You are on page 1of 8

Brain, Behavior, and Immunity 25 (2011) 6–13

Contents lists available at ScienceDirect

Brain, Behavior, and Immunity


journal homepage: www.elsevier.com/locate/ybrbi

Invited Review

A short review on the psychoneuroimmunology of posttraumatic stress


disorder: From risk factors to medical comorbidities
Thaddeus W.W. Pace ⇑, Christine M. Heim
Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA

a r t i c l e i n f o a b s t r a c t

Article history: Posttraumatic stress disorder (PTSD) is a serious and debilitating condition with a prevalence rate of
Received 20 July 2010 approximately 8% in the United States. Given the number of veterans returning from conflicts around
Received in revised form 2 October 2010 the globe with PTSD, and the substantial number of civilians experiencing traumas, new perspectives
Accepted 4 October 2010
on the biology of PTSD are needed. Based on the concept that PTSD is a disorder of stress response sys-
Available online 8 October 2010
tems, numerous studies have suggested changes in hypothalamic–pituitary–adrenal (HPA) axis and sym-
pathetic–adrenal–medullary (SAM) system function in patients with PTSD. Given that both
Keywords:
glucocorticoids and catecholamines exert powerful effects on the immune system, it is surprising that
PTSD
Immune system
relatively few studies have examined immune changes in patients with PTSD. Moreover, patients with
Inflammation PTSD are known to have increased rates of comorbidity with somatic disorders that involve immune
Catecholamines and inflammatory processes. Patients with PTSD have been found to exhibit a number of immune
Glucocorticoids changes including increased circulating inflammatory markers, increased reactivity to antigen skin tests,
Neuroendocrine–immune interactions lower natural killer cell activity, and lower total T lymphocyte counts. Studies with humans and rodents
PTSD risk factors suggest that certain proinflammatory cytokines are able to induce neurochemical and behavioral changes
that resemble some key features of PTSD. This short article reviews immune alterations in PTSD, and con-
siders possible mechanisms by which such changes may be related to neuroendocrine alterations and
medical comorbidities of PTSD.
Ó 2010 Elsevier Inc. All rights reserved.

1. Introduction 2. History and central features of PTSD

Although neuroendocrine systems have been studied exten- PTSD was first introduced as a diagnosis in 1980. At the time,
sively in posttraumatic stress disorder (PTSD), relatively few pa- traumatic events sufficient to produce clinical PTSD symptoms
pers have explored immune alterations in patients with PTSD. were thought to be relatively rare (Vieweg et al., 2006). However,
Given that neuroendocrine systems are known to regulate immune subsequent investigations revealed that 50–90% of people in the
function, it is reasonable to predict that patients with PTSD may civilian population are exposed to events that may produce PTSD,
show immune changes. This expectation is bolstered by the fact and some 8% go on to develop the disorder in their lifetime (View-
that PTSD is often comorbid with medical disorders that involve eg et al., 2006; Yehuda and LeDoux, 2007). The nature of the trau-
inflammatory or autoimmune dysregulation. This short article dis- ma is important for understanding the risk for developing the
cusses studies that have indentified changes in inflammatory disorder, as PTSD may be more likely to develop after repeated
markers and lymphocyte function in patients with PTSD compared trauma such as torture or abuse (as high as 65%) compared to sin-
to controls. We also consider whether immune changes may play a gle incident exposures involving accidents such as car crashes and
role in the development and severity of PTSD symptoms as op- natural disasters (10–15%) (Bichescu et al., 2005). Traumas experi-
posed to being merely epiphenomena of PTSD. Possible mecha- enced early in life have also been shown to increase the risk for
nisms by which the described immune changes take place are developing PTSD following subsequent stressors (Keane et al.,
discussed. Finally, a psychoneuroimmunological model is pre- 2006). Finally, women appear to be more likely than men to devel-
sented in which immune alterations serve as mediators between op PTSD after traumatic events (Kessler et al., 1995).
PTSD and medical disease. PTSD is especially a concern for combat veterans or individuals
exposed to war. Although the exact numbers are a matter of de-
bate, between 1.4% and 31% of military personnel deployed to Iraq
⇑ Corresponding author. Address: Department of Psychiatry and Behavioral
Sciences, Emory University School of Medicine, 1365-B Clifton Rd., Rm B5105,
and Afghanistan develop PTSD upon returning home (Sundin et al.,
Atlanta, GA 30322, USA. Fax: +1 404 778 3965. 2010). This variability in prevalence rates likely stems from differ-
E-mail address: thaddeus.pace@emory.edu (T.W.W. Pace). ences in trauma exposure and differences in methodologies used to

