovulation on
hormone levels in ovulatory cycles. Hum Reprod Oxf Engl.
2013;28:168794.
53. Ellison PT, Panter-Brick C, Lipson SF, ORourke MT. The
ecological context of human ovarian function. Hum Reprod Oxf
Engl. 1993;8:224858.
54. Lobo RA, Granger L, Goebelsmann U, Mishell DR. Elevations
in unbound serum estradiol as a possible mechanism for inap-
propriate gonadotropin secretion in women with PCO. J Clin
Endocrinol Metab. 1981;52:1568.
55. Cover KK, Maeng LY, Lebrón-Milad K, Milad MR. Mechan-
isms of estradiol in fear circuitry: implications for sex differences
in psychopathology. Transl Psychiatry. 2014;4:e422.
56. Maner JK, Miller SL. Hormones and social monitoring: men-
strual cycle shifts in progesterone underlie womens sensitivity
to social information. Evol Hum Behav. 2014;35:916.
57. Sheppard PAS, Choleris E, Galea LAM. Structural plasticity of
the hippocampus in response to estrogens in female rodents. Mol
Brain. 2019;12:22.
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
58. Wang ACJ, Hara Y, Janssen WGM, Rapp PR, Morrison JH.
Synaptic estrogen receptor-αlevels in prefrontal cortex in female
rhesus monkeys and their correlation with cognitive performance. J
Neurosci. 2010;30:127706.
59. Engman J, Sundström Poromaa I, Moby L, Wikström J, Fredrikson
M, Gingnell M. Hormonal cycle and contraceptive effects on
amygdala and salience resting-state networks in women with pre-
vious affective side effects on the pill. Neuropsychopharmacology.
2018;43:55563.
60. Engman J, Linnman C, Van Dijk KRA, Milad MR. Amygdala
subnuclei resting-state functional connectivity sex and estrogen
differences. Psychoneuroendocrinology. 2016;63:3442.
61. Miedl SF, Wegerer M, Kerschbaum H, Blechert J, Wilhelm FH.
Neural activity during traumatic lm viewing is linked to
endogenous estradiol and hormonal contraception. Psychoneur-
oendocrinology. 2018;87:206.
62. Protopopescu X, Butler T, Pan H, Root J, Altemus M, Pola-
necsky M, et al. Hippocampal structural changes across the
menstrual cycle. Hippocampus. 2008;18:9858.
63. Lisofsky N, Mårtensson J, Eckert A, Lindenberger U, Gallinat J,
Kühn S. Hippocampal volume and functional connectivity changes
during the female menstrual cycle. NeuroImage. 2015;118:15462.
64. Pletzer B, Harris T-A, Scheuringer A, Hidalgo-Lopez E. The
cycling brain: menstrual cycle related uctuations in hippo-
campal and fronto-striatal activation and connectivity during
cognitive tasks. Neuropsychopharmacology. 2019;44:186775.
65. Albert K, Pruessner J, Newhouse P. Estradiol levels modulate
brain activity and negative responses to psychosocial stress
across the menstrual cycle. Psychoneuroendocrinology. 2015;
59:1424.
66. Arélin K, Mueller K, Barth C, Rekkas PV, Kratzsch J, Burmann I,
et al. Progesterone mediates brain functional connectivity changes
during the menstrual cyclea pilot resting state MRI study. Front
Neurosci [Internet]. 2015 Feb 23 [cited 2020 Apr 11];9. Available
from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4337344/
67. Pritschet L, Santander T, Layher E, Taylor CM, Yu S, Miller
MB, et al. Functional reorganization of brain networks across the
human menstrual cycle. bioRxiv. 2019 Dec 6;866913.
68. Auerbach RP, Pagliaccio D, Allison GO, Alqueza KL, Alonso
MF. Neural correlates associated with suicide and nonsuicidal
self-injury in youth. Biol Psychiatry. 2020;89:11933.
69. Rybaczyk LA, Bashaw MJ, Pathak DR, Moody SM, Gilders
RM, Holzschu DL. An overlooked connection: serotonergic
mediation of estrogen-related physiology and pathology. BMC
Womens Health. 2005;5:12.
70. Sullivan GM, Oquendo MA, Milak M, Miller JM, Burke A,
Ogden RT, et al. Positron emission tomography quantication of
serotonin(1 A) receptor binding in suicide attempters with major
depressive disorder. JAMA Psychiatry. 2015;72:16978.
71. Oquendo MA, Galfalvy H, Sullivan GM, Miller JM, Milak MM,
Sublette ME, et al. Positron emission tomographic imaging of the
serotonergic system and prediction of risk and lethality of future
suicidal behavior. JAMA Psychiatry. 2016;73:104855.
72. Mann JJ The serotonergic system in mood disorders and suicidal
behaviour. Philos Trans R Soc B Biol Sci [Internet]. 2013 [cited
2020 Apr 11];368. Available from: https://www.ncbi.nlm.nih.
gov/pmc/articles/PMC3638390/
73. Steinberg LJ, Rubin-Falcone H, Galfalvy HC, Kaufman J, Miller
JM, Sublette ME, et al. Cortisol stress response and in vivo PET
imaging of human brain serotonin 1A receptor binding. Int J
Neuropsychopharmacol. 2019;22:32938.
74. Underwood MD, Kassir SA, Bakalian MJ, Galfalvy H, Dwork
AJ, Mann JJ, et al. Serotonin receptors and suicide, major
depression, alcohol use disorder and reported early life adversity.
Transl Psychiatry. 2018;8:115.
75. Mellon SH, Grifn LD. Neurosteroids: biochemistry and clinical
signicance. Trends Endocrinol Metab TEM. 2002;13:3543.
76. Giordano R, Picu A, Bonelli L, Balbo M, Berardelli R,
Marinazzo E, et al. Hypothalamus-pituitary-adrenal axis eva-
luation in patients with hypothalamo-pituitary disorders: com-
parison of different provocative tests. Clin Endocrinol (Oxf).
2008;68:93541.
77. Gunn JF, Lester D, McSwain S. Testing the warning signs of
suicidal behavior among suicide ideators using the 2009 National
survey on drug abuse and health. Int J Emerg Ment Health.
