You are on page 1of 13

Journal of Inherited Metabolic Disease (2018) 41:585–596

https://doi.org/10.1007/s10545-018-0168-1

REVIEW

The role of suboptimal mitochondrial function in vulnerability


to post-traumatic stress disorder
Graeme Preston 1 & Faisal Kirdar 1 & Tamas Kozicz 1,2,3

Received: 20 October 2017 / Revised: 28 February 2018 / Accepted: 2 March 2018 / Published online: 28 March 2018
# SSIEM 2018

Abstract
Post-traumatic stress disorder remains the most significant psychiatric condition associated with exposure to a traumatic event,
though rates of traumatic event exposure far outstrip incidence of PTSD. Mitochondrial dysfunction and suboptimal mitochon-
drial function have been increasingly implicated in several psychopathologies, and recent genetic studies have similarly sug-
gested a pathogenic role of mitochondria in PTSD. Mitochondria play a central role in several physiologic processes underlying
PTSD symptomatology, including abnormal fear learning, brain network activation, synaptic plasticity, steroidogenesis, and
inflammation. Here we outline several potential mechanisms by which inherited (genetic) or acquired (environmental) mito-
chondrial dysfunction or suboptimal mitochondrial function, may contribute to PTSD symptomatology and increase suscepti-
bility to PTSD. The proposed pathogenic role of mitochondria in the pathophysiology of PTSD has important implications for
prevention and therapy, as antidepressants commonly prescribed for patients with PTSD have been shown to inhibit mitochon-
drial function, while alternative therapies shown to improve mitochondrial function may prove more efficacious.

Incidence and prevalence of PTSD hyperarousal — symptoms pointing to potential defects in


and traumatic event exposure neural circuits related to fear learning, threat detection, con-
text processing, and executive function (Shalev et al 2017;
Post-traumatic stress disorder (PTSD) is a debilitating psy- Flory and Yehuda 2015). While the cause of the disorder —
chiatric condition that affects millions of individuals world- experience of a traumatic event (TE) — remains clear, a
wide. The current DSM-V requires an individual be exposed robust description of the etiology of PTSD remains elusive.
— either directly or indirectly — to a traumatic event in the For a given individual, the estimated risk of developing
form of actual or threatened death, serious injury, or sexual PTSD following trauma exposure can vary from 1% to
violence, to oneself or a close family member, or to repeat- greater than 50% (Spoont et al 2015). Lifetime prevalence
ed exposure to the details of such a traumatic event. of PTSD worldwide varies from 1.3 to 12.2%; the 1-year
Diagnosis requires the presence of symptoms of three types: prevalence varies from 0.2 to 3.8%. And while PTSD sus-
intrusive symptoms (such as memories, dreams, and flash- ceptibility varies between high- and low-income countries,
backs) associated with the traumatic event, symptoms of and occurs more frequently in soldiers, first responders and
avoidance, and symptoms of hypervigilance and other groups regularly exposed to trauma, these differences
may be the result of differences in traumatic event exposure,
Communicated by: Eva Morava or social factors, rather than due to some underlying biolog-
ic factor (Karam et al 2014, Shalev et al 2017). The severity
* Graeme Preston of TE does not appear to be predictive of subsequent PTSD
gpreston@tulane.edu severity (Karam et al 2014, Rubin and Feeling 2013); how-
ever, the number of TEs experienced appears to be highly
1
Hayward Genetics Center, Tulane University School of Medicine, predictive of PTSD susceptibility and symptom severity,
1430 Tulane Ave, New Orleans, LA 70112, USA with PTSD associated with 4 or more TEs (4+/PTSD)
2
Department of Clinical Genomics, Mayo Clinic, Rochester, MN, showing significantly greater PTSD vulnerability and more
USA severe symptomatology compared to PTSD associated with
3
Department of Anatomy, Radboud University Medical Center, 3 or fewer TEs (3-/PTSD) (Karam et al 2014, Benjet et al
Nijmegen, Netherlands 2015). There are also data to suggest that TE type may
586 J Inherit Metab Dis (2018) 41:585–596

predict PTSD vulnerability (Ozer et al 2003, Roberts et al contribute to PTSD susceptibly by influencing these biologi-
2011, Kessler et al 2014). Interestingly, those TE types most cal domains of PTSD pathophysiology Fig. 1.
predictive of PTSD vulnerability, such as sexual assault and
physical violence are also those TEs most associated with 4
+/PTSD, and those highly predictive TE types may be as- Allostasis, allostatic load, stress, and brain
sociated with revictimization and multiple TE exposure mitochondria
(Karam et al 2014, Benjet et al 2015). The incidence of
trauma, notably, remains much more prevalent than that of Mitochondria are the primary metabolic organelles of eukary-
PTSD, and most individuals exposed to a TE do not devel- otic organisms, and responding to a stressful environment,
op PTSD: 64–70% of adults worldwide experience a TE at such as exposure to a traumatic event, requires energy.
some time in their lives and 31% experience four or more Living organisms continually attempt to maintain homeosta-
events, though under-reporting of TE exposure is expected sis, a physiologic state in which metabolic functions are kept
(Shalev et al 2017; Benjet et al 2016). Based on this epide- within an optimal range of parameters necessary to support the
miologic data, it seems likely that an underlying physiolog- essential functions of life in the face of ever-changing envi-
ic, genetic, or neurologic vulnerability factor exists. ronments. It often occurs, however, that a living thing encoun-
ters some environmental condition beyond what its normal
homeostatic functions can accommodate. In this case, physi-
Identified genetic susceptibility factors ologic parameters must be altered in such a way to retain its
for PTSD ability to restore homeostasis. This process is known as
allostasis (McEwen 1998). Allostasis is an essential process
Early genetic studies into PTSD vulnerability focused on for maintaining life functions through the various stages of a
genes involved in neurotransmitters and endocrine brain ef- normal life span. The process of undergoing allostasis, the
fectors, including serotonin, dopamine, glucocorticoids, and allostatic state, is necessarily hallmarked by an imbalance of
GABA, among others; however, results were inconsistent the biochemicals that mediate the change, such as corticoste-
(Broekman et al 2007). A recent study using a gene expression roids, cytokines and catecholamines (McEwen and Wingfield
panel focusing on mitochondrial genes in the prefrontal cortex 2003). The combined energetic and physiologic requirements
of post-mortem brains from six PTSD patients and six controls to complete allostasis are known as allostatic load. If there are
showed altered expression of eight mitochondria-related insufficient resources to achieve the necessary allostatic
genes, including six genes of the OXPHOS pathway, three change (the allostatic load is too great), or if the allostatic
genes of the TCA cycle, and three genes of the pyruvate path- change is inhibited by some other mechanism or does not
way (Su et al 2008). In a group of 87 male combat veterans, reach its proper end-point, and homeostatic capacity is not
mitochondrial DNA copy number was reduced in restored, a state of allostatic overload can result (McEwen
granulocytes of PTSD-affected individuals compared to unaf- and Wingfield 2003; Picard et al 2014).
fected individuals, and was significantly correlated with pos- The potential hazards of allostatic load are of particular
itive affectivity (Bersani et al 2015). A subsequent study of significance to the brain. Allostatic state is a highly ener-
978 single nucleotide polymorphisms in the mtDNA of 1238 getic process, and the brain is a highly energetic organ.
individuals showed significant correlation between PTSD se- Constituting only 2% of the human body mass by weight,
verity and the heteroplasmy levels in leukocytes of two the brain is responsible for 20% of the total energy me-
mtDNA SNPs in genes for respiratory chain proteins tabolism of the body (Harris et al 2012). Mitochondria
(Flaquer et al 2015). play a central role in stress response and allostasis, par-
These pieces of genetic evidence for mitochondrial in- ticularly in the brain (Picard et al 2014; Jou et al 2009).
volvement in PTSD imply that mitochondrial dysfunction One of the primary biological functions of mitochondria
may play a role in the etiology of PTSD (Jou et al 2009). is the production of usable cellular energy in the form of
We argue that mitochondria could play crucial roles in many ATP through oxidative phosphorylation (OXPHOS) by
of the physiologic processes involved in PTSD symptomatol- the enzymatic complexes of the electron transport chain,
ogy, including hypocortisolemia, activation/inhibition of brain a pathway responsible for 90% of the cellular energy pro-
networks involved in fear learning and extinction; neuronal duction in the human body (Chance et al 1979).
plasticity including neurogenesis, apoptosis, and synaptic ar- Mitochondria are also instrumental in calcium ion (Ca2+)
chitecture (Cheng et al 2010); the regulation of stress hormone sequestration (Rizzuto et al 2012), fatty acid oxidation
production and release (Miller 2013; Smith and Vale 2006); (Wanders et al 2010), nucleic acid metabolism (Wang
and inflammatory response (Lopez-Armada et al 2013). Here, 2016), are the primary producers of ROS production
we outline several mechanisms by which mitochondrial dys- (Murphy 2009), and are the prime mediators of apoptotic
function or suboptimal mitochondrial function (SMF) may cell death (Wang and Youle 2016). Mitochondria undergo
J Inherit Metab Dis (2018) 41:585–596 587