0889-1591/$ - see front matter Ó 2010 Elsevier Inc. All rights reserved.
doi:10.1016/j.bbi.2010.10.003
T.W.W. Pace, C.M. Heim / Brain, Behavior, and Immunity 25 (2011) 6–13 7

study PTSD in military personnel. Differences in deployed popula- concentrations of cortisol are decreased (for a review, see Wessa
tions may also contribute to this variability in prevalence rates. and Rohleder, 2007; Heim and Nemeroff, 2009). Reduced cortisol
Individuals living with PTSD bear a large burden. Patients with levels in PTSD patients have been reported most often when the
PTSD are six times more likely to commit suicide, have increased hormone has been assessed in urine or saliva (Wessa and Rohleder,
risk of dropping out of high school or college, and are more likely 2007). The disagreement between indicators of HPA axis activity in
to experience marital instability (Kessler, 2000). Financial penalties central versus peripheral components suggests that endocrine
are significant, as work and family lives are severely interrupted. glands downstream of the hypothalamus are either downregu-
Indeed, the overall work impairment suffered by PTSD patients is lated, inhibited by glucocorticoid negative feedback, or insensitive
comparable to that found for individuals with major depression, to the increased activity of stress response systems in the brain.
highlighting the devastating nature of the disorder. In 2000, Kess- However, it must be noted that not all studies have reported lower
ler estimated that the annual loss in productivity because of PTSD circulating concentrations of cortisol in patients with PTSD (Mee-
in the United States was some $3 billion (Kessler, 2000), which wisse et al., 2007). This variability of results may be due to various
equates to $3.8 billion (EUR 2.9 billion) today. This estimate does factors including comorbid depression, current versus lifetime
not take into account the increased number of veterans suffering PTSD, severity of PTSD symptoms, time since the traumatic event,
from PTSD as a result of conflicts in Iraq and Afghanistan, which duration of trauma exposure, and whether or not control groups
would likely increase the annual loss in productivity. have a history of trauma (Wessa and Rohleder, 2007; Meewisse
The Diagnostic and Statistical Manual for Mental Disorders et al., 2007). It may also be more likely to find an association be-
(DSM)-IV requires that a person has been exposed to an extraordi- tween lower cortisol and PTSD when studies involve women, peo-
nary traumatic event in order to make a diagnosis of PTSD. Trau- ple with a history of physical or sexual abuse, or when samples are
matic events involve actual or threatened death or serious injury collected in the afternoon (Meewisse et al., 2007).
of the patient, or a threat to other persons such as friends and loved In addition to decreased cortisol at specific points over the
ones. The diagnosis also requires that key symptoms are present course of the day, patients with PTSD may exhibit a ‘‘flattening’’
from three distinct clusters: the first symptom cluster involves of the cortisol slope. More specifically, patients with PTSD have
reexperiencing the traumatic event with flashbacks, dreams, or been found to display less of a cortisol ‘‘spike’’ upon awakening
recollections, and/ or psychological or physical distress when and higher circulating concentrations of cortisol in the evening
encountering cues of the trauma. The second symptom cluster in- (Wessa and Rohleder, 2007). Interestingly, flattening of the daily
volves hypervigilance for danger and/or the expression of in- cortisol rhythm has been associated with a number of chronic
creased arousal manifested as difficulties sleeping, concentrating, medical illnesses including CVD and insulin resistance, as well as
and controlling anger. The third symptom cluster includes persis- other psychiatric disorders including major depression and chronic
tent avoidance of cues that evoke memories of the traumatic event. fatigue syndrome (e.g. Nater et al., 2008).
The avoidance cluster also includes features such as emotional With respect to HPA axis function, several studies suggest that