2011;13:14754.
78. Skilbeck KJ, Johnston GAR, Hinton T. Stress and GABAA
receptors. J Neurochem. 2010;112:111530.
79. Hu W, Zhang M, Czéh B, Flügge G, Zhang W. Stress impairs
GABAergic network function in the hippocampus by activat-
ing nongenomic glucocorticoid receptors and affecting the
integrity of the parvalbumin-expressing neuronal network.
Neuropsychopharmacology 2010;35:1693707.
80. Zhao J, Verwer RWH, Gao S-F, Qi X-R, Lucassen PJ, Kessels
HW, et al. Prefrontal alterations in GABAergic and glutamater-
gic gene expression in relation to depression and suicide. J
Psychiatr Res. 2018;102:26174.
81. Archer J. Testosterone and human aggression: an evaluation of the
challenge hypothesis. Neurosci Biobehav Rev. 2006;30:
31945.
82. van Anders SM. Beyond masculinity: testosterone, gender/sex,
and human social behavior in a comparative context. Front
Neuroendocrinol. 2013;34:198210.
83. Halpern CT, Udry JR, Campbell B, Suchindran C. Relationships
between aggression and pubertal increases in testosterone: a
panel analysis of adolescent males. Soc Biol. 1993;40:824.
84. Rice TR, Sher L Adolescent suicide and testosterone: Postnatal
testosterone may be an important mediator of the association
between prematurity and male neurodevelopmental disorders: A
Hypothesis. Int J Adolesc Med Health [Internet]. 2015 Nov 4
[cited 2020 Apr 11];29. Available from: https://www.degruyter.
com/view/journals/ijamh/29/4/article-20150058.xml
85. Perez-Rodriguez MM, Lopez-Castroman J, Martinez-Vigo M, Diaz-
Sastre C, Ceverino A, Núñez-Beltrán A, et al. Lack of association
between testosterone and suicide attempts. Neuropsychobiology.
2011;63:12530.
86. McGirr A, Turecki G. The relationship of impulsive aggres-
siveness to suicidality and other depression-linked behaviors.
Curr Psychiatry Rep. 2007;9:4606.
87. Eisenegger C, Haushofer J, Fehr E. The role of testosterone in
social interaction. Trends Cogn Sci. 2011;15:26371.
88. Duke SA, Balzer BWR, Steinbeck KS. Testosterone and its
effects on human male adolescent mood and behavior: a sys-
tematic review. J Adolesc Health. 2014;55:31522.
89. van Bokhoven I, van Goozen SHM, van Engeland H, Schaal B,
Arseneault L, Séguin JR, et al. Salivary testosterone and
aggression, delinquency, and social dominance in a population-
based longitudinal study of adolescent males. Horm Behav.
2006;50:11825.
90. Schaal B, Tremblay RE, Soussignan R, Susman EJ. Male
testosterone linked to high social dominance but low physical
aggression in early adolescence. J Am Acad Child Adolesc
Psychiatry. 1996;35:132230.
91. Milner A, Page A, Morrell S, Hobbs C, Carter G, Dudley M,
et al. The effects of involuntary job loss on suicide and suicide
attempts among young adults: evidence from a matched case-
control study. Aust N Z J Psychiatry. 2014;48:33340.
92. Stefansson J, Chatzittos A, Nordström P, Arver S, Åsberg M,
Jokinen J. CSF and plasma testosterone in attempted suicide.
Psychoneuroendocrinology 2016;74:16.
T. C. Ho et al.
93. Gustavsson G, Träskman-Bendz L, Higley JD, Westrin Å. CSF
testosterone in 43 male suicide attempters. Eur Neuropsycho-
pharmacol. 2003;13:1059.
94. Zhang J, Jia C-X, Wang L-L. Testosterone differs between sui-
cide attempters and community controls in men and women of
China. Physiol Behav. 2015;141:405.
95. Roland BC, Morris JL, Zelhart PF. Proposed relation of testos-
terone levels to male suicides and sudden deaths. Psychol Rep.
1986;59:1002.
96. Sher L, Grunebaum MF, Sullivan GM, Burke AK, Cooper TB,
Mann JJ, et al. Testosterone levels in suicide attempters with
bipolar disorder. J Psychiatr Res. 2012;46:126771.
97. Sher L, Grunebaum MF, Sullivan GM, Burke AK, Cooper TB,
Mann JJ, et al. Association of testosterone levels and future
suicide attempts in females with bipolar disorder. J Affect Dis-
ord. 2014;166:98102.
98. Tripodianakis J, Markianos M, Rouvali O, Istikoglou C. Gonadal
axis hormones in psychiatric male patients after a suicide
attempt. Eur Arch Psychiatry Clin Neurosci. 2007;257:1359.
99. Markianos M, Tripodianakis J, Istikoglou C, Rouvali O, Christo-
poulos M, Papageorgopoulos P, et al. Suicide attempt by jumping:
a study of gonadal axis hormones in male suicide attempters versus
men who fell by accident. Psychiatry Res. 2009;170:825.
100. Shay DA, Vieira-Potter VJ, Rosenfeld CS Sexually Dimorphic
Effects of Aromatase on Neurobehavioral Responses. Front Mol
Neurosci [Internet]. 2018 [cited 2020 Apr 17];11. Available
from: https://www.frontiersin.org/articles/10.3389/fnmol.2018.
00374/full
101. Fitzgerald ML, Kassir SA, Underwood MD, Bakalian MJ, Mann JJ,
Arango V. Dysregulation of Striatal Dopamine Receptor Binding in
Suicide. Neuropsychopharmacology. 2017;42:97482.
102. Pitchot W, Hansenne M, Ansseau M. Role of dopamine in non-
depressed patients with a history of suicide attempts. Eur Psy-
chiatry. 2001;16:4247.
103. Roy A, Karoum F, Pollack S. Marked reduction in indexes of
dopamine metabolism among patients with depression who
attempt suicide. Arch Gen Psychiatry. 1992;49:44750.