Fig. 1 Mitochondrial dysfunction, either inherited (genetic) or acquired resulting in abnormal physiologic response to stress in the neurons of the
(environmental), disrupts production of cellular energy (ATP), buffering brain, contributing to PTSD symptomatology, and increasing vulnerabil-
of cytosolic Ca2+, ROS production, and mitochondrial fission and fusion, ity to PTSD

fission and fusion, and the formation and fracturing of et al 2005). These corticosteroid hormones have been
these fused mitochondrial superstructures is directly relat- shown to alter the function and structure of the brain,
ed to cellular metabolic function, and promotes cell sur- influencing neurogenesis, neural atrophy, cell turnover,
vival, particularly in response to stress (Friedman and and neural physiology and excitability (Datson et al 2001;
Nunnari 2014). Each mitochondrion possesses thousands Gould et al 1990; Sloviter et al 1989; Hu et al 1997;
of copies of its own genome, and while most of the genes Wossink et al 2001; Nair et al 2004; Almeida et al 2017;
coding for the ~1500 mitochondrial proteins have been Diamond et al 1992; Kim and Diamond 2002; Pavlides
relocated to nuclear chromosomes, the mitochondrion re- et al 1993; Xu et al 1997). The physiologic effects of
tains 22 tRNA genes, and 13 protein-coding genes, all glucocorticoids are of great importance to the understanding
essential equipment for mitochondrial gene transcription of PTSD. Paradoxically, patients with PTSD show reduced
and assembly of the electron transport chain (Taanman levels of plasma cortisol (hypocortisolaemia) and increased
1999). Exposed to the highly oxidative environment of levels of corticotropin-releasing hormone (CRH) in cerebro-
the mitochondrial matrix, and without the DNA repair spinal fluid (Baker et al 1999), indicating an HPA axis
mechanisms of the nucleus, the mitochondrial genome is dysregulation. Mitochondria play potentially pathogenic
highly susceptible to oxidative damage and DNA muta- roles in many of the steps of HPA axis-mediated glucocor-
tion (Shokolenko et al 2009). These nucleoid genomes ticoid release and regulation (Chow et al 2017).
can be reshuffled through mitochondrial fission and fu- Glucocorticoid synthesis takes place within mitochondria
sion, diluting deleterious mutations, potentially mitigating of the steroidogenic cells of the adrenal cortex, and is de-
the resultant bioenergetic disruptions. Mitochondrial mu- pendent on enzymes responsible for sensing cholesterol on
tations induced by oxidative damage can result in mito- the outer mitochondrial matrix, transport of the cholesterol
chondrial dysfunction resulting in additional oxidative into the mitochondrial matrix, and the ultimate synthesis of
damage (Cui et al 2012). The delicate balance of mito- glucocorticoids from cholesterol (Miller 2013). Primary ad-
chondrial function can easily be disrupted, causing a cas- renal insufficiency (Addison’s Disease) has been reported in
cade of disfunction which can ultimately result in apopto- mitochondrial disease cases with mtDNA mutations, oxida-
tic cell death, necrosis, or severe cellular disfunction tive phosphorylation defects, and large scale mtDNA dele-
(Marchi et al 2012). Patients with inherited mitochondrial tions (North et al 1996; Nicolino et al 1997; Bruno et al
dysfunction or SMF may have sufficient mitochondrial 1998; Boles et al 1998; Artuch et al 1998). Steroid hor-
capacity to meet the energetic needs of their day-to-day mone synthesis by steroidogenic cells of the adrenal cortex
lives, but may have insufficient mitochondrial capacity to is stimulated by binding of adrenocorticotropic hormone
respond to a highly stressful environment like a traumatic (ACTH) to melanocortin type-2 receptors (MCR-2) and
event exposure. Alternatively, mounting a physiologic re- subsequent Ca2+ influx and cAMP induction (Kojima
sponse to a traumatic event exposure may induce addi- et al 1985; Durroux et al 1991; Yamazaki et al 1998).
tional mitochondrial dysfunction in patients with inherited However, maintenance of optimal steroidogenic capacity
mitochondrial dysfunction or SMF. requires chronic activation by ACTH, and prolonged enzy-
matic induction by cAMP activation (Simpson and
Waterman 1988). Mitochondrial dysfunction or reduced mi-
PTSD, hypocortisolemia, and mitochondria tochondrial capacity may allow for sufficient Ca2+ buffering
to facilitate the initial glucocorticoid synthesis induced by
Glucocorticoid hormone release is a key aspect of stress ACTH activity in response to stress, but may be insuffi-
response, with plasma concentrations of glucocorticoids cient to reliably maintain that buffering capacity in response
peaking 15–30 min following a stressful stimulus and to chronic, long term ACTH activation, resulting in ste-
returning to pre-stress levels after 60–90 min (de Kloet roidogenic dysfunction and hypocortisolemia.
588 J Inherit Metab Dis (2018) 41:585–596

PTSD, neuronal circuits of fear, response (Li et al 2014). A subsequent 2015 study found 34
and mitochondria genes related to mitochondria upregulated in the amygdalae of
stressed rats (Zhang et al 2015). Pharmacologic increase in
PTSD represents an abnormal response to a stressful environ- mitochondrial cytochrome oxidase activity has been shown
ment (de Kloet et al 2005). Abnormal activity in the neural to increase fear extinction (Telch et al 2014). The glutamate
circuits associated with a healthy stress and fear response, receptor N-methyl-D-aspartate (NMDA) acts as a calcium ion
including fear and safety learning, threat detection, executive channel; buffering of intracellular Ca2+ levels by mitochondria
function and emotional regulation, and contextual processing, is crucial for NMDA-mediated neuronal activation, and dys-
may play a significant role in the pathophysiology of PTSD functional mitochondrial Ca2+ buffering has been implicated
(Shalev et al 2017, VanElzakker et al 2014, Quirk and Mueller in NMDA-mediated excitotoxicity and irreversible neuronal
2007, Hayes et al 2012, Liberzon and Abelson 2016, Olff et al damage and loss (Peng et al 1998). NMDA-mediated neuronal
2014, Aupperle et al 2012, Polak et al 2012). activation also induces the production of reactive oxygen spe-
Consolidation of fear memory is crucial for an animal’s cies (ROS) and nitric oxide by mitochondria, which may be
defense against threats. The consolidation of memories asso- responsible for NMDA-mediated excitotoxicity (Dugan et al
ciated with fear has been repeatedly mapped to the amygdala 1995; Gunasekar et al 1995; Reynolds and Hastings 1995).
and is dependent upon neuroplasticity in that area (Fanselow Maintenance of proper ROS levels has also been shown to
and LeDoux 1999). Closely related to fear conditioning is the mediate GABA-ergic inhibitory signaling (Accardi et al
process of fear extinction, a progressive reduction in fear re- 2014). We propose that mitochondrial dysfunction, or subop-
sponse to a previously dangerous environment in which no timal mitochondrial function (SMF), results in disrupted or
adverse conditions result (Myers and Davis 2007). Inefficient dysfunctional signaling by GABA-ergic interneurons respon-
fear extinction in patients with PTSD results in exaggerated sible for inhibition of the amygdala by the medial prefrontal
fear response in situations reminiscent of trauma, and leads to cortex, and subsequent fear extinction, leading to the persis-
symptoms of hyperarousal and increased startle response tent and inappropriate fear response characteristic of PTSD,
(Francati et al 2007). Ca 2+ -dependent post synaptic either through insufficient cellular energy to maintain proper
long-term potentiation (LTP) is instrumental in both the con- ion gradients, deficient Ca2+ buffering leading to aberrant
solidation of fear memories (Huang and Kandel 1998) as well NMDA mediated signaling, disrupted levels of ROS, or some
as fear extinction (Charney 2004). Fear extinction involves combination of these effects.
inhibition of the amygdala (LeDoux 1993), and functional
neuroimaging studies have shown increased activity in the
amygdalae of patients with PTSD (Liberzon et al 1999; PTSD, inflammation, and mitochondria
Pissiota et al 2002; Shin et al 1997; Hendler et al 2003;
Yang et al 2004; Shin et al 2004) suspected to result in defi- Altered inflammatory markers have been repeatedly identified
cient fear extinction and subsequent hyperarousal (Bremner in patients with PTSD. Increased concentrations of the
et al 1995). Initial activation of the amygdala is modulated pro-inflammatory cytokines IL-1β, TNFα, IL-2, IL-6,
by the medial prefrontal cortex (mPFC), and insufficient in- Interferon gamma (INF-y), and C-reactive protein (CRP) are
hibitory feedback may result in the hyperactivation of the all associated with trauma exposure (Tursich et al 2014). The
amygdala in PTSD patients (Nutt and Malizia 2004). inflammatory response is tightly associated with mitochondri-
Amygdalar inhibition by the mPFC is mediated by GABA al function: mitochondria can induce an inflammatory re-
inter-neurons (Charney 2004), high energy neurons character- sponse as part of the innate immune response, and inflamma-
ized by repetitive, high frequency action potentials (Kann et al tion can influence mitochondrial function (Lopez-Armada
2014). To maintain the ion gradients required for this activity, et al 2013). A primary mediator of the inflammatory response
GABA interneurons consume significantly more cellular en- is ROS production, which can activate the NLRP3
ergy than other cell types in the brain, and contain a high inflammasome (Kepp et al 2011). Ros production induced
density of mitochondria and cytochrome oxidase (Kageyama by mitochondrial dysfunction can lead to activation and secre-
and Won-Riley 1982; Gulyas et al 2006). These cells are sub- tion of IL-1β (Strowig et al 2012). Inhibition of ATP synthesis
sequently highly sensitive to defects in the mitochondrial re- by oligomycin, which has been shown to increase ROS pro-
spiratory chain (Kann et al 2011; Whittaker et al 2011), and duction (Tan et al 1998) increased the sensitivity of IL-1β
interneuron dysfunction and loss have been implicated in the tenfold in human sinoviocytes (Valcarcel-Ares et al 2014).
neuropathology of patients with mitochondrial disease. A Degraded mtDNA has been shown to induce IL-1β, IL-6,
2014 study found several up- and down-regulated genes in and TNFα in mouse astrocytes (Mathew et al 2012).
the amygdalae of rats with PTSD characterized by exaggerat- Pro-apoptotic signaling promotes release of IL-1β in mouse
ed startle, several of which have previously been associated macrophages (Nakahira et al 2011). Both IL-1β and TNFα
with neuronal dysfunction, psychiatric disorders, and stress inhibit complex I activity and ATP synthesis, resulting in
J Inherit Metab Dis (2018) 41:585–596 589