numbing, decreased interest in activities, a sense of a foreshort- PTSD patients may exhibit enhanced glucocorticoid sensitivity.
ened future, and a lack of positive emotions including love, con- This has been assessed by the low-dose dexamethasone suppres-
tentment, or happiness. sion test (DST). Yehuda and colleagues were the first to demon-
strate that patients with PTSD showed enhanced sensitivity to
glucocorticoids during the DST, especially when a low dose of
3. Neuroendocrine alterations in PTSD
dexamethasone was used (Yehuda, 2009). However, later studies
using the DST have found little or no differences between PTSD pa-
Over the last three decades, biological PTSD research has fo-
tients and trauma-exposed controls without PTSD (Wessa and
cused on two major hormone systems that form the backbone of
Rohleder, 2007). Thus, it is somewhat unclear if PTSD patients con-
the physiological response to stress: the hypothalamic–pituitary–
sistently display enhanced glucocorticoid sensitivity during the
adrenal (HPA) axis and the sympathetic–adrenal–medullary
DST. Heim and colleagues found lower cortisol levels during the
(SAM) system. Attention on these systems stemmed from the facts
DEX–CRH test in men with abuse-related PTSD, which is consistent
that (1) PTSD develops subsequent to exposure to a traumatic and
with the idea of enhanced feedback suppression under conditions
stressful event and (2) glucocorticoids and catecholamines are
of additional challenge (Heim et al., 2008).
‘‘stress hormones’’ that may regulate responses to trauma. In the
Glucocorticoid sensitivity can also be measured in circulating
1990’s, it became clear that PTSD is not a normative response to
immune cells. Several original reports suggest that circulating im-
extreme stressors, as only a subset of people exposed to trauma
mune cells from patients with PTSD may be more sensitive to glu-
go onto develop the disorder. Yehuda therefore reconceptualized
cocorticoids compared to immune cells from healthy individuals
PTSD as a disorder of stress response systems, leading to maladap-
(reviewed in Rohleder et al., 2010). For example, lysozyme activity
tive responses and a failure to cope with the stressor (Yehuda,
has been found to be more sensitive to dexamethasone in bulk leu-
2009). A search of the literature reveals multiple papers document-
kocytes collected from PTSD patients compared to cells obtained
ing changes of the HPA axis and the SAM system in PTSD patients.
from healthy controls. In addition, lipopolysaccharide (LPS)-in-
Although a comprehensive discussion of HPA axis and SAM system
duced cytokine production from whole blood, a measure of mono-
changes in PTSD is beyond the scope of this review, we will briefly
cyte responsiveness, has been reported to be more sensitive to
consider the highpoints of the literature, in order to set the stage
dexamethasone in samples collected from PTSD patients compared
for considering the possible origins of immune changes in PTSD pa-
to samples collected from controls (Rohleder et al., 2010). How-
tients. Readers are referred to thorough reviews on the topic (e.g.
ever, not all findings point to enhanced glucocorticoid sensitivity
Wessa and Rohleder, 2007; Yehuda, 2009; Heim and Nemeroff,
in immune tissues from PTSD patients: de Kloet and colleagues
2009).
compared the sensitivity of various immune parameters to dexa-
methasone in cell samples collected from patients with PTSD, trau-
3.1. HPA axis alterations and glucocorticoid sensitivity in PTSD ma exposed controls, and controls not exposed to trauma (de Kloet
et al., 2007). While they found that the sensitivity of LPS-induced
While cerebrospinal fluid (CSF) levels of corticotropin cytokine production to dexamethasone did not vary as a function
releasing hormone (CRH) are increased in patients with PTSD of PTSD or trauma exposure, the sensitivity of phytohemagglutinin
(e.g. Bremner et al., 1997), many studies have found that peripheral (PHA)-induced T cell proliferation to dexamethasone was actually
8 T.W.W. Pace, C.M. Heim / Brain, Behavior, and Immunity 25 (2011) 6–13