104. Tobiansky DJ, Wallin-Miller KG, Floresco SB, Wood RI, Soma
KK Androgen Regulation of the Mesocorticolimbic System and
Executive Function. Front Endocrinol [Internet]. 2018 [cited
2020 Apr 14];9. Available from: https://www.frontiersin.org/a
rticles/10.3389/fendo.2018.00279/full
105. Sinclair D, Purves-Tyson TD, Allen KM, Weickert CS. Impacts
of stress and sex hormones on dopamine neurotransmission in
the adolescent brain. Psychopharmacol. 2014;231:158199.
106. Wahlstrom D, Collins P, White T, Luciana M. Developmental
changes in dopamine neurotransmission in adolescence: beha-
vioral implications and issues in assessment. Brain Cogn.
2010;72:14659.
107. Pitchot W, Hansenne M, Moreno AG, Ansseau M. Dopamine and
history of suicide attempts. Clin Neuropharmacol. 1992;15:296B.
108. Happe S, Tings T, Koch W, Welsch J, Helmschmied K, Baier
PC, et al. Growth hormone response in low-dose apomorphine
test correlates with nigrostriatal dopamine transporter binding in
patients with Parkinsons disease. J Neural Transm Vienna
Austria 1996. 2007;114:58994.
109. Oquendo MA, Sullivan GM, Sudol K, Baca-Garcia E, Stanley
BH, Sublette ME, et al. Toward a Biosignature for Suicide. Am J
Psychiatry. 2014;171:125977.
110. Barendse MEA, Simmons JG, Byrne ML, Seal ML, Patton G,
Mundy L, et al. Brain structural connectivity during adrenarche:
associations between hormone levels and white matter micro-
structure. Psychoneuroendocrinology. 2018;88:707.
111. Menzies L, Goddings A-L, Whitaker KJ, Blakemore S-J, Viner
RM. The effects of puberty on white matter development in
boys. Dev Cogn Neurosci. 2015;11:11628.
112. Peper JS, Reus MA, de, Heuvel MP, van den, Schutter DJLG.
Short fused? associations between white matter connections, sex
steroids, and aggression across adolescence. Hum Brain Mapp.
2015;36:104352.
113. Bos PA, van Honk J, Ramsey NF, Stein DJ, Hermans EJ.
Testosterone administration in women increases amygdala
responses to fearful and happy faces. Psychoneur-
oendocrinology. 2013;38:80817.
114. Spielberg JM, Olino TM, Forbes EE, Dahl RE. Exciting fear in
adolescence: does pubertal development alter threat processing?
Dev Cogn Neurosci. 2014;8:8695.
115. Cservenka A, Stroup ML, Etkin A, Nagel BJ. The effects of age,
sex, and hormones on emotional conict-related brain response
during adolescence. Brain Cogn. 2015;99:13550.
116. Op de Macks ZA, Moor BG, Overgaauw S, Güroğlu B, Dahl RE,
Crone EA. Testosterone levels correspond with increased ventral
striatum activation in response to monetary rewards in adoles-
cents. Dev Cogn Neurosci. 2011;1:50616.
117. Laube C, van den Bos W, Fandakova Y. The relationship between
pubertal hormones and brain plasticity: Implications for cognitive
training in adolescence. Dev Cogn Neurosci. 2020;42:100753.
118. Braams BR, van Duijvenvoorde ACK, Peper JS, Crone EA.
Longitudinal changes in adolescent risk-taking: a comprehensive
study of neural responses to rewards, pubertal development, and
risk-taking behavior. J Neurosci J Soc Neurosci. 2015;35:722638.
119. White SF, Lee Y, Schlund MW, Shirtcliff EA, Ladouceur CD.
Testosterone reactivity is associated with reduced neural
response to reward in early adolescence. Behav Brain Res. 2020;
387:112593.
120. Coates JM, Herbert J. Endogenous steroids and nancial risk
taking on a London trading oor. Proc Natl Acad Sci. 2008;
105:616772.
121. Stanton SJ, Liening SH, Schultheiss OC. Testosterone is posi-
tively associated with risk taking in the Iowa Gambling Task.
Horm Behav. 2011;59:2526.
122. Richard-Devantoy S, Berlim MT, Jollant F. A meta-analysis of
neuropsychological markers of vulnerability to suicidal behavior
in mood disorders. Psychol Med. 2014;44:166373.
123. Gifuni AJ, Perret LC, Lacourse E, Geoffroy M-C, Mbekou V,
Jollant F, et al. Decision-making and cognitive control in ado-
lescent suicidal behaviors: a qualitative systematic review of the
literature. Eur Child Adolesc Psychiatry [Internet]. 2020 May 9
[cited 2021 Mar 8]; Available from: https://doi.org/10.1007/
s00787-020-01550-3
124. Chatzittos A, Nordström P, Hellström C, Arver S, Åsberg M,
Jokinen J. CSF 5-HIAA, cortisol and DHEAS levels in suicide
attempters. Eur Neuropsychopharmacol. 2013;23:12807.
125. Buttereld MI, Stechuchak KM, Connor KM, Davidson JRT,
Wang C, MacKuen CL, et al. Neuroactive steroids and suicidality
in posttraumatic stress disorder. Am J Psychiatry. 2005;162:3802.
126. Sher L, Flory J, Bierer L, Makotkine I, Yehuda R. Dehy-
droepiandrosterone and dehydroepiandrosterone sulfate levels in
combat veterans with or without a history of suicide attempt.
Acta Psychiatr Scand. 2018;138:5561.
127. Sequeira A, Shen K, Gottlieb A, Limon A.Human brain tran-
scriptome analysis nds region- and subject-specic expression
signatures of GABA A R subunits. Commun Biol. 2019;2:114.
128. Pineles SL, Nillni YI, Pinna G, Irvine J, Webb A, Arditte Hall KA,
et al. PTSD in women is associated with a block in conversion of
progesterone to the GABAergic neurosteroids allopregnanolone
and pregnanolone measured in plasma. Psychoneuroendocrinology.
2018;93:13341.
129. Locci A, Pinna G. Neurosteroid biosynthesis downregulation
and changes in GABAA receptor subunit composition: a bio-
marker axis in stressinduced cognitive and emotional impair-
ment. Br J Pharm. 2017;174:322641.
Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of. . .