increased ROS production in human lymphocytes (Guidarelli mitochondrial bioenergetics, mitochondrial dynamics (fission
et al 2007, Kim et al 2010), and complex I deficiency can and fusion), Ca2+ homeostasis, ROS production, and subcellular
induce IL-1β in the hypothalamus (Yi et al 2007). IL-6 can mitochondrial distribution (Prigione et al 2010; Voccoli and
increase dopamine activity in the amygdala and hippocampus, Colombaioni 2009). Levels of PGC1α, the primary regulator
and may influence consolidation and extinction of fear learning, of mitochondrial biogenesis, are increased in neural progenitor
and context processing (Pervanidou et al 2007). Increased IL-6 cells and newly-developed neurons in brains of developing rats
levels have been implicated in the development of PTSD in (Cowell et al 2007), and PGC1α-knockout mice display loss of
children following motor vehicle accidents (Pervanidou et al brain matter and behavioral anomalies (Lin et al 2004).
2007). INF-y has been shown to inhibit the expression of Mitochondrial uncoupling protein (UCP) has recently been
SIRT1, an important regulator of mitochondrial biogenesis shown to mediate ATP production, ROS production, and
and turnover (Li et al 2012). Cytokine levels also appear to Ca2+ homeostasis (Chan et al 2006; Liu et al 2006), and
directly influence the pathophysiology of PTSD. In patients neuron-specific UCP has been implicated in regulating
with PTSD, TNFα and CRP levels correlate with PTSD symp- neuronal differentiation (Smorodchenko et al 2009).
tomatology generally (von Kanel et al 2007, Michopoulos et al Differentiation-inducing transcription factors influence
2015), and TNFα levels correlate with avoidance, regulation of several mitochondrial genes, including genes
re-experiencing, and hyperarousal specifically (von Kanel et al for mitochondrial transport, and are highly involved in
2007). TNFα inhibits LTP in the CA1 and dentate gyrus regions neurite outgrowth (Baxter et al 2009). Mitochondria appear
of the hippocampus in a partially MAPK-dependent manner to be instrumental in all phases of neurite outgrowth, from
(Butler et al 2004). Increased levels of circulating IL-1, IL-6, determination of the neuronal axis (Mattson and Partin
TNFα, and CRP have also been identified in patients with major 1999) to extension of the axon (Ruthel and Hollenbeck
depressive disorder (MDD) (Dowlati et al 2010) and reduced 2003; Dedov et al 2000), a role that goes beyond simple
hippocampal volumes similar to those observed in PTSD pa- production of ATP and which may involve mitochondrial
tients have been reported in MDD patients with high levels of Ca2+ buffering and its effects on cytoskeletal reorganiza-
IL-6 and CRP (Frodl and Amico 2014). Immune challenge in tion (Mattson and Partin 1999). Localized treatment with
healthy individuals induced increased levels of TNFα, IL-6 and nerve growth factor (NGF) induces actin-dependent trans-
IL-1, and decreased levels of brain derived neurotrophic factor port and retention of mitochondria to the site of NGF in-
(BDNF) in the hippocampus (Dunn et al 2005), while also fusion, and an increase in mitochondrial membrane poten-
increasing TNFα levels in the PFC (Feleder et al 2010), and tial (Verburg and Hollenbeck 2008; Chada and Hollenbeck
decreasing connectivity between the ACC, mPFC, and amyg- 2004). Evidence suggests that mitochondrial dysfunction
dala (Frodl and Amico 2014), resulting in impaired learning and has an even greater effect on dendritic morphology than
memory (Dunn et al 2005). We propose that mitochondrial axon morphology, as induced mitochondrial dysfunction
dysfunction or SMF may increase an individual’s susceptibility insufficient to significantly disrupt axonal morphology sig-
to the kinds of abnormal inflammatory response observed in nificantly disrupted dendritic tree formation (Chihara et al
patients suffering from PTSD, and that this abnormal inflamma- 2007). Mitochondria are significantly more active in the
tory response may contribute to PTSD symptomatology. dendrites than in the axons of cultured hippocampal neu-
rons (Overly et al 1996). Activation of post-synaptic glu-
tamate receptors such as NMDA and AMDA (a—
PTSD, neuronal plasticity, and mitochondria amino-3-hydroxy-5-methylisoxazole-4-propionic acid) in-
duces significant influx of extracellular and endoplasmic
Mitochondria play an important role in synaptic plasticity by reticulum Ca2+ into the cytosol (Toresson and Grant 2005)
facilitating neurogenesis, neuron remodeling, and neurotransmit- which must be sequestered by mitochondria (Duchen
ter release, as well as providing the cellular energy required for 2000; Nicholls and Budd 2000; Toescu 2000). The number
the gene regulation changes, protein synthesis, cytoskeletal re- of mitochondria in the dendritic spines, where synaptic activa-
modeling, organelle trafficking, and intracellular transport re- tion occurs, increases in response to repeated depolarization,
quired for these processes (Cheng et al 2010). Neurogenesis is and this redistribution of mitochondria is facilitated by mito-
a highly energetic process, requiring large-scale cytoskeletal re- chondrial fission and fusion (Li et al 2004). NMDA is a potent
arrangement, organelle transport, and neuronal sprouting, mak- mediator of LTP (Lüscher and Malenka 2012) and has been
ing adequate mitochondrial function essential (Xavier et al 2015). shown to be important in memory and learning (Williams
Neuronal differentiation, a crucial step in neurogenesis, is marked et al 1998; Calabresi et al 2001; Li et al 2010). Induction of
by an increase in mitochondrial mass (Cheng et al 2010), as well LTP in the dentate gyrus of rats induced expression of several
as key mitochondrial proteins and transcription factors (Mattson mitochondrial genes within the dentate gyrus, as well as in
and Liu 2003; Kann and Kovacs 2007). The differentiation of neighboring regions of the hippocampus where LTP had not
neuronal stem cells into functional neurons involves shifts in occurred (Williams et al 1998). Mitochondrial activation of
590 J Inherit Metab Dis (2018) 41:585–596

caspase-9 and caspase-3 in dendrites, independent of apoptotic dysfunction, who experience a stressor, may experience a pro-
cell death, facilitates AMDA receptor internalization and sub- gressive degeneration of mitochondrial function, which could
sequent long-term depression (LTD) without effecting LTP (Li disrupt otherwise healthy neurogenesis, neuronal differentiation,
et al 2010). BDNF is a potent inducer of LTP in hippocampal neurite outgrowth, synaptic plasticity, and LTP, and may disrupt
neurons (Ying et al 2002) and has been shown to be instrumen- the balance of pro-and apoptotic factors, leading to cell
tal in hippocampal learning and memory (Lu et al 2009). Both death-induced neuropathy and psychopathology.
glutamate and BDNF are important mediators of synaptic
neurogenesis, synaptic plasticity, and neuronal structure
(Martin and Finsterwald 2011). Activation of glutamate recep- Therapeutic implications
tors in hippocampal neurons increased glucose metabolism
(Shuttleworth et al 2003), and BDNF promotes increased glu- The proposed roles of mitochondrial dysfunction in the above
cose utilization by neuronal mitochondrial in response to in- outlined biological domains of PTSD pathophysiology and
creased energy demand (Burkhalter et al 2003) and increased susceptibility to PTSD have several implications for treat-
mitochondrial complex I activity (Markham et al 2004). ment, management, and prevention. Current treatment for
While necrotic cell death has historically been implicated in PTSD focuses on antidepressant (AD) drug therapies that alter
neurodegenerative disease, apoptotic cell death is increasingly neurotransmissions by serotonin, dopamine, and norepineph-
recognized as the primary mediator (Tatton and Olanow 1999). rine. Currently, the selective serotonin reuptake inhibitors
The intrinsic apoptotic pathway is initiated by mitochondria, (SSRIs) Sertraline and Paroxetine are the only
which release cytochrome c into the cytosol, triggering the cas- FDA-approved drug treatments for PTSD, and are the pre-
pase cascade that results in apoptotic cell death (Reed 1997; ferred initial class of medications used to treat PTSD (Brady
Green and Reed 1998; Varfolomeev et al 1998; Juo et al 1998; et al 2000; Marshall et al 2001). Additional antidepressants
Kuida et al 1998; Hakem et al 1998). The intrinsic apoptotic commonly prescribed as first-line treatments for PTSD in-
pathway is controlled by the Bcl-2 family of proteins, which clude fluoxetine, mirtazapine, venlafazine, and nefazodone.
may have either pro- or anti-apoptotic effects; the ratio of Bcl-2 Tricyclic antidepressants and monoamine oxidase inhibitors
family species ultimately dictates the sensitivity of a cell to var- (MAOIs) can be used as second-line treatments. Several
ious pro- or anti-apoptotic signaling (Chen et al 1996, Middleton ADs show diverse adverse effects on mitochondrial function,
et al 1996, Song et al 1999, Inohara et al 1998). In hippocampal including energy metabolism and apoptosis. Fluoxetine in-
neurons, activation of the NMDA receptor by L-glutamate in- hibits OXPHOS in rat brain and liver mitochondria by affect-
creases elevated cytosolic Ca2+, inducing BAD/Bcl2-induced ing electron transport and ATP synthesis (Souza et al 1994;
apoptosis (Wang et al 1998). Apoptotic cell death mediated by Curti et al 1999). Fluoxetine and paroxetine increase apoptosis
Bcl-2 family proteins has been identified in numerous psycho- in glioma and neuroblastoma cells in vitro (Levkovitz et al
logical disorders including Alzheimers (MacGibbon et al 1997). 2005). Mirtazapine inhibited complex I activity in mitochon-
Ultimately, Bcl-2 family proteins mediate the intrinsic apoptotic dria isolated from pig brain in vitro (Hroudova and Fisar
pathway by controlling the release of cytochrome c from the 2010). Acute in vitro treatment with SSRIs, SNRIs, and
mitochondria, with pro-apoptotic Bcl-2 family proteins facilitat- MAOIs decreased mitochondrial bioenergetics (Klinedinst
ing release, and anti-apoptotic species inhibiting its release. In and Regenold 2015). We have outlined several potential
addition to cytochrome c, mitochondria contain several addition- mechanisms by which mitochondrial dysfunction or reduced
al pro-apoptotic species within their intermembrane space, which mitochondrial capacity could increase a patient’s susceptibili-
aid in the induction of apoptotic cell death in the event of Blc-2 ty to PTSD or exacerbate PTSD symptoms. In such cases, the
family protein mediated apoptosis induction or mitochondrial use of ADs with known mitochondrial interactions should be
damage (Mancini et al 1998; Krajewski et al 1999; Susin et al avoided, to avoid inducing further mitochondrial dysfunction.
1999; Du et al 2000; Verhagen et al 2000). Mitochondria are A mitochondrial dysfunction mechanism of PTSD suscepti-
very susceptible to oxidative stress: nitric oxide, lipid peroxida- bility has several implications in terms of novel PTSD preven-
tion, and ROS can lead to oxidative stress, which can result in tion and treatment. Recent developments in the metabolic pro-
loss of mitochondrial membrane potential, respiratory chain dis- filing have introduced the potential to screen for mitochondri-
ruption, and decreased ATP production (Kroemer and Reed al disease patients, and biomarkers for mitochondrial dysfunc-
2000; Brown 2003). Ca2+ influx in both pre-synaptic axon ter- tion may help identify PTSD-susceptible individuals in at-risk
minals and post-synaptic dendrites can induce significant oxida- populations (Thompson Legault et al 2015). Additionally, in-
tive and metabolic stress on mitochondria (Bezprozvanny and formation about an SMF patient’s metabolic profile could
Mattson 2008). This process of mitochondrial dysfunction and suggest personalized metabolic interventions and greatly im-
oxidative damage can be self-catalyzing, with oxidative stress prove recovery outcomes (Parr and Martin 2012). Lifestyle
disrupting mitochondrial function, which induces further oxida- and dietary interventions like increased exercise, caloric re-
tive stress. Patients with congenital or acquired mitochondrial striction, and cognitive enrichment, which have been shown
J Inherit Metab Dis (2018) 41:585–596 591