reduced in samples collected from PTSD patients versus those ob- These results raised the intriguing possibility that neuroendo-
tained from controls with or without trauma exposure (de Kloet crine dysregulation may be a pre-existing risk factor that is present
et al., 2007). The disagreement between measures of glucocorticoid well before and independent of the traumatic event that elicits
sensitivity when using LPS-induced cytokines as an endpoint may PTSD. Several prospective studies have now assessed neuroendo-
be explained by differences in assay protocols or control conditions crine changes prior to trauma exposure. Indeed, increased PBMC
(de Kloet et al., 2007). The finding of decreased sensitivity of T cell glucocorticoid binding before deployment has been found to pre-
proliferation to glucocorticoids may not necessarily conflict with dict the development of PTSD 6 months after deployment in army
studies using LPS-induced cytokines or lysozyme activity end- personnel (van Zuiden et al., 2010). In addition, exaggerated startle
points, as each explores different aspects of immune function. In responses in police academy cadets, measured prospectively, have
other words, there is the intriguing possibility that PTSD involves also been found to predict the development of PTSD (Pole et al.,
decreased glucocorticoid sensitivity in some immune tissues and 2009). This suggests that healthy people who show enhanced star-
increased glucocorticoid sensitivity in others. tle responses are at increased risk for developing PTSD.
There is also a lack of clarity with respect to changes of GR den- Risk for PTSD may also be transmitted across generations. Low-
sity in immune cells as a correlate of PTSD. While a number of er plasma concentrations of cortisol have been found in children of
studies have reported increased expression of GR in circulating im- Holocaust survivors (who also displayed enhanced HPA axis sensi-
mune cells, other reports suggest that GR density may actually be tivity during the DST), and in babies from mothers who developed
decreased (for a review, see Gill et al., 2009). As with measures of PTSD after the World Trade Center attack in New York City (Yehu-
glucocorticoid sensitivity, this disagreement in GR expression in da, 2009).
patients with PTSD could involve clinical differences between stud-
ies regarding patient groups. Methodological difference could also
4. Comorbidity with somatic illness
be important, including different techniques to assess GR density
or expression levels, as well as assessments of these parameters
Chronic sympathetic activation in PTSD may have important
in different subpopulations of immune cells.
implications for the individual risk to develop cardiovascular dis-
ease (CVD) as well as other chronic medical illnesses. Indeed, pa-
3.2. SAM system alterations in PTSD
tients with PTSD have increased rates of comorbidity with CVD
and other chronic somatic conditions (Boscarino, 2004, 2008; Ki-
Central and peripheral concentrations of norepinephrine appear
bler, 2009). A recent study by Heppener and colleagues reported
to be elevated in patients with PTSD. One study found increased
that PTSD severity predicted the presence of metabolic syndrome
CSF concentrations of norepinephrine in veterans with the disorder
(Heppner et al., 2009). However, PTSD does not appear to be asso-
(Geracioti et al., 2001). And seven studies have found increased ur-
ciated with diabetes mellitus (Qureshi et al., 2009) or waist-to-hip
ine norepinephrine levels in PTSD patients exposed to various
ratio (Heppner et al., 2009). This suggests that PTSD may enhance
types of trauma including domestic abuse, childhood abuse, com-
the risk for insulin resistance, especially in the context of obesity.
bat, automobile accidents, and exposure to war as a refugee (re-
PTSD has also been linked to rheumatoid arthritis (Qureshi et al.,
viewed in Wessa and Rohleder, 2007). Urinary epinephrine
2009), independent of genetic and familial factors (Boscarino
concentrations are also increased in patients with PTSD (Wessa
et al. 2010). Other chronic medical conditions that have been asso-
and Rohleder, 2007). Only one study has attempted to measure
ciated with PTSD include psoriasis and thyroid disease (Boscarino,
norepinephrine levels in blood (Yehuda et al., 1998). Beyond cate-
2004). Interestingly, each of these chronic somatic disorders has
cholamines, studies investigating cardiovascular variables in PTSD
inflammatory or autoimmune underpinnings. It is therefore rea-
patients support the notion of increased SAM reactivity. Thus, pa-
sonable to suspect that patients with PTSD may display immune
tients with PTSD exhibit increased heart rate, blood pressure, and
alterations in addition to neuroendocrine changes.
tremor responses when presented with reminders of traumatic
events (Bedi and Arora, 2007).
5. Immune alterations in PTSD
3.3. Is neuroendocrine dysregulation a risk factor for the development
of PTSD? 5.1. Central and peripheral inflammatory markers