130. Ellis R, Fernandes A, Simmons JG, Mundy L, Patton G, Allen
NB, et al. Relationships between adrenarcheal hormones, hip-
pocampal volumes and depressive symptoms in children. Psy-
choneuroendocrinology. 2019;104:5563.
131. Barendse MEA, Simmons JG, Patton G, Mundy L, Byrne ML,
Seal ML, et al. Adrenarcheal timing longitudinally predicts
anxiety symptoms via amygdala connectivity during emotion
processing. J Am Acad Child Adolesc Psychiatry [Internet].
2019 May 2 [cited 2020 Apr 12]. Available from: https://www.
jaacap.org/article/S0890-8567(19)30286-2/abstract
132. Whittle S, Simmons JG, Byrne ML, Strikwerda-Brown C, Ker-
estes R, Seal ML, et al. Associations between early adrenarche,
affective brain function and mental health in children. Soc Cogn
Affect Neurosci. 2015;10:128290.
133. Sripada RK, Marx CE, King AP, Rajaram N, Garnkel SN,
Abelson JL, et al. DHEA enhances emotion regulation neuro-
circuits and modulates memory for emotional stimuli. Neu-
ropsychopharmacology. 2013;38:1798807.
134. McLoughlin AB, Gould MS, Malone KM. Global trends in
teenage suicide: 20032014. QJM Mon J Assoc Phys. 2015;
108:76580.
135. WHO | Preventing suicide: a global imperative [Internet]. WHO.
World Health Organization; [cited 2020 Mar 26]. Available from:
http://www.who.int/mental_health/suicide-prevention/world_report_
2014/en/
136. Ruch DA, Sheftall AH, Schlagbaum P, Rausch J, Campo JV,
Bridge JA.Trends in suicide among youth aged 10 to 19 years
in the United States, 1975 to 2016. JAMA Netw Open.2019;2:
e193886.
137. Lewinsohn PM, Rohde P, Seeley JR, Baldwin CL.
Gender differences in suicide attempts from adolescence to
young adulthood. J Am Acad Child Adolesc Psychiatry. 2001;
40:42734.
138. Boeninger DK, Masyn KE, Feldman BJ, Conger RD. Sex dif-
ferences in developmental trends of suicide ideation, plans, and
attempts among European American adolescents. Suicide Life
Threat Behav. 2010;40:45164.
139. Miranda-Mendizabal A, Castellví P, Parés-Badell O, Alayo I,
Almenara J, Alonso I, et al. Gender differences in suicidal
behavior in adolescents and young adults: systematic review and
meta-analysis of longitudinal studies. Int J Public Health. 2019;
64:26583.
140. Brent DA, Baugher M, Bridge J, Chen T, Chiappetta L. Age- and
sex-related risk factors for adolescent suicide. J Am Acad Child
Adolesc Psychiatry. 1999;38:1497505.
141. Dorn LD, Biro FM. Puberty and its measurement: a decade in
review. J Res Adolesc. 2011;21:18095.
142. Graber JA. Pubertal timing and the development of psychopathology
in adolescence and beyond. Horm Behav. 2013;64:2629.
143. Kaltiala-Heino R, Marttunen M, Rantanen P, Rimpelä M. Early
puberty is associated with mental health problems in middle
adolescence. Soc Sci Med. 2003;57:105564.
144. Lee D, Ahn I-Y, Park C-S, Kim B-J, Lee C-S, Cha B, et al.
Early menarche as a risk factor for suicidal ideation in girls: The
Korea youth risk behavior web-based survey. Psychiatry Res.
2020;285:112706.
145. Nacinovich R, Buzi F, Oggiano S, Rossi S, Spada S, Broggi F,
et al. Body experiences and psychopathology in idiopathic central
precocious and early puberty. Minerva Pediatr. 2016;68:118.
146. Stice E, Presnell K, Bearman SK. Relation of early menarche to
depression, eating disorders, substance abuse, and comorbid
psychopathology among adolescent girls. Dev Psychol. 2001;
37:60819.
147. Roberts E, Fraser A, Gunnell D, Joinson C, Mars B. Timing of
menarche and self-harm in adolescence and adulthood: a
population-based cohort study. Psychol Med. 2020;50:20108.
148. Belsky J, Steinberg LD, Houts RM, Friedman SL, DeHart G,
Cauffman E, et al. Family rearing antecedents of pubertal timing.
Child Dev. 2007;78:130221.
149. Turecki G, Ernst C, Jollant F, Labonté B, Mechawar N. The
neurodevelopmental origins of suicidal behavior. Trends Neu-
rosci. 2012;35:1423.
150. Wichstrøm L, Hegna K. Sexual orientation and suicide attempt:
A longitudinal study of the general Norwegian adolescent
population. J Abnorm Psychol. 2003;112:14451.
151. Klonsky ED, Saffer BY, Bryan CJ. Ideation-to-action theories of
suicide: a conceptual and empirical update. Curr Opin Psychol.
2018;22:3843.
152. Beauchaine TP, Crowell SE, Hsiao RC. Post-dexamethasone cor-
tisol, self-inicted injury, and suicidal ideation among depressed
adolescent girls. J Abnorm Child Psychol. 2015;43:61932.
153. Shalev A, Porta G, Biernesser C, Zelazny J, Walker-Payne M,
Melhem N, et al. Cortisol response to stress as a predictor for
suicidal ideation in youth. J Affect Disord. 2019;257:106.
154. King LS, Graber MG, Colich NL, Gotlib IH. Associations of
waking cortisol with DHEA and testosterone across the pubertal
transition: effects of threat-related early life stress. Psychoneur-
oendocrinology. 2020;115:104651.
155. Maninger N, Wolkowitz OM, Reus VI, Epel ES, Mellon SH.
Neurobiological and neuropsychiatric effects of dehydroepian-
drosterone (DHEA) and DHEA sulfate (DHEAS). Front Neu-
roendocrinol. 2009;30:6591.
156. Oyola MG, Handa RJ. Hypothalamic-pituitary-adrenal and
hypothalamic-pituitary-gonadal axes: sex differences in regula-
tion of stress responsivity. Stress Amst Neth 2017;20:47694.
157. Toufexis D, Rivarola MA, Lara H, Viau V. Stress and the
reproductive axis. J Neuroendocrinol. 2014;26:57386.