to improve mitochondrial capacity and mitochondrial health in determining whether a patient with mitochondrial dysfunc-
(Lopez-Lluch et al 2006, Lanza et al 2012, Eluamai and tion or SMF develops PTSD-like symptomatology rather than
Brooks 2013, Menshikova et al 2006), and which have been symptomatology consistent with anxiety or mood disorder.
shown to be neuro-protective (Maalouf et al 2009; Zigmond We have provided evidence that mitochondrial dysfunction
and Smeyne 2014; Milgram et al 2006), may offer alternative is associated with PTSD susceptibility, and have provided
therapeutic strategies which avoid drug-induced mitochondri- several mechanisms by which mitochondrial dysfunction
al inhibition or dysfunction. Increased physical activity is as- may contribute to the pathophysiology of PTSD. We have also
sociated with a decreased incidence of psychopathology, and cited mechanisms by which stress can induce mitochondrial
studies have shown that exercise can be used as a possible dysfunction, particularly in individuals who already display
treatment for anxiety and depression, though the mechanism reduced mitochondrial capacity. If mitochondrial capacity
is not fully understood (Strohle 2009). does in fact play a causal role in the psychopathology of
PTSD, and contributes to PTSD susceptibility, additional in-
vestigation will be required to determine whether this mito-
Outstanding questions chondrial dysfunction or suboptimal mitochondrial function is
congenital or acquired, and whether this mitochondrial dys-
Mitochondrial dysfunction and suboptimal mitochondrial function can be induced by the stress of a traumatic event
function have been increasingly implicated in several psycho- exposure. Both inborn and induced mitochondrial dysfunction
pathologies, and recent genetic studies have similarly sug- have been shown to induce susceptibility to anxiety and mood
gested a pathogenic role of mitochondria in PTSD. disorders, which share significant pathophysiology’s to
Mitochondria play a central role in several physiologic pro- PTSD. Pharmacologic inhibition of mitochondrial complex I
cesses underlying PTSD symptomatology, including abnor- and II has been shown to induce anxiety-like behavior in mice
mal fear learning, brain network activation, synaptic plasticity, (Hollis et al 2015), while mice harboring a defective mtDNA
steroidogenesis, and inflammation. Not surprisingly, PTSD polymerase transgene displayed increased depression-like
shares many of these pathophysiologic features with other behavior.
psychopathologies associated with mitochondrial dysfunc- Mitochondrial dysfunction is increasingly implicated in
tion. Anxiety disorders have been associated with abnormal psychiatric disease, however, not all mitochondrial disease
fear learning, inappropriate neuronal activation (viz. in the patients develop psychiatric symptoms. What is the biologic
amygdala, medial prefrontal cortex, and hippocampus), and mechanism by which a mitochondrial dysfunction increases
HPA axis dysregulation (Craske et al 2017). There is growing susceptibility to PTSD in a particular individual? Does mito-
evidence that abnormal inflammatory signaling secondary to chondrial dysfunction play a role in any or all previously
mitochondrial ROS production plays a role in the pathophys- mentioned physiologic processes involved in PTSD patho-
iology of schizophrenia (Rajasekaran et al 2015). Given the physiology? If SMF plays a role in only some features of
large degree of overlap in pathophysiologic pathways among PTSD pathophysiology but not all, why does it play a role in
anxiety and mood disorders, it is perhaps not surprising that in the particular mechanisms in which it does and not in others?
a national comorbidity survey of 12-month DSM-IV disor- How does a particular mitochondrial dysfunction in a partic-
ders, PTSD showed a high comorbidity with all anxiety and ular individual interact with the environment in terms of a
mood disorders investigated (Kessler et al 2005). Ultimately, particular traumatic event or set of traumatic events and the
psychopathologies appear to precipitate from a complex inter- stress response mobilized to contribute to a particular feature
action of genetic, physiologic, and environmental factors. of PTSD pathophysiology to ultimately increase PTSD sus-
While further investigation will be required to elucidate exact- ceptibility? Is mitochondrial dysfunction sufficient to increase
ly how any one particular collection of these overlapping ge- susceptibility to PTSD in a patient, or is it one aspect in a
netic, physiologic, and environmental factors combines to pre- multi-hit disease model?
cipitate a particular psychopathology, current evidence sug-
gests that mitochondrial dysfunction plays a significant and
important role in vulnerability to PTSD, perhaps by some of Conclusion
the same pathways that lead to increased vulnerability to anx-
iety and mood disorders, as well as other psychopathologies. Here we have outlined several potential mechanisms by which
It may prove that traumatic event exposure, which plays a mitochondrial dysfunction or reduced mitochondrial capacity
unique role in the precipitation of PTSD as opposed to other may increase susceptibility to PTSD or exacerbate PTSD
psychopathologies, and abnormal physiologic processes asso- symptomatology. Specifically, inability to efficiently affect
ciated with response to that traumatic event exposure — such critical inhibitory neural circuits or affect long-term potentia-
as steroid hormone release and signaling — secondary to mi- tion through bioenergetic- and Ca2+ homeostasis-sensitive
tochondrial dysfunction, may prove to play a significant role GABA-ergic neurons, disrupted neurogenesis, cell
592 J Inherit Metab Dis (2018) 41:585–596