While most studies conceptualized neuroendocrine changes as In 1997, Spivak and colleagues were the first to measure plasma
correlates of the disease state of PTSD, several studies now suggest cytokine levels in PTSD patients. They discovered that compared to
that neuroendocrine dysregulation may in fact be a risk factor that healthy, non-trauma exposed controls, veterans with PTSD dis-
predicts the development of the disorder upon exposure to ex- played increased circulating concentrations of interleukin IL-1b
treme stress. Some studies have evaluated whether altered neuro- that were also correlated with the duration of PTSD symptoms
endocrine responses observed in the immediate aftermath of a (Spivak et al., 1997). This finding has been bolstered by subsequent
trauma predict PTSD. SAM and HPA axis responses to traumatic studies. Higher tumor necrosis factor (TNF)-a was found in PTSD
events have been shown to predict later development of PTSD. patients with mixed trauma experience versus non-PTSD controls
According to Yehuda’s model, PTSD develops because key aspects (von Känel et al., 2007). In addition, higher plasma IL-6 levels
of the biological response to trauma interfere with recovery after and soluble IL-6 receptors were found in victims of an automobile
the trauma ends (Yehuda, 2009). Indeed, low plasma concentra- accident or a hotel fire compared to healthy controls (Maes et al.,
tions of cortisol and elevated plasma catecholamines in the period 1999). Higher IL-6 levels have also been reported in patients with
of time immediately after trauma have been found to predict PTSD PTSD subsequent to myocardial infarction (MI) as compared to MI
development (for a review of original studies, see Yehuda, 2009). If patients who did not develop PTSD, when controlling for depres-
cortisol responses to trauma normally facilitate containment of sive symptoms (von Känel et al. 2010). And refugees with PTSD fol-
catecholamine responses to the same event, then reduced cortisol lowing Hurricane Katrina exhibited higher circulating IL-6
following a trauma may promote an excessive catecholamine re- concentrations compared to refugees without PTSD (Tucker et al.,
sponse. This enhanced sympathetic activity may amplify encoding 2010). Alterations in inflammatory markers are evident not just
of traumatic memories, which would encourage expression of key in the peripheral circulation, but in the central nervous system as
PTSD features (Yehuda, 2009). well. Hence, elevated IL-6 concentrations have been measured in
T.W.W. Pace, C.M. Heim / Brain, Behavior, and Immunity 25 (2011) 6–13 9