158. Mastorakos G, Pavlatou MG, Mizamtsidi M. The hypothalamic-
pituitary-adrenal and the hypothalamic- pituitary-gonadal axes
interplay. Pediatr Endocrinol Rev. 2006;3:17281.
159. Mendle J, Beltz AM, Carter R, Dorn LD. Understanding puberty
and its measurement: ideas for research in a new generation. J
Res Adolesc. 2019;29:8295.
160. Kleiman EM, Turner BJ, Fedor S, Beale EE, Picard RW, Huff-
man JC, et al. Digital phenotyping of suicidal thoughts. Depress
Anxiety. 2018;35:6018.
161. Roseneld RL. Adolescent anovulation: maturational mechanisms
and implications. J Clin Endocrinol Metab. 2013;98:357283.
162. Østby Y, Tamnes CK, Fjell AM, Westlye LT, Due-Tønnessen P,
Walhovd KB. Heterogeneity in subcortical brain development: a
structural magnetic resonance imaging study of brain maturation
from 8 to 30 years. J Neurosci. 2009;29:1177282.
T. C. Ho et al.
... Adolescence is a period of vulnerability that increases the likelihood of developing STB, which can persist into adulthood and become chronic, hindering positive personal development. The adolescence phase is normally characterized by significant changes in the brain and body, including hormonal fluctuations associated with puberty, which impact mood, impulse control, and adequate decision-making abilities (12). Heightened impulsivity, quick mood changes and difficulties rationalizing risks and consequences, are all risk factor that render adolescents more vulnerable to STB (13,14). ...
... There are some physical and biological evidences, on these risks. Brain imaging shows that group differences in surface area in the prefrontal, temporal and parietal regions, as well as in the volume of several subcortical nuclei are associated with the occurrence of suicide attempts in depressed adolescents (12,15). Additionally, distinct patterns of serotonergic abnormalities, disruptions in the hypothalamicpituitary-adrenal (HPA) axis, and irregularities in growth hormone (GH) secretion are associated with suicidal behaviors in adolescents (12). ...
... Brain imaging shows that group differences in surface area in the prefrontal, temporal and parietal regions, as well as in the volume of several subcortical nuclei are associated with the occurrence of suicide attempts in depressed adolescents (12,15). Additionally, distinct patterns of serotonergic abnormalities, disruptions in the hypothalamicpituitary-adrenal (HPA) axis, and irregularities in growth hormone (GH) secretion are associated with suicidal behaviors in adolescents (12). Exposure to suicidal behavior, particularly within the family context, and childhood adversities like abuse, neglect, and family conflict also increase the risk of STB (16). ...
Article
Full-text available
Introduction Despite the extensive implementation of suicide prevention strategies targeting suicidal thoughts and behaviors (STB) in adolescents, there remains a concerning lack of improvement in the situation. In this comprehensive scoping review, our objective was to provide insights into prevention methods for suicidal thoughts and behaviors directed towards adolescents, including their effectiveness, public perception, and potential adaptations. Method A scoping review was conducted, encompassing 71 articles including systematic review, clinical trials and qualitative studies for a wider understanding. Most articles included focus generally on adolescents aged 10–20. Results No single intervention has shown expected effectiveness, collective efforts have laid a solid foundation for suicide prevention. Promising interventions include cognitive-behavioral therapy (CBT) and incorporating Technology-based interventions. However, challenges persist in promoting help-seeking behaviors and addressing barriers such as stigma, the natural impulsive nature of adolescents and difficulty in selecting and defining data and designs. Discussion This review underscores the need for a holistic approach to suicide prevention, integrating social, emotional, and psychological dimensions. Successful interventions target underlying issues like depression and loneliness rather than solely focusing on suicidal thoughts and behaviors (STB). Combining direct and indirect interventions is a sensible approach for both immediate and long-term results. Understanding Generation Z's unique needs, influenced by technology and diverse perspectives, is crucial for effective prevention. Conclusion Involving adolescents and adopting patient-centered healthcare with outcome measures like Patient Perceived Outcome Measures can enhance suicide prevention efforts by prioritizing safety and patient experiences.
... Although existing theories incorporate important aspects of suicide vulnerability (e.g., hopelessness, perceived burdensomeness, thwarted belongingness, pain, acquired capability) that are applicable across the lifespan, studies testing these models have not typically included factors that strongly shape interpersonal relationships during adolescence, most notably pubertal development. Puberty is a key biological process of adolescent development that involves brain development, which can increase sensitivity to social stress [119] and STB risk [120]. Interpersonal reactions to physical changes during puberty also are of key importance, especially in explaining gender differences in STB. ...
Article
Full-text available
Suicide is a leading cause of death among adolescents, and recent suicide theories have sought to clarify the factors that facilitate the transition from suicide ideation to action. Specifically, the Interpersonal Theory of Suicide (IPTS), Integrated Motivational-Volitional Model (IMV), and Three Step Theory (3ST) have highlighted risk factors central to the formation of suicidal ideation and suicidal behaviors, which is necessary for suicide death. However, these models were initially developed and tested among adults, and given core socioemotional and neurodevelopmental differences in adolescents, the applicability of these models remains unclear. Directly addressing this gap in knowledge, this systematic review aimed to (1) describe the evidence of leading ideation-to-action theories (i.e., IPTS, IMV, 3ST) as they relate to suicide risk among adolescents, (2) integrate ideation-to-action theories within prevailing biological frameworks of adolescent suicide, and (3) provide recommendations for future adolescent suicide research. Overall, few studies provided a complete test of models in adolescent samples, and empirical research testing components of these theories provided mixed support. Future research would benefit from integrating neurodevelopmental and developmentally sensitive psychosocial frameworks to increase the applicability of ideation-to-action theories to adolescents. Further, utilizing real-time monitoring approaches may serve to further clarify the temporal association among risk factors and suicide.
... The neurobiological correlates of suicide risk in youth, however, remain unclear (see (2) for a review). Given that adolescence represents a time of significant brain plasticity governing the growth of cognitive and social competencies (3) as well as puberty-related changes (4), clarifying the neural correlates of suicide risk in this vulnerable population will have important implications for developing adolescent-specific interventions to reduce suicidality during this high-risk period. ...