remodeling, and apoptosis in response to stress signaling, ex- Bezprozvanny I, Mattson MP (2008) Neuronal calcium mishandling and
the pathogenesis of Alzheimer’s disease. Trends Neurosci 31(9):
cessive or imbalanced pro-inflammatory signaling secondary
454–463. https://doi.org/10.1016/j.tins.2008.06.005
to inappropriate ROS production, and an inability to maintain Boles RG, Roe T, Senadheera D (1998) Mitochondrial DNA deletion
appropriate synthesis of glucocorticoid hormones, may singu- with Kearns Sayre syndrome in a child with Addison disease. Eur
larly, or in concert, affect PTSD disease etiology. However, J Pediatr 157(8):643–647
Voccoli V, Colombaioni L (2009) Mitochondrial remodeling in differen-
much research is still required to elucidate whether any or all
tiating neuroblasts. Brain Res 1252:15–29
of these mechanisms contribute to PTSD vulnerability in pa- Brady K, Pearlstein T, Asnis GM et al (2000) Efficacy and safety of
tients, how these individual mechanisms interact within a par- sertraline treatment of posttraumatic stress disorder: a randomized
ticular individual, with a particular mitochondrial dysfunction, controlled trial. JAMA 283:1837–1844
Bremner JD, Krystal JH, Southwick SM, Charney DS (1995) Functional
and in response to a particular traumatic event exposure, and
neuroanatomical correlates of the effects of stress on memory. J
whether those particular interactions reveal biomarkers for Trauma Stress 8:527–553
identifying susceptible patients, or personalized strategies for Broekman BFP, Olff M, Boer F (2007) The genetic background to PTSD.
treatment and/or prevention. Neurosci Behav Rev 31:348–362
Brown GC (2003) NO says yes to mitochondria. Science 299(5608):838–
839
Funding No funding was provided for this work.
Bruno C, Minetti C, Tang Y (1998) Primary adrenal insufficiency in a
child with a mitochondrial DNA deletion. J Inherit Metab Dis 21(2):
Compliance with ethical standards 155–161
Burkhalter J, Fiumelli H, Allaman I, Chatton JY, Martin JL (2003) Brain-
Conflict of interest None. derived neurotrophic factor stimulates energy metabolism in devel-
No IACUC approval was required for this work. oping cortical neurons. J Neurosci 23(23):8212–8220
Butler MP, O’Connor JJ, Moynagh PN (2004) Dissection of tumor-
necrosis factor-alpha inhibition of long-term potentiation (LTP) re-
veals a p38 mitogen-activated protein kinase-dependent mechanism
which maps to early-but not late-phase LTP. Neurosci 124:319–326
References Calabresi P, Gubellini P, Picconi B, Centonze D, Pisani A, Bonsi P,
Greengard P, Hipskind RA, Borrelli E, Bernardi G (2001)
Inhibition of mitochondrial complex II induces a long-term potenti-
Accardi MV, Daniels BA, Brown PMGE, Fritschy J-M, Tyagarajan SK, ation of NMDA-mediated synaptic excitation in the striatum requir-
Bowie D (2014) Mitochondrial reactive oxygen species regulate the ing endogenous dopamine. J Neurosci 21:5110–5120
strength of inhibitory GABA-mediated synaptic transmission. Nat Chada SR, Hollenbeck PJ (2004) Nerve growth factor signaling regulates
Commun 5:3168 motility and docking of axonal mitochondria. Curr Biol 14:1272–
Almeida OFX, Conde GI, Crochemore C, Demeneix BA, Fischer D, 1276
Hassan AHS, Meyer M, Holsboer F, Michaelidis TM (2017) Chan SL, Liu D, Kyriazis GA, Bagsiyao P, Ouyang X, Mattson MP
Subtle shifts in the ratio between pro- and antiapoptotic molecules (2006) Mitochondrial uncoupling protein-4 regulates calcium ho-
after activation of corticosteroid receptros decide neuronal fate. meostasis and sensitivity to store depletion-induced apoptosis in
FASEB J 14(5):779–790 neural cells. J Biol Chem 281:37391–37403
Artuch R, Pavia C, Playan A (1998) Multiple endocrine involvement in Chance B, Sies H, Boveris A (1979) Hydroperoxide metabolism in mam-
two pediatric patients with Kearns-Sayre syndrome. Horm Res 50 malian organs. Physiol Rev 59(3):527–605
(2):99–104 Charney DS (2004) Psychobiological mechanisms of resilience and vul-
Aupperle RL, Melrose AJ, Stein MB, Paulus MP (2012) Executive func- nerability: implications for successful adaptation to extreme stress.
tion and PTSD: disengaging from trauma. Neuropharmacology 62 Am J Psychiatry 161(2):195–216
(2):686–694 Chen GQ, Zhu J, Shi XG, Ni JH, Zhong HJ, Si GY, Jin XL, Tang W, Li
Baker DG, West SA, Nicholson WE, Ekhator NN, Kasckow JW, Hill KK, XS, Xong SM, Shen XZ, Sun GL, Ma J, Zhang P, Zhang TD, Gazin
Bruce AB, Orth DN (1999) Geracioti TD Jr. serial CSF C, Naoe T, Chen SJ, Wang ZY, Chen Z (1996) In vitro studies on
corticotropin-releasing hormone levels and adrenocortical activity cellular and moleular mechanisms of arsenic trioxid (As2O3) in the
in combat veterans with posttraumatic stress disorder. Am J treatment of acute promyelocytic leukemia: As2O3 induces NB4
Psychiatry 156(4):585–588 cell apoptosis with downregulation of Bcl-2 expression and modu-
Baxter KK, Uittenbogaard M, Yoon J, Chiaramello A (2009) The neuro- lation of PML-RAR alpha/PML proteins. Blood 88(3):1052–1061
genic basic helix–loop–helix transcription factor neuroD6 concom- Cheng A, Hou Y, Mattson MP (2010) Mitochondria and neuroplasticity.
itantly increases mitochondrial mass and regulates cytoskeletal or- ASN NEURO 2(5):e00045. https://doi.org/10.1042/AN20100019.
ganization in the early stages of neuronal differentiation. ASN Chihara T, Luginbuhl D, Luo L (2007) Cytoplasmic and mitochondrial
NEURO 1(4):art:e00016. https://doi.org/10.1042/AN20090036 protein translation in axonal and dendritic terminal arborization. Nat
Benjet C, Bromet E, Karam EG et al (2016) The epidemiology of trau- Neurosci 10:828–837
matic event exposure worldwide: results from the World Mental Chow J, Rahman J, Achermann JC, Dattani MT, Rahman S (2017)
health survey consortium. Psychol Med 46(2):327–343 Mitochondrial disease and endocrine dysfunction. Nat Rev
Bersani FS, Morley C, Lindqvist D, Epel ES, Picard M, Yehuda R, Flory Endocrinol 13:92–104
J, Bierer LM, Makotkine I, Abu-Amara D, Coy M, Reus VI, Lin J, Cowell RM, Blake KR, Russell JW (2007) Localization of the transcrip-
Blackburn E, Marmar C, Wolkowitz OM, Mellon SH (2015) tional coactivator PGC-1alpha to GABAergic neurons during mat-
Mitochondrial DNA copy number is reduced in male combat vet- uration of the rat brain. J Comp Neurol 502:1–18
erans with PTSD. Progress Neuro-Psychoparmacol Biol Psychiatry Craske MG, Stein MB, Elay TC, Milad MR, Holmes A, Rapee RM,
64:10–17 Wittchen HU (2017) Anxiety disorders. Nat Rev Disease Primers
3:17100
J Inherit Metab Dis (2018) 41:585–596 593