CSF of PTSD patients with combat trauma exposure compared to Proportions of natural killer (NK) cells and monocytes in the cir-
healthy controls (Baker et al., 2001). Given that IL-6 is thought to culation have also been examined in patients with PTSD using flow
drive C-reactive protein (CRP) production from the liver, several re- cytometry. Skarpa and colleagues found increased proportions of
cent studies have also examined CRP levels in patients with PTSD. CD16+ positive cells in PTSD patients who were prisoners of war
In one of these studies, PTSD patients were found to have a nearly (Skarpa et al., 2001). All other studies (e.g. Laudenslager et al.,
twofold higher chance of having elevated CRP levels, even after 1998) have failed to detect enumerative differences in NK cells be-
controlling for possible confounds including sex, age, and alcohol tween PTSD and control participants. Monocytes also appear un-
use (Spitzer et al., 2010). changed in patients with PTSD (Rohleder et al., 2004). Thus, it
Despite multiple reports of increased circulating and central remains unclear whether or not the number of circulating NK cells
nervous system cytokines in PTSD patients, two studies have found or monocytes is altered in patients with PTSD.
decreased inflammatory markers. Most recently, von Känel and
colleagues found lower CRP in patients with PTSD subsequent to 5.3. T and B cell function
MI (von Känel et al., 2007). Lower CRP was also found in Iraqi ref-
ugees with PTSD (Sondergaard et al., 2004). The lack of agreement The function of T and B cells has been examined in patients with
between these studies may involve unique participant characteris- PTSD using a number of techniques including vaccine response, la-
tics. For example, Sondergaard and colleagues noted that control tent virus antibody titers, delayed-type hypersensitivity (DTH)
participants in their study may have been more likely than PTSD tests, and cell proliferation assays.
patients to have infections (Sondergaard et al., 2004), leading to Only one study has examined vaccine responses in patients
higher circulating CRP concentrations in the control group. Results with PTSD. In that report, patients with combat-related PTSD did
from studies with MI patients should be interpreted carefully as not differ from healthy controls regarding antibody titers and sero-
well, given the complicated medical status of these individuals. conversion rates after influenza vaccination (Kosor Krnic et al.,
Finally, a recent report suggests that trauma exposure and PTSD 2007). This result suggests that B cell function is not impaired in
may instill epigenetic changes that impact long-term inflammatory PTSD. Interestingly, the same study found that patients with PTSD
function (Uddin et al., 2010). In that study, patients with PTSD had significantly lower numbers of T cells positive for human leu-
were found to exhibit a greater number of unmethylated genes re- kocyte antigen-A*0201-restricted influenza A haemagglutinin anti-
lated to innate immune and inflammatory function compared to gens before vaccination (Kosor Krnic et al., 2007), indicating that
healthy controls, which could eventually encourage the expression patients may have had greater pre-vaccination vulnerability to
of altered immune function and enhanced inflammatory activity. influenza infection.
Immune function related to latent virus infection in patients
with PTSD has also been examined by only one study. In that re-
5.2. Monocyte and natural killer cells port, physically or psychologically abused women with increased
PTSD symptoms (but who were not diagnosed with syndromal
Several studies have examined natural killer cell activity PTSD according to DSM-IV criteria) were compared to healthy con-
(NKCA) in patients with PTSD using in vitro approaches. On the trols. Physically abused women exhibited significantly lower her-
whole, it appears that NKCA is impaired in PTSD. The most recent pes simplex virus 1 (HSV-1) viral neutralization compared to
report in this area examined NKCA in Croatian veterans with com- healthy controls and psychologically abused women. The physi-
bat-related PTSD and found that patients had reduced NKCA com- cally abused women also tended to have lower HSV-1 specific anti-
pared to healthy controls (Gotovac et al., 2010). Earlier studies body levels (Garcia-Linares et al., 2004). This suggests that in vivo
have reported the same finding in patients with non-combat re- cellular immunity may be impaired in PTSD patients. A search of
lated PTSD, such as male workers with PTSD who were victims of the literature for studies on the relationship between PTSD and Ep-
an earthquake in Japan and residents with PTSD exposed to Hurri- stein–Barr antibody titers, another common latent virus, did not
cane Andrew (Inoue-Sakurai et al., 2000; Kawamura et al., 2001; yield any findings.
Ironson et al., 1997). In contrast, negative findings have been re- Several investigations have measured DTH responses in pa-
ported in a small sample of Vietnam veterans with PTSD versus tients with PTSD. The majority of these suggest that antigen-spe-
Vietnam veterans without PTSD (Laudenslager et al., 1998). How- cific T cell function is enhanced in PTSD. Altemus and colleagues
ever, in that study scores on the Beck Depression Inventory were found that women with PTSD who were victims of early life phys-
positively correlated with NKCA, suggesting that NKCA alterations ical and sexual abuse had enhanced DTH responses compared to
may have been more related to comorbid depression than to PTSD healthy controls (Altemus et al., 2006). Similarly, Boscarino and
in those patients. Chang reported enhanced DTH in PTSD patients that served during
In vitro mitogen-stimulated cytokine tests have also been con- the Vietnam War compared to healthy veterans who also served in
ducted in PTSD patients. Samples containing immune cells (most Vietnam (Boscarino and Chang, 1999). These studies suggest that
often whole blood) are removed from participants and treated with patients have enhanced T cell responses, and they may explain in-
a mitogen such as LPS. LPS stimulates cytokine production primar- creased rates of autoimmune conditions such as psoriasis or atopic
ily from monocytes. These tests provide information about im- dermatitis in individuals suffering from PTSD.
mune responsiveness, without exposing participants to mitogens. Besides DTH, T cell function has also been examined in PTSD pa-
Only three studies have been performed using this approach, and tients using in vitro mitogen-induced cytokine or proliferation as-
two have found increased production of proinflammatory cyto- says. These techniques involve treating whole blood or PBMCs with
kines in samples from PTSD patients. In the first, Rohleder and col- the T cell mitogen PHA, or more recently with anti-human CD3
leagues reported increased LPS-induced IL-6 and TNF-a production monoclonal antibody. On the whole it appears that PTSD patients
in whole blood collected from Bosnian war refugees with PTSD display attenuated T cell responses when using these in vitro tests.
compared to healthy controls (Rohleder et al., 2004). In the second For example, patients with non-combat related PTSD demon-
study, enhanced LPS-induced production of interferon (IFN)-gam- strated lower IFN-gamma and IL-4 responses to PHA (Kawamura
ma was found in domestic abuse victims with PTSD (Woods et al., 2001). In another study, women displaced by the Bosnian
et al., 2005). However, a study by de Kloet and colleagues did not war who had increased fear and anxiety features displayed re-
find enhanced cytokine responses after LPS challenge in samples duced PHA-induced lymphocyte proliferation compared to non-
from veterans with PTSD (de Kloet et al., 2007). displaced controls (Sabioncello et al., 2000). Finally, greater T cell
10 T.W.W. Pace, C.M. Heim / Brain, Behavior, and Immunity 25 (2011) 6–13