Article
Full-text available
Introduction Suicide is a current leading cause of death in adolescents and young adults. The neurobiological underpinnings of suicide risk in youth, however, remain unclear and a brain-based model is lacking. In adult samples, current models highlight deficient serotonin release as a potential suicide biomarker, and in particular, involvement of serotonergic dysfunction in relation to the putamen and suicidal behavior. Less is known about associations among striatal regions and relative suicidal risk across development. The current study examined putamen connectivity in depressed adolescents with (AT) and without history of a suicide attempt (NAT), specifically using resting-state functional magnetic resonance imaging (fMRI) to evaluate patterns in resting-state functional connectivity (RSFC). We hypothesized the AT group would exhibit lower striatal RSFC compared to the NAT group, and lower striatal RSFC would associate with greater suicidal ideation severity and/or lethality of attempt. Methods We examined whole-brain RSFC of six putamen regions in 17 adolescents with depression and NAT (MAge [SD] = 16.4[0.3], 41% male) and 13 with AT (MAge [SD] = 16.2[0.3], 31% male). Results Only the dorsal rostral striatum showed a statistically significant bilateral between-group difference in RSFC with the superior frontal gyrus and supplementary motor area, with higher RSFC in the group without a suicide attempt compared to those with attempt history (voxel-wise p<.001, cluster-wise p<.01). No significant associations were found between any putamen RSFC patterns and suicidal ideation severity or lethality of attempts among those who had attempted. Discussion The results align with recent adult literature and have interesting theoretical and clinical implications. A possible interpretation of the results is a mismatch of the serotonin transport to putamen and to the supplementary motor area and the resulting reduced functional connectivity between the two areas in adolescents with attempt history. The obtained results can be used to enhance the diathesis-stress model and the Emotional paiN and social Disconnect (END) model of adolescent suicidality by adding the putamen. We also speculate that connectivity between putamen and the supplementary motor area may in the future be used as a valuable biomarker of treatment efficacy and possibly prediction of treatment outcome.
... Research has reported that an increased risk of suicide attempts and more severe suicidal thoughts and intentions are associated with relatively low levels of Estradiol and Progesterone (i.e., during the follicular/menstrual or early premenstrual phases). 26 ...
Article
Full-text available
Adolescence is established as an important stage in the human being, we try to show how positive or negative this stage can be in the development of an adequate environment, elucidating the benefits or consequences that can affect the psychomotor development of a person. Adolescence is the time when the personality of an individual is defined, influenced by various factors that can help the subject to develop adequately in society, as well as factors that can affect the subject conditioning him/her to an environment that is not favorable for his/her development. Both physical and psychological changes are mentioned, as well as hormonal factors which play an important role in development. It is intended to demonstrate the hormonal mechanisms that condition the changes in the development of the human being. TRANSLATE with x English Arabic Hebrew Polish Bulgarian Hindi Portuguese Catalan Hmong Daw Romanian Chinese Simplified Hungarian Russian Chinese Traditional Indonesian Slovak Czech Italian Slovenian Danish Japanese Spanish Dutch Klingon Swedish English Korean Thai Estonian Latvian Turkish Finnish Lithuanian Ukrainian French Malay Urdu German Maltese Vietnamese Greek Norwegian Welsh Haitian Creole Persian // TRANSLATE with COPY THE URL BELOW Back EMBED THE SNIPPET BELOW IN YOUR SITE Enable collaborative features and customize widget: Bing Webmaster Portal Back //
Article
Relevance. Over the past decade, the number of children who are characterized by suicidal behavior has increased dramatically. This factor is caused by various reasons, which can be both external and internal. At the same time, the number of such reasons is only growing, which, in turn, determines the relevance of this study. Purpose. The purpose of the work was to establish the features of the implementation of psychological profiling and correctional work with adolescents in the context of preventing them from committing suicide. Methodology. The study used the method of analysis and synthesis, comparison, deduction, modeling, testing. Results. As a result, it was found that the effective organization of psychological profiling among students should be based on an integrated approach, namely, a system of methods, forms and tools designed to identify and prevent suicidal tendencies among young people. In addition, it has been proven that the above system should include the education of parents regarding possible psycho-emotional problems among children, teachers and students. Conclusions. Attention was paid to bullying, in particular, in the school environment, attention was focused on tools for combating it, among which it is worth noting the observation of students, the control and analysis of their behavior, the conduct of timely psychological consultations. In addition, the factors that influence the deformation of the psycho-emotional state in adolescents were studied, which as a result provokes the appearance of suicidal behavior. Given this, it was noted that the results of this study can be used by the above categories of persons for their own development, as well as for the development of special methodological materials regarding psychological work with students in educational institutions.
Article
Full-text available
Previous research has identified how menstruation is an important factor in both attempted and completed suicides for women. The purpose of this review was to outline (a) the risk profile for suicidality in women who were identified to experience Premenstrual Dysphoric Disorder (PMDD), a condition characterized by severe physical and psychological changes that occur during the luteal menstrual phase, and (b) the implications of these findings for clinical practice. A systematic literature review was conducted using five databases to identify any peer-reviewed articles published between 1989 and 2019. Ten papers eligible for inclusion were identified: three pertaining to suicide cognitions, five to suicide attempts and two to both cognitions and attempts. Findings showed that suicidal thoughts, ideation, plans and attempts were strongly associated with experiences of PMDD and that these findings were independent of psychiatric co-morbidities. However, women with PMDD did not present with more severe risk profiles for suicide attempts (in terms of frequency, impulsivity and lethality) or make more frequent attempts during the luteal menstrual phase compared with suicide attempters without PMDD. Women with PMDD should be considered a high risk group for suicidality; thus, identifying and treating symptoms are vital in reducing suicide attempts. Implications for clinical practice are outlined in the discussion.