Cui H, Kong Y, Zhang H (2012) Oxidative stress, mitochondrial dysfunc- Guidarelli A, Cerioni L, Cantoni O (2007) Inhibition of complex III
tion, and aging. J Signal Transduct 2012:646354 promotes loss of Ca2+ dependence for mitochondrial superoxide
Curti C, Mingatto FE, Plizello AC, Galastri LO, Uyemura SA, Santos AC formation and permeability transition evoked by peroxynitrite. J
(1999) Fluoxetine interacts with the lipid bilayer of the inner mem- Cell Sci 120(Pt 11):1908–1914
brane of isolated rat brain mitochondria, inhibiting electron transport Gulyas AI, Buzsaki G, Freund TF, Hirase H (2006) Populations of hip-
and F1F0-ATPase activity. Mol Cell Biochem 199(1–2):103–109 pocampal inhibitory neurons express different levesl of cytochrome
Datson NA, van der Perk J, de Kloet ER, Vreugdenhil E (2001) c. Eur J Nerosci 23:2581–2594
Identification of corticosteroid-responsive genes in rat hippocampus Gunasekar PG, Kanthasamy AG, Borowitz JL, Isom GE (1995) NMDA
using serial analysis of gene expression. Eur J Neurosci 14:675–689 receptor activation produces a concurrent generation of nitric oxide
de Kloet ER, Joels M, Holsboer F (2005) Stress and the brain: from and reactive oxygen species: implication for cell death. J Neurochem
adaptation to disease. Nat Rev Neurosci 6(6):463–475 65:2016–2021
Dedov VN, Armati PJ, Roufogalis BD (2000) Three-dimensional orga- Hakem R, Hakem A, Duncan GS, Henderson JT, Woo M, Soengas MS,
nisation of mitochondrial clusters in regenerating dorsal root gangli- Elia A, de la Pompa JL, Kagi D, Khoo W, Potter J, Yoshida R,
on (DRG) neurons from neonatal rats: evidence for mobile mito- Kaufman SA, Lowe SW, Penninger JM, Mak TW (1998)
chondrial pools. J Peripher Nerv Syst 5(1):3–10 Differential requirement for caspase 9 in apoptotic pathways
Diamond DM, Bennett MC, Fleshner M, Rose GM (1992) Inverted-U in vivo. Cell 94(3):339–352
relationship between the level of peripheral corticosterone and the Harris JJ, Jolivet R, Attwell D (2012) Synaptic energy use and supply.
magnitude of hippocampal primed burst potentiation. Hippocampus Neuron 75:762–777
2:421–430 Hayes JP, VanElzakker MB, Shin LM (2012) Emotion and cognition
Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, interactions in PTSD: a review of neurocognitive and neuroimaging
Lanctot KLA (2010) Meta-analysis of cytokines in major depres- studies. Front Integr Neurosci 6:89
sion. Biol Psychiatry 67(5):446–457 Hendler T, Rotshtein P, Yeshurun Y, Weizmann T, Kahn I, Ben-Bashat D,
Du C, Fang M, Li Y, Li L, Wang X (2000) Smac, a mitochondrial protein Malach R, Bleich A (2003) Sensing the inveisible: differential sen-
that promotes cytochrome c-dependent caspase activation by elimi- sitivity of visual cortex and amygdala to traumatic context.
nating IAP inhibition. Cell 102(1):33–42 NeuroImage 19:587–600
Duchen MR (2000) Mitochondria and Ca2+ in cell physiology and path- Hroudova J, Fisar Z (2010) Activities of respiratory chain complexes and
ophysiology. Cell Calcium 28:339–348 citrate synthase influenced by pharmacologically different antide-
Dugan LL, Sensi SL, Canzoniero LM, Handran SD, Rothman SM, Lin pressants and mood stabilizers. Neuro Endocrinol Lett 31(3):336–
TS, Goldberg MP, Choi DW (1995) Mitochondrial production of 342
reactive oxygen species in cortical neurons following exposure to N-
Hu Z, Yuri K, Ozawa H, Lu H, Kawata M (1997) The in vivo time course
methyl-D-aspartate. J Neurosci 15(10):6377–6388
for elimination of adrenalectomy-induced apoptotic profiles from
Dunn AJ, Swiergiel AH, de Beaurepaire R (2005) Cytokines as mediators
the granule cell layer fo the rat hippocampus. J Neurosci 17(11):
of dperession: what can we learn from animal studies? Neurosci
3981–3989
Biobehav Rev 29(4–5):891–909
Huang YY, Kandel ER (1998) Postsynaptic induction and PKA-
Durroux T, Gallo-Payet N, Payet M (1991) Effects of adrenocorticotropin
dependent expression of LTP in the lateral amygdala. Neuron 21
on action potential and calcium currents in cultured rat and bovine
(1):169–178
glomerulosa cells. Endocrinology 129:2139–2147
Inohara N, Gourley TS, Carrio R, Muniz M, Merino J, Garcia I, Koseki T,
Eluamai A, Brooks K (2013) Effect of aerobic exercise on mitochondrial
Hu Y, Chen S, Diva NG (1998) A Bcl-2 homologue that binds
DNA and aging. J Exerc Sci Fit 11(1):1–5.
directly to Apaf-1 and induces BH3-independent cell death. J Biol
Fanselow MS, LeDoux JE (1999) Why we think plasticity underlying
Chem 273:32479–32486
Pavlovian fear conditioning occurs in the basolateral amygdala.
Neuron 23:229–232 Jou SH, Chiu NY, Liu CS (2009) Mitochondrial dysfunction and psychi-
Feleder C, Tseng KY, Calhoon GG, O'Donnell P (2010) Neonatal intra- atric disorders. Chang Gung Med J 32(4):370–379
hippocampal immune challenge alters dopamine modulation of pre- Juo P, Kuo CJ, Yuan J, Blenis J (1998) Essential requirement for caspase-
frontal cortical interneurons in adult rats. Biol Psychiatry 67(4):386– 8/FLICE in the initiation of the Fas-induced apoptotic cascade. Curr
392 Biol 8(18):1001–1008
Flaquer A, Baumbach C, Ladwig K-H et al (2015) Mitochondrial genetic Kageyama GH, Won-Riley MTT (1982) Histochemical localization of
variants identified to be associated with posttraumatic stress disor- cytochrome oxidase in the hippocampus: correlation with specific
der. Transl Psychiatry 5(3):e524. https://doi.org/10.1038/tp.2015.18 neuronal types and afferent pathways. Neuroscience 7:2337–2361
Flory JD, Yehuda R (2015) Comorbidity between post-traumatic stress Kann O, Kovacs R (2007) Mitochondria and neuronal activity. Am J
disorder and major depressive disorder: alternative explanations and Physiol Cell Physiol 292:C641–C657
treatment considerations. Dialogues Clin Neurosci 17(2):141–150 Kann O, Huchzermeyer C, Kovacs R, Wirtz S, Schuelke M (2011)
Francati V, Vermetten E, Bremner JD (2007) Functional neuroimaging Gamma oscillations in the hippocampus require high complex I
studies in posttraumatic stress disorder: review of current methods gene expression and strong functional performance of mitochondria.
and findings. Depression and anxiety. 24(3):202–218. https://doi. Brain 134:345–358
org/10.1002/da.20208 Kann O, Papageorgiou IE, Draguhn A (2014) Highly energized inhibitory
Friedman JR, Nunnari J (2014) Mitochondrial form and function. Nature interneurons are a central element for information processin gin
505(7483):335–343 cortical networks. J Cereb Blood Flow Metab 34:1270–1282
Frodl T, Amico F (2014) Is there an association between peripheral im- Karam EG, Friedman MJ, Hill ED et al (2014) Cumulative traumas and
mune markers and structural/functional neuroimaging findings? risk thresholds: 12-month PTSD in the WORLD MENTAL health
Prog Neuropsychoparmacol Biol Psychiatry 48:295–303 (WMH) SURVEYS. Depression and anxiety 31(2):130–142.
Gould E, Woolley CS, McEwen BS (1990) Short-term glucocorticoid https://doi.org/10.1002/da.22169
manipulations affect neuronal morphology and survival in the adult Kepp O, Galluzzi L, Martins I, Schlemmer F, Adjemian S, Michaud M,
rat dentate gyrus. Neuroscience 37:367–375 Sukkurwala AQ, Menger L, Zitvogel L, Kroemer G (2011)
Green DR, Reed JC (1998) Mitochondria and apoptosis. Science 281 Molecular determinants of immunogenic cell death elicited by anti-
(5381):1309–1312 cancer chemotherapy. Cancer Metastasis Rev 30(1):61–69
594 J Inherit Metab Dis (2018) 41:585–596

Kessler RC, Rose S, Koenen KC et al (2014) How well can post- Lu B, Wang KH, Nose A (2009) Molecular mechanisms underlying
traumatic stress disorder be predicted from pre-trauma risk factors? neural circuit formation. Curr Opin Neurobiol 19:162–167
An exploratory study in the WHO World Mental health Surveys. Lüscher C, Malenka RC (2012) NMDA receptor-dependent long-term
World Psychiatry 13(3):265–274 potentiation and long-term depression (LTP/LTD). Cold Spring
Kim JJ, Diamond DM (2002) The stressed hippocampus, synaptic plas- Harb Perspect Biol 4(6):a005710. https://doi.org/10.1101/
ticity and lost memories. Nat Rev Neurosci 3:453–462 cshperspect.a005710
Klinedinst NJ, Regenold WTA (2015) Mitochondrial bioenergetic basis Maalouf MA, Rho JM, Mattson MP (2009) The neuroprotective proper-
of depression. J Bioenerg Biomembr 47(1–2):155–171 ties of calorei restriction, the ketogenic diet, and ketone bodies.
Kojima I, Kojima K, Rasmussen II (1985) Characteristics of angiotensin Brain Res Rev 59(2):293–315
II, K+, and ACTH-induced calcium influx in adrenal glomerulosa MacGibbon GA, Lawlor PA, Sirimanne ES, Walton MR, Connor B,
cells. J Biol Chem 260:9171–9176 Young D, Williams C, Gluckman P, Faull RL, Hughes P,
Krajewski S, Krajewska M, Ellerby LM, Welsh K, Xie Z, Deveraux QL, Dragunow M (1997) Bax expression in mammalian neurons under-
Salvesen GS, Bredesen DE, Rosenthal RE, Fiskum G (1999) going apoptosis, and in Alzheimer's disease hippocampus. Brain
Release of caspase-9 from mitochondria during neuronal apoptosis Res 750(1–2):223–234
and cerebral ischemia. Proc Natl Acad Sci U S A 96:5752–5757 Mancini M, Nicholson DW, Roy S, Thornberry NA, Peterson EP,
Kroemer G, Reed JC (2000) Mitochondrial control of cell death. Nat Med Casciola-Rosen LA, Rosen A (1998) The caspase-3 precursor has
6:513–519 a cytosolic and mitochondrial distribution: implications for apopto-
Kuida K, Haydar TF, Kuan CY, Gu Y, Taya C, Karasuyama H, Su MS, tic signaling. J Cell Biol 140:1485–1495
Rakic P, Flavell RA (1998) Reduced apoptosis and cytochrome c- Marchi S, Giorgi C, Suski JM, Agnoletto C, Bononi A, Bonora M, De
mediated caspase activation in mice lacking caspase 9. Cell 94(3): Marchi E, Missiroli S, Patergnani S, Poletti F, Rimessi A, Duszynski
325–337 J, Wiechowski MR, Pinton P (2012) Mitochondria-Ros crosstalk in
Lanza IR, Zabielski P, Klaus KA et al (2012) Chronic caloric restriction the control of cell death and aging. J Signal Transduct. 2012:329635
preserves mitochondrial function in senescence without increasing Markham A, Cameron I, Franklin P, Spedding MBDNF (2004) Increases
mitochondrial biogenesis. Cell Metab 16(6):777–788. https://doi. rat brain mitochondrial respiratory coupling at complex I, but not
org/10.1016/j.cmet.2012.11.003 complex II. Eur J Neurosci 20(5):1189–1196
LeDoux JE (1993) Emotional memory systems in the brain. Behav Brain Marshall RD, Beebe KL, Oldham M et al (2001) Efficacy and safety of
Res 58(1–2):69–79 paroxetine treatment for chronic PTSD: a fixed-dose, placebo-
Levkovitz Y, Gil-Ad I, Zeldich E, Dayag M, Weizman A (2005) controlled study. Am J Psychiatry 158:1982–1988
Differential induction of apoptosis by antidepressants in glioma Martin J-L, Finsterwald C (2011) Cooperation between BDNF and glu-
and neuroblastoma cell lines. J Mol Neurosci 27:29–42 tamate in the regulation of synaptic transmission and neuronal de-
Li Z, Okamoto K, Hayashi Y, Sheng M (2004) The importance of den- velopment. Commun Integrat Biol 4(1):14–16. https://doi.org/10.
dritic mitochondria in the morphogenesis and plasticity of spines 4161/cib.4.1.13761
and synapses. Cell 119:873–887 Mathew A, Lindsley TA, Sheridan A, Bhoiwala DL, Hushmendy SF,
Li Z, Jo J, Jia JM, Lo SC, Whitcomb DJ, Jiao S, Cho K, Sheng M (2010) Yager EJ, Ruggiero EA, Crawford DR (2012) Degraded mitochon-
Caspase-3 activation via mitochondria is required for long-term de- drial DNA Isa a newly identified subtype of the damage associated
pression and AMPA receptor internalization. Cell 141:859–871 molecular pattern (DAMP) family and possible trigger of neurode-
Li P, Zhao Y, Wu X et al (2012) Interferon gamma (IFN-γ) disrupts generation. J Alzheimers Dis 30(3):617–627
energy expenditure and metabolic homeostasis by suppressing Mattson MP, Liu D (2003) Mitochondrial potassium channels and
SIRT1 transcription. Nucleic Acids Res 40(4):1609–1620. https:// uncoupling proteins in synaptic plasticity and neuronal cell death.
doi.org/10.1093/nar/gkr984 Biochem Biophys Res Commun 304:539–549. https://doi.org/10.
Li H, Li X, Smerin SE et al (2014) Mitochondrial gene expression pro- 1016/S0006-291X(03)00627-2
files and metabolic pathways in the amygdala associated with exag- Mattson MP, Partin J (1999) Evidence for mitochondrial control of neu-
gerated fear in an animal model of PTSD. Front Neurol 5:164. ronal polarity. J Neurosci Res 56:8–20
https://doi.org/10.3389/fneur.2014.00164 McEwen BS, Wingfield JC (2003) The concept of allostasis in biology
Liberzon I, Abelson JL (2016) Context processing and the neurobiology and biomedicine. Horm Behav 43(1):2–15
of post-traumatic stress disorder. Neuron 92(1):14–30 Menshikova EV, Ritov VB, Fairfull L, Ferrell RE, Kelley DE,
Liberzon I, Taylor SF, Amdur R, Jung TD, Chamberlain KR, Minoshima Goodpaster BH (2006) Effects of exercise on mitochondrial content
S, Koeppe RA, Fig LM (1999) Brain activation in PTSD in response and function in aging human skeletal muscle. J Gerontol A Biol Sci
to trauma-related stimuli. Biol Psychiatry 45:817–826 Med Sci 61(6):534–540
Liu D, Chan SL, de Souza-Pinto NC, Slevin JR, Wersto RP, Zhan M, Michopoulos V, Rothbaum AO, Jovanovic T et al (2015) CRP genetic
Mustafa K, de Cabo R, Mattson MP (2006) Mitochondrial UCP4 variation and CRP levels are associated with increased PTSD symp-
mediates an adaptive shift in energy metabolism and increases the toms and physiological responses in a highly traumatized civilian
resistance of neurons to metabolic and oxidative stress. population. Am J Psychiatry 172(4):353–362. https://doi.org/10.
NeuroMolecular Med 8:389–414 1176/appi.ajp.2014.14020263.
Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J, Zhang CY, Mootha Middleton G, Nunez G, Davies AM (1996) Bax promotes neuronal sur-
VK, Jager S, Vianna CR, Reznick RM (2004) Defects in adaptive vival and antagonises the survival effects of neurotrophic factors.
energy metabolism with CNS-linked hyperactivity in PGC-1alpha Development 122(2):695–701
null mice. Cell 119:121–135 Milgram NW, Siwak-Tapp CT, Araujo J, Head E (2006) Neuroprotective
Lopez-Armada MJ, Riveiro-Naveira RR, Vaamonde-Garcia C, Valcarcel- effects of cognitive enrichment. Agein Res Rev 5(3):354–369
Ares MN (2013) Mitochondrial dysfunction and the inflammatory Miller WL (2013) Steroid hormone synthesis in mitochondria. Mol Cell
response. Mitochondrion 13(2):106–118 Endocrinol 379(1–2):62–73
Lopez-Lluch G, Hunt N, Jones B, Zhu M, Jamieson H, Hilmer S, Cascajo Murphy MP (2009) How mitochondria produce reactive oxygen species.
MV, Allard J, Ingram DK, Navas P, de Cabo R (2006) Calorie Biochem J 417(Pt 1):1–13
restriction induces mitochondrial biogenesis and bioenergectic effi- Myers KM, Davis M (2007) Mechanisms of fear extinction. Mol
ciency. Proc Natl Acad Sci U S A 103(6):1768–1773 Psychiatry 12(2):120–150
J Inherit Metab Dis (2018) 41:585–596 595