proliferation was found in PBMCs collected from refugees with 5.5. Are immune changes risk factors for the development of PTSD?
PTSD and stimulated with the anti-CD3 monoclonal antibody
(measured with a carboxyfluorescein succinimidyl ester [CFSE]- As discussed above, decreased concentrations of cortisol and in-
based assay), indicating enhanced T cell responsiveness (Sommers- creased sympathetic activation in the immediate aftermath of a
hof et al., 2009). Taken together, these studies suggest that patients trauma have been relatively consistently associated with the
with PTSD exhibit enhanced T cell function. development of PTSD. Interestingly, immune responses to a trauma
may also be a risk factor for developing subsequent PTSD. Pervan-
5.4. T and B cell counts idou and colleagues found that increased circulating concentra-
tions of IL-6 measured 24 hours after an automobile accident
Several studies have examined circulating lymphocytes num- predicted the development of PTSD 6 months later in children (Per-
bers in PTSD patients. Results are extremely mixed, and general vanidou et al., 2007). This is the first and only study to examine the
conclusions cannot be drawn. However, a brief review of the topic relationship between an immune endpoint at the time of trauma
is still warranted given recent interest in the role that T cells may and the later development of PTSD. To date no prospective studies
play in other psychiatric disorders, including major depression have examined immune variables recorded well before and sepa-
(Miller, 2010). In general, T cells may play important neuroprotec- rate from a traumatic event. Immune risk factors for PTSD may also
tive roles in situations of stress and inflammation. Along with func- be governed by genetic influences, as inflammatory responsiveness
tional changes described above, changes in T cell subsets may may be inherited (e.g. Danik and Ridker, 2007).
encourage the development and maintenance of psychiatric ill- Insight into how altered cytokine levels may be related to the
nesses like major depression (Miller, 2010), and possibly PTSD. development of PTSD is provided by a study by Reichenberg and
Total lymphocytes do not appear to be altered in patients with colleagues, in which participants were acutely treated with LPS.
PTSD, as measured in women with childhood sexual abuse-related Feelings of anxiety were increased in the LPS group compared to
PTSD (Wilson et al., 1999), in Croatian war veterans with PTSD vehicle controls, and the severity of anxiety was positively corre-
(Vidovic et al., 2007), in Bosnian refugees with PTSD (Rohleder lated with the magnitude of the TNF-a response to LPS treatment
et al., 2004), or in Dutch veterans with PTSD (de Kloet et al., (Reichenberg et al., 2001). These results suggest that proinflamma-
2007). These negative results with the lymphocyte population as tory cytokines may exert anxiogenic effects. Of note, LPS also led to
a whole encourage examination of lymphocytes subsets. an increase in both core body temperature and depressive features.
Findings from two studies suggest that B cell counts do not dif- The latter suggests that proinflammatory cytokines, or mitogens
fer as a function of PTSD. Specifically, B cells were not different be- that induce inflammatory responses, have the ability to induce a
tween Vietnam era veterans with and without PTSD (Boscarino and diverse set of behavioral changes, including anxiety. However, it
Chang, 1999), or traumatized Dutch veterans with and without is also important to point out that not all cytokines or mitogens in-
PTSD (de Kloet et al., 2007). duce behavioral changes.
Mixed results have been found for T cell counts. Vietnam veter- Studies with rodents suggest that cytokines may have actions in
ans with PTSD were found to have higher total T cells compared to the brain that promote changes in the neurocircuitry that is in-
veterans without PTSD (Boscarino, 2004; Boscarino and Chang, volved in the mediation of anxiety. IL-6 has been shown to increase
1999). However, fewer T cells were found in non-combat related dopamine levels in the hippocampus and prefrontal cortex in mice
PTSD (Kawamura et al., 2001). For T helper (CD4+) cells, one study (Zalcman et al., 1994). Change in these regions can have powerful
reported increased proportions in combat veterans with PTSD effects on the amygdala, which is central to the neurocircuitry of
(Boscarino and Chang, 1999), while others have reported decreased the fear response and overactive in PTSD (Yehuda and LeDoux,
proportions in patients with different types of trauma history 2007). Interestingly, the frequency and severity of PTSD symptoms
(Ironson et al., 1997; Kawamura et al., 2001). With respect to cyto- (reexperiencing, avoidance, and arousal) are correlated with circu-
toxic T (CD8+) cells, some studies have reported lower counts lating concentrations of TNF-a (von Känel et al., 2007). Taken to-
(Ironson et al., 1997; Kawamura et al., 2001), while others have gether, these results may suggest that cytokine actions are
found no differences between participants with and without PTSD causally linked to PTSD symptoms, possibly by way of limbic
(Altemus et al., 2006; de Kloet et al., 2007; Laudenslager et al., circuits.
1998; Vidovic et al., 2007; Wilson et al., 1999).
A recent article by Sommershof and colleagues suggested that it 5.6. The hippocampus and immune function in PTSD
may be important to consider T cell activation and differentiation
states when performing enumerative tests of T cells (Sommershof Multiple studies have reported reduced hippocampal volumes
et al., 2009). For this approach, T cells are categorized according to in patients with PTSD (Bremner et al., 2008). Twins without trauma
the lineage markers CD45RA and CCR7, which identifies naive T or PTSD have been found to have the same reduced hippocampal
cells. This assumes a model in which naive T cells (CD45RA+ volume as siblings with PTSD (Yehuda and LeDoux, 2007). Child-
CCR7+) become activated with antigen exposure, differentiate into hood adversity, a known risk factor for PTSD, has also been associ-
T memory cells, and then partly develop into effector/T cytolytic ated with reduced hippocampal volume (Stein et al., 1997;
cells. Vythilingam et al., 2002). This suggests that reduced hippocampal
Sommershof and colleagues found that compared to a healthy volume may be a preexisting risk factor for PTSD.
controls, war refugees with PTSD had reduced percentages of Interestingly, proinflammatory cytokines have been shown to
CD3+ naive T cells along with an increased percentage of central mediate chronic stress-induced impairments in hippocampal neu-
and effector memory T cells. Among CD8+ T cells, PTSD patients rogenesis (Goshen et al., 2008), and this effect has been shown to
displayed reduced naive and increased effector memory cells depend on NF-jB (Koo et al., 2010). Given that early life adversity
(Sommershof et al., 2009). CD4+ cells of all lineages were not sig- (i.e. childhood abuse) has been associated with elevated circulating
nificantly altered between groups. PTSD patients also had reduced concentrations of CRP in childhood (Danese et al., 2010), it is
regulatory T cells compared to healthy participants. Along with possible that increased inflammation subsequent to childhood
functional alterations in patients with PTSD (i.e. enhanced re- adversity impairs neurogenesis via NF-jB at critical periods
sponses to anti-CD3 monoclonal antibody discussed above), these throughout development. This may ultimately encourage
shifts in T cells might be related to increased comorbidities with decreased hippocampal volume in adulthood. Reduced hippocam-
PTSD such as CVD and autoimmune disorders. pal volume may then promote malfunction of the fear response
T.W.W. Pace, C.M. Heim / Brain, Behavior, and Immunity 25 (2011) 6–13 11