Article
Full-text available
The brain is an endocrine organ, sensitive to the rhythmic changes in sex hormone production that occurs in most mammalian species. In rodents and nonhuman primates, estrogen and progesterone’s impact on the brain is evident across a range of spatiotemporal scales. Yet, the influence of sex hormones on the functional architecture of the human brain is largely unknown. In this dense-sampling, deep phenotyping study, we examine the extent to which endogenous fluctuations in sex hormones alter intrinsic brain networks at rest in a woman who underwent brain imaging and venipuncture for 30 consecutive days. Standardized regression analyses illustrate estrogen and progesterone’s widespread associations with functional connectivity. Time-lagged analyses examined the temporal directionality of these relationships and suggest that cortical network dynamics (particularly in the Default Mode and Dorsal Attention Networks, whose hubs are densely populated with estrogen receptors) are preceded—and perhaps driven—by hormonal fluctuations. A similar pattern of associations was observed in a follow-up study one year later. Together, these results reveal the rhythmic nature in which brain networks reorganize across the human menstrual cycle. Neuroimaging studies that densely sample the individual connectome have begun to transform our understanding of the brain’s functional organization. As these results indicate, taking endocrine factors into account is critical for fully understanding the intrinsic dynamics of the human brain.
Article
Full-text available
There is no definitive neural marker of suicidal thoughts and behaviors (STB) or non-suicidal self-injury (NSSI), and relative to adults, research in youth is more limited. This comprehensive review focuses on magnetic resonance imaging (MRI) studies reporting structural and functional neural correlates of STB and NSSI in youth to: (i) elucidate shared and independent neural alternations, (ii) clarify how developmental processes may interact with neural alterations to confer risk, and (iii) provide recommendations based on convergence across studies. Forty-seven articles were reviewed (STB = 27; NSSI = 20), and notably, 63% of STB articles and 45% of NSSI articles were published in the previous 3 years. Structural MRI research suggests reduced volume in the ventral prefrontal and orbitofrontal cortices among youth reporting STB, and there is reduced anterior cingulate cortex volume related to STB and NSSI. With regard to functional alterations, blunted striatal activation may characterize STB and NSSI youth, and there is reduced frontolimbic task-based connectivity in suicide ideators and attempters. Resting state functional connectivity findings highlight reduced positive connectivity between the default mode network and salience network in attempters, and self-injurers exhibit frontolimbic alterations. Together, suicidal and non-suicidal behaviors are related to top-down and bottom-up neural alterations, which may compromise approach, avoidance, and regulatory systems. Future longitudinal research with larger and well-characterized samples, especially those integrating ambulatory stress assessments, will be well-positioned to identify novel targets that may improve early identification and treatment for youth with STB and NSSI.
Article
Full-text available
Suicide and suicidal behaviors represent a leading cause of morbidity and mortality during adolescence. While several lines of evidence suggest that suicidal behaviors are associated with risky decisions and deficient cognitive control in laboratory tasks in adults, comparatively less is known about adolescents. Here, we systematically reviewed the literature on the association between these neurocognitive variables and adolescent suicidal behaviors. The online search strategy identified 17 neurocognitive studies examining either cognitive control or decision-making processes in adolescents with past suicidal behaviors. Several studies have reported that adolescents with a history of suicidal behaviors present neuropsychological differences in the cognitive control (using Go/NoGo, suicide Stroop Test, continuous performance test, suicide/death Implicit Association Test), and decision-making (Iowa Gambling Task, Cambridge Gambling Task, cost computation, delay discounting, loss aversion tasks) domains. Due to a lack of replication or conflicting findings, our systematic review suggests that no firm conclusion can be drawn as to whether altered decision-making or poor cognitive control contribute to adolescent suicidal behaviors. However, these results collectively suggest that further research is warranted. Limitations included scarcity of longitudinal studies and a lack of homogeneity in study designs, which precluded quantitative analysis. We propose remediating ways to continue neuropsychological investigations of suicide risk in adolescence, which could lead to the identification of novel therapeutic targets and predictive markers, enabling early intervention in suicidal youth.
Article
Full-text available
Atypical regulation of the hypothalamic-pituitary-adrenal (HPA) axis is a putative mechanism underlying the association between exposure to early life stress (ELS) and the subsequent development of mental and physical health difficulties. Recent research indicates that puberty is a period of HPA-axis plasticity during which the effects of exposure to ELS on cortisol regulation may change. In particular, increases in the sex hormones that drive pubertal maturation, including dehydroepiandrosterone (DHEA) and testosterone, may be implicated in pubertal recalibration of cortisol regulation. In the current study, we examined the associations among levels of objectively-rated threat-related ELS and salivary waking cortisol, DHEA, and testosterone in a sample of 178 adolescents (55% female) who were in early puberty at baseline (Tanner stages 1-3; mean Tanner stage[SD] = 1.93[0.64]; mean age[SD] = 11.42[1.04]) and were followed up approximately two years later (mean Tanner stage[SD] = 3.46[0.86]; mean age[SD] = 13.38[1.06]). Using multi-level modeling, we disaggregated the effects of between-individual levels and within-individual increases in pubertal stage and sex hormones on change in cortisol. Controlling for between-individual differences in average pubertal stage, the association between levels of cortisol and DHEA was more strongly positive among adolescents who evidenced greater within-individual increases in pubertal stage across time. Both higher average levels and greater within-individual increases in DHEA and testosterone were associated with increases in cortisol across time, indicating positive coupling of developmental changes in these hormones; however, coupling was attenuated in adolescents who were exposed to more severe threat-related ELS prior to puberty. These findings advance our understanding of the development of the HPA-axis and its association with childhood environmental risk during puberty.
Article
Full-text available
Adolescence is characterized by rapid brain development in white matter (WM) that is attributed in part to surges in gonadal hormones. To date, however, there have been few longitudinal investigations relating changes in gonadal hormones and WM development in adolescents. We acquired diffusion-weighted MRI to estimate mean fractional anisotropy (FA) from 10 WM tracts and salivary testosterone from 51 females and 29 males (ages 9–14 years) who were matched on pubertal stage and followed, on average, for 2 years. We tested whether interactions between sex and changes in testosterone levels significantly explained changes in FA. We found positive associations between changes in testosterone and changes in FA within the corpus callosum, cingulum cingulate, and corticospinal tract in females (all ps<0.05, corrected) and non-significant associations in males. We also collected salivary estradiol from females and found that increases in estradiol were associated with increases in FA in the left uncinate fasciculus (p = 0.04, uncorrected); however, this effect was no longer significant after accounting for changes in testosterone. Our findings indicate there are sex differences in how changes in testosterone relate to changes in WM microstructure of tracts that support impulse control and emotion regulation across the pubertal transition.