Nair SM, Karst H, Dumas T, Phillips R, Sapolsky RM, Rumpff-van Essen Reynolds IJ, Hastings TG (1995) Glutamate induces the production of
L, Maslam S, Lucassen PJ, Joels M (2004) Gene expression profiles reactive oxygen species in culture forebrain nerons following nmda
associated with survival of individual rat dentat cells after endoge- receptor activation. J Neurosci 15(5 Pt 1):3318–3327
nous corticosteroid deprivation. Eur J Neurosci 20(12):3233–3243 Rizzuto R, De Stefani D, Raffaello A, Mammucari C (2012)
Nakahira K, Haspel JA, Rathinam VA et al (2011) Autophagy proteins Mitochondria as sensors and regulators of calcium signalling. Nat
regulate innate immune response by inhibiting NALP3 Rev Mol Cell Biol 13:566–578
inflammasome-mediated mitochondrial DNA release. Nat Roberts AL, Gilman SE, Breslau J, Breslau N, Koenen KC (2011) Race/
Immunol 12(3):222–230. https://doi.org/10.1038/ni.1980. ethnic differences in exposure to traumatic events, development of
Nicholls DG, Budd SL (2000) Mitochondria and neuronal survival. post-traumatic stress disorder, and treatment-seeking for post-
Physiol Rev 80:315–360 traumatic stress disorder in the United States. Psychol Med 41:71–
Nicolino M, Ferlin T, Forest M (1997) Identification of a large-scale 83
mitochondrial deoxyribonucleic acid deletion in endocrinopathies Rubin DC, Feeling N (2013) Measuring the severity of negative and
and deafness: report of two unrelated cases with diabetes mellitus traumatic events. Clin Psychol Sci 1(4):375–389
and adrenal insufficiency, respectively. J Clin Endocrinol Metab 82 Ruthel G, Hollenbeck PJ (2003) Response of mitochondrial traffic to
(9):3063–3067 axon determination and differential branch growth. J Neurosci 23:
North K, Korson MS, Krawiecki N (1996) Oxidative phosphorylation 8618–8624
defect associated with primary adrenal insufficiency. J Pediatr 128 Shalev A, Liverzon I, Marmar C (2017) Post-traumatic stress disorder. N
(5 Pt 1):688–692 Engl J Med 376:2459–2469
Nutt DH, Malizia AL (2004) Structural and functional brain changes in Shin LM, Kozzlyn SM, McNally RJ, Alpert NM, Thmopson WL, Rauch
postraumatic stress diosrder. J Clin Psychiatry 65(Suppl 1):11–17 SL, Macklin ML, Pitman RK (1997) Visual imagery and perception
Olff M, Polak AR, Witteveen AB, Denys D (2014) Executive function in in posttraumatic stress disorder a positron emission tomographic
posttaumatic stress disorder (PTSD) and the influence of comorbid investigation. Arch Gen Psychiatry 54:233–241
depression. Neurobiol Learn Mem 112:114–121 Shin LM, Orr SP, Carson MA, Rauch SL, Macklin ML, Lasko NB, Peters
Overly CC, Rieff HI, Hollenbeck PJ (1996) Organelle motility and me- PM, Metzger LJ, Dougherty DD, Cannistraro PA, Alpert M,
tabolism in axons vs dendrites of cultured hippocampal neurons. J Fischman AJ, Pitman RK (2004) Regional cerebral blood flow in
Cell Sci 109:971–980 the amygdala and medial prefrontal cortex during traumatic imagery
Ozer EJ, Best SR, Lipsey TL, Weiss DS (2003) Predictors of posttrau- in male and female Vietnam beterans with PTSD. Arch Gen
matic stress disorder and symptoms in adults: a meta-analysis. Psychiatry 61:168–176
Psychol Bull 129(1):52–73 Shokolenko I, Venediktova N, Bochkareva A, Wilson BL, Alexeyev MF
Parr RL, Martin LH (2012) Mitochondrial and nuclear genomics and the (2009) Oxidative stress induces degradation of mitochondrial DNA.
emergence of personalized medicine. Human Genomics 6(1):3. Nucleic Acids Res 37(8):2539–2548
https://doi.org/10.1186/1479-7364-6-3 Shuttleworth CW, Brennan AM, Connor JA (2003) NAD(P)H fluores-
Pavlides C, Watanabe Y, McEwen BS (1993) Effects of glucocorticoids cence imaging of postsynaptic neuronal activation in murine hippo-
on hippocampal long-term potentiation. Hippocampus 3:183–19210 campal slices. J Neurosci 23:3196–3208
Peng TI, Jou MJ, Sheu SS, Greenamyre JT (1998) Visualization of Simpson ER, Waterman MR (1988) Regulation of the synthesis of ste-
NMDA receptor-induced mitochondrial calcium accumluation in roidogenic enzymes in adrenal cortical cells by ACTH. Annu Rev
striatal neurons. Exp Neurol 149(1):1–12 Physiol 50:427–424
Pervanidou P, Kolaitis G, Charitaki S, Lazaropoulou C, Papassotiriou I, Sloviter RS, Valiquette G, Abrams GM, Ronk EC, Sollas AI, Paul LA,
Hindmarsh P, Bakoula C, Tsiantis J, Chrousos GP (2007) The nat- Neubort SL (1989) Selective loss of hippocampal granule cells in
ural history of neuroendocrine changes in pediatric posttraumatic the mature rat brain after adrenalectomy. Science 243:535–538
stress disorder (PTSD) after motor vehicle accidents: progressive Smith SM, Vale WW (2006) The role of the hypothalamic-pituitary-
divergence of noradrenaline and cortisol concentrations over time. adrenal axis in neuroendocrine responses to stress. Dialogues Clin
Biol Psychiatry 62(10):1095–1102. https://doi.org/10.1016/j. Neurosci 8(4):383–395
biopsych.2007.02.008 Smorodchenko A, Rupprecht A, Sarilova I, Ninnemann O, Bräuer AU,
Picard M, Juster RP, McEwen BS (2014) Mitochondrial allostatic load Franke K, Schumacher S, Techritz S, Nitsch R, Schuelke M, Pohl
puts the "gluc" back in glucocorticoids. Nat Rev Endocrinol 10: EE (2009) Comparative analysis of uncoupling protein 4 distribu-
303–310 tion in various tissues under physiological conditions and during
Pissiota A, Frans O, Fernandez M, von Knorring L, Fisher H, Fredrikson development. Biochim Biophys Acta 1788:2309–2319
M (2002) Neurofunctional correlates of posttraumatic stress disor- Song Q, Kuang Y, Dixit VM, Vincenz C (1999) Boo, a novel negative
der; a PET symptom provocation study. Eur Arch Psychiatry Clin regulator of cell death, interacts with Apaf-1. EMBO J 18:167–178
Neurosci 252:68–75 Souza MEJ, Polizello ACM, Uyemur SA, Curti C (1994) Effect of flu-
Polak AR, Witteveen AB, Reitsma JB, Olff M (2012) The role of exec- oxetine on rat liver mitochondria. Biochem Pharmacol 48(3):535–
utive funciton in posttraumatic stress disorder: a systematic review. J 541
Affect Disord 141(1):11–21 Spoont MR, Williams JW, Kehle-Forbes S, Nieuwsma JA, Mann-Wrobel
Prigione A, Fauler B, Lurz R, Lehrach H, Adjaye J (2010) The MC, Gross R (2015) Does this patient have posttraumatic stress
senescence-related mitochondrial/oxidative stress pathway is re- disorder? Rational clinical examination systematic review. JAMA
pressed in human induced pluripotent stem cells. Stem Cells 28: 314(5):501–510. https://doi.org/10.1001/jama.2015.7877
721–733 Stress MEBS (1998) Adaptation, and disease: Allostasis and allostatic
Quirk GJ, Mueller D (2007) Neural mechanisms of extinction learning load. Ann N Y Acad Sci 840:33–44
and retreival. Neuropsychopharmacology 33(1):56–72 Strohle A (2009) Physical activity, exercise, depression and anxiety dis-
Rajasekaran A, Venkatasubramanian G, Berk M, Debnath M (2015) orders. J Neural Transm 116:777–784
Mitochondrial dysfunction in schizophrenia: pathways, mechanisms Strowig T, Henao-Mejia J, Elinav E, Flavell R (2012) Inflammasomes in
and implications. Neurosci Biobehav Rev 48:10–21 health and disease. Nature 481(7381):278–286
Reed JC (1997) Bcl-2 family proteins: regulators of apoptosis and Su YA, Wu J, Zhang L et al (2008) Dysregulated mitochondrial genes and
chemoresistance in hematologic malignancies. Semin Hematol 34 networks with drug targets in postmortem brain of patients with
(4 Suppl 5):9–19 posttraumatic stress disorder (PTSD) revealed by human
596 J Inherit Metab Dis (2018) 41:585–596