Article
Full-text available
Adolescence may mark a sensitive period for the development of higher-order cognition through enhanced plasticity of cortical circuits. At the same time, animal research indicates that pubertal hormones may represent one key mechanism for closing sensitive periods in the associative neocortex, thereby resulting in decreased plasticity of cortical circuits in adolescence. In the present review, we set out to solve some of the existing ambiguity and examine how hormonal changes associated with pubertal onset may modulate plasticity in higher-order cognition during adolescence. We build on existing age-comparative cognitive training studies to explore how the potential for change in neural resources and behavioral repertoire differs across age groups. We review animal and human brain imaging studies, which demonstrate a link between brain development, neurochemical mechanisms of plasticity, and pubertal hormones. Overall, the existent literature indicates that pubertal hormones play a pivotal role in regulating the mechanisms of experience-dependent plasticity during adolescence. However, the extent to which hormonal changes associated with pubertal onset increase or decrease brain plasticity may depend on the specific cognitive domain, the sex, and associated brain networks. We discuss implications for future research and suggest that systematical longitudinal assessments of pubertal change together with cognitive training interventions may be a fruitful way toward a better understanding of adolescent plasticity. As the age of pubertal onset is decreasing across developed societies, this may also have important educational and clinical implications, especially with respect to the effects that earlier puberty has on learning.
Preprint
Full-text available
The brain is an endocrine organ, sensitive to the rhythmic changes in sex hormone production that occurs in most mammalian species. In rodents and nonhuman primates, estrogen and progesterone’s impact on the brain is evident across a range of spatiotemporal scales. Yet, the influence of sex hormones on the functional architecture of the human brain is largely unknown. In this dense-sampling, deep phenotyping study, we examine the extent to which endogenous fluctuations in sex hormones alter intrinsic brain networks at rest in a woman who underwent brain imaging and venipuncture for 30 consecutive days. Standardized regression analyses illustrate estrogen and progesterone’s widespread associations with functional connectivity. Time-lagged analyses examined the temporal directionality of these relationships and suggest that cortical network dynamics (particularly in the Default Mode and Dorsal Attention Networks, whose hubs are densely populated with estrogen receptors) are preceded—and perhaps driven—by hormonal fluctuations. A similar pattern of associations was observed in a follow-up study one year later. Together, these results reveal the rhythmic nature in which brain networks reorganize across the human menstrual cycle. Neuroimaging studies that densely sample the individual connectome have begun to transform our understanding of the brain’s functional organization. As these results indicate, taking endocrine factors into account is critical for fully understanding the intrinsic dynamics of the human brain. Highlights Intrinsic fluctuations in sex hormones shape the brain’s functional architecture. Estradiol facilitates tighter coherence within whole-brain functional networks. Progesterone has the opposite, reductive effect. Ovulation (via estradiol) modulates variation in topological network states. Effects are pronounced in network hubs densely populated with estrogen receptors.
Article
Full-text available
Identifying brain alterations that contribute to suicidal thoughts and behaviors (STBs) are important to develop more targeted and effective strategies to prevent suicide. In the last decade, and especially in the last 5 years, there has been exponential growth in the number of neuroimaging studies reporting structural and functional brain circuitry correlates of STBs. Within this narrative review, we conducted a comprehensive review of neuroimaging studies of STBs published to date and summarize the progress achieved on elucidating neurobiological substrates of STBs, with a focus on converging findings across studies. We review neuroimaging evidence across differing mental disorders for structural, functional, and molecular alterations in association with STBs, which converges particularly in regions of brain systems that subserve emotion and impulse regulation including the ventral prefrontal cortex (VPFC) and dorsal PFC (DPFC), insula and their mesial temporal, striatal and posterior connection sites, as well as in the connections between these brain areas. The reviewed literature suggests that impairments in medial and lateral VPFC regions and their connections may be important in the excessive negative and blunted positive internal states that can stimulate suicidal ideation, and that impairments in a DPFC and inferior frontal gyrus (IFG) system may be important in suicide attempt behaviors. A combination of VPFC and DPFC system disturbances may lead to very high risk circumstances in which suicidal ideation is converted to lethal actions via decreased top-down inhibition of behavior and/or maladaptive, inflexible decision-making and planning. The dorsal anterior cingulate cortex and insula may play important roles in switching between these VPFC and DPFC systems, which may contribute to the transition from suicide thoughts to behaviors. Future neuroimaging research of larger sample sizes, including global efforts, longitudinal designs, and careful consideration of developmental stages, and sex and gender, will facilitate more effectively targeted preventions and interventions to reduce loss of life to suicide.
Article
The marked increase in adolescent reward-seeking behavior has important implications for adaptive and maladaptive development. Reward-seeking is linked to increased testosterone and increased neural responses to reward cues. How acute testosterone changes modulate neural reward systems remains unclear. Based on previous work, adolescents, particularly males, showing an increase in endogenous testosterone reactivity were hypothesized to show increased neural response to reward in ventromedial prefrontal cortex, ventral striatum, and posterior cingulate cortex. Sixty-one healthy adolescents aged 10 to 13 (38 female, mean age = 12.01 [SD = 1.00]) completed a reward-cue processing task during fMRI. Saliva samples to be assayed for testosterone were collected immediately before and after scanning. Acute testosterone changes were not associated with variation in behavioral performance. Within ventromedial prefrontal and posterior cingulate cortices, increased acute testosterone change was associated with reduced discrimination between rewarded and un-rewarded trials. Results suggest that increasing levels of testosterone may result in reduced attention to/salience of task irrelevant information. In contrast to previous studies that found a positive association between testosterone and neural response to reward, the reward information in the current paradigm was irrelevant to success in task performance. These results are consistent with theoretical conceptualization of testosterone’s role in reproduction, which involves a shift in salience to short-term relative to long-term goals. These data further emphasized the need to consider context in the study of hormones; specific behaviors will be up- or down-regulated by a hormone based on the fit of the behavior with the broader contextual goal being orchestrated by the hormone.