mitochondria-focused cDNA microarrays. Int J Biol Sci 4(4):223– von Kanel R, Hepp U, Kraemer B, Traber R, Keel M, Mica L, Schnyder
235 U (2007) Evidence for low grade systemic proinflammatory activity
Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, in patients with posttraumatic stress disorder. J Psychatric Res 41(9):
Mangion J, Jacotot E, Constantini P, Loeffler M, Larochette N, 744–752
Goodlett DR, Aebersold R, Siderovski DP, Penninger JM, Wanders RJA, Ruiter JPN, IJLst L, Waterham HR, Houten SM (2010)
Kroemer G (1999) Molecular characterization of mitochondrial The enzymology of mitochondrial fatty acid beta-oxidation and its
apoptosis-inducing factor. Nature 397(6718):441–446 application to followup analysis of positive neonatal screening re-
Taanman J-W (1999) The mitochondrial genome: structure, transcription, sults. J Inherit Metab Dis 33(5):479–494
translation, replication. Biochim Biophys Acta 1410(2):103–123 Wang L (2016) Mitochondrial purine and pyrimidine metabolism and
Tan S, Sagara Y, Liu Y, Maher P, Schubert D (1998) The regulation of beyond. Nucelosides Nucleotides Nucleic Acids 35(10–12):578–
reactive oxygen species production during programmed cell death. J 594
Cell Biol 141(6):1423 Wang C, Youle RJ (2016) The role of mitochondria in apoptosis. Annu
Tatton WG, Olanow CW (1999) Apoptosis in neurodegenerative dis- Rev Genet 43:95–118
eases: the role of mitochondria. Biochim Biophys Acta 1410(2): Wang C-U, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS Jr
195–213 (1998) NF-kB antiapoptosis: induction of TRAF1 and TRAF2 and
Telch MJ, Bruchey AK, Rosenfield D et al (2014) Post-session adminis- c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 281
tration of USP methylene blue facilitates the retention of patholog- (5383):1680–1683
ical fear extinction and contextual memory in phobic adults. Am J Whittaker RG, Turnbull DM, Whittington MA, Cunningham MO (2011)
Psychiatry 171(10):1091–1098. https://doi.org/10.1176/appi.ajp. Impaired mitochondrial function abolishes gamma oscillations in
2014.13101407. the hippocampus through an effect on fast-spiking interneurons.
Thompson Legault J, Strittmatter L, Tardif J et al (2015) A metabolic Brain 134:e180
signature of mitochondrial dysfunction revealed through a mono- Williams JM, Thompson VL, Mason-Parker SE, Abraham WC, Tate WP
genic form of Leigh syndrome. Cell Rep 13(5):981–989. https:// (1998) Synaptic activity-dependent modulation of mitochondrial
doi.org/10.1016/j.celrep.2015.09.054 gene expression in the rat hippocampus. Mol Brain Res 60:50–56
Toescu EC (2000) Mitochondria and Ca2+ signaling. J Cell Mol Med 4:
Wossink J, Karst H, Mayboroda O, Morphological JM (2001) Functional
164–175
properties of rat dentate granule cells after adrenalectomy.
Toresson H, Grant SG (2005) Dynamic distribution of endoplasmic retic-
Neuroscience 108(2):263–272
ulum in hippocampal neuron dendritic spines. Eur J Neurosci 22:
Xavier JM, Rodrigues CMP, Mitochondria SS (2015) Major regulators of
1793–1798
neural development. Neuroscientist 22(4):346–358
Tursich M, Neufeld RW, Frewen PA, Harricharan S, Kibler JL, Rhind SG
et al (2014) Association of trauma exposure with proinflammatory Xu L, Anwyl R, Rowan MJ (1997) Behavioural stress facilitates the
activity: a transdiagnostic meta-analysis. Transl Psychiatry 4:e413. induction of long-term depression in the hippocampus. Nature
https://doi.org/10.1038/tp.2014.56 387:497–500
Valcarcel-Ares MN, Riveiro-Naveira RR, Vaamonde-Garcia C, Lureiro J, Yamazaki T, Kimoto T, Higuchi K, Ohta Y, Kawato S, Kominami S (1998)
Hermida-Carballo L, Blanco FJ, Lopez-Armada MJ (2014) Calcium ion as a second messenger for o-nitrophenylsulfenyl-
Mitochondrial dysfunction promotes and aggravates the inflamma- adrenocorticotropin (NPS-ACTH) and ACTH in bovine adrenal ste-
tory response in normal human synoviocytes. Rheumatology 53: roidogenesis. Endocrinology 139(2):4765–4771
1332–1343 Yang P, Wu MT, Hsu CC, Ker JH (2004) Evidence of early neurobiolog-
VanElzakker MB, Dahlgren MK, Davis FC, Dubois S, Shin LM (2014) ical alternations in adolescents with posttraumatic stress disorder: a
From Pavlov to PTSD: the extinction of conditioned fear in rodents, functional MRI study. Neurosci Lett 370:13–18
humans, and in anxiety disorders. Neurobiol Learn Mem 113:3–18 Yi PL, Tsai CH, Lu MK, Liu HJ, Chen YC, Chang FC (2007) Interleukin-
Varfolomeev EE, Schuchmann M, Luria V, Chiannilkulchai N, 1beta mediates sleep alteration in rats with rotenone-induced parkin-
Beckmann JS, Mett IL, Rebrikov D, Brodianski VM, Kemper OC, sonism. Sleep 30(4):413–425
Kollet O, Lapidot T, Soffer D, Sobe T, Avraham KB, Goncharov T, Ying SW, Futter M, Rosenblum K, Webber MJ, Hunt SP, Bliss TV,
Holtmann H, Lonai P, Wallach D (1998) Targeted disruption of the Bramham CR (2002) Brain-derived neurotrophic factor induces
mouse caspase 8 gene ablates cell death induction by the TNF re- long-term potentiation in intact adult hippocampus: requirement
ceptors, Fas/Apo1, and DR3 and is lethal prenatally. Immunity 9(2): for ERK activation coupled to CREB and upregulation of arc syn-
267–276 thesis. J Neurosci 22(5):1532–1540
Verburg J, Hollenbeck PJ (2008) Mitochondrial membrane potential in Zhang L, Li H, Hu X et al (2015) Mitochondria-focused gene expression
axons increases with local nerve growth factor or semaphorin sig- profile reveals common pathways and CPT1B dysregulation in both
naling. J Neurosci 28:8306–8315 rodent stress model and human subjects with PTSD. Transl
Verhagen AM, Ekert PG, Pakush M, Silke J, Connolly LM, Reid GE, Psychiatry 5(6):e580. https://doi.org/10.1038/tp.2015.65
Moritz RL, Simpson RJ, Vaux DL (2000) Indentification of Zigmond MJ, Smeyne RJ (2014) Exercise: is it neuroprotective and if so,
DIABLO, a mammalian protein that promotes apoptosis by binding how does it work? Parkinsonism Relat Disord 20(Suppl 1):S123–
to an d antagonizing IAP proteins. Cell 102(1):43–53 S127
Reproduced with permission of copyright owner. Further reproduction
prohibited without permission.

You might also like