circuit in context-dependent situations (Yehuda and LeDoux, systems may help to explain the inflammatory excess found in a
2007), with the eventual development of PTSD symptoms. portion of patients with PTSD.
Of course, neither early life adversity nor reduced hippocampal Because glucocorticoids are well known for their anti-inflam-
volume are prerequisites for PTSD, and many patients with the dis- matory effects, it is reasonable to theorize that reduced circulating
order will not have these risk factors. In addition, early life adver- concentrations of cortisol seen in some patients with PTSD may be
sity is not the only factor that may promote increased responsible for increased inflammation in the same individuals.
inflammation. For example, elevated circulating inflammatory While we are unaware of studies directly relating cortisol and
markers have also been associated with personality traits such as inflammatory markers in PTSD, attenuated cortisol responses have
hostility (e.g. Graham et al., 2006). Individual traits should there- been associated with enhanced IL-6 and IL-1b responses to stress
fore be considered along with adverse early life experiences when in healthy individuals (Kunz-Ebrecht et al., 2003). This suggests
attempting to understand the relationship between inflammation that low circulating concentrations of cortisol may foster a hyper-
and PTSD. inflammatory state, especially in the context of stress.
But the actions of hormones are not determined solely as a
6. Neuroendocrine–immune interactions in PTSD function of circulating concentrations. Instead, hormone effects
also depend on tissue sensitivity, which is determined by receptor
Neuroendocrine studies suggest that PTSD is not exclusively density and receptor function. As discussed above, PTSD patients
dependent on exposure to trauma. Instead, the onset and course have been found to exhibit altered glucocorticoid sensitivity in
of the disorder may involve a number of biological variables endocrine tissues and immune cells. In addition, multiple studies
including altered cortisol levels, enhanced glucocorticoid sensitiv- suggest that GR density may be increased on immune cells. Assum-
ity, altered GR numbers on immune cells, altered startle responses, ing that patients with PTSD exhibit enhanced glucocorticoid sensi-
and increased SAM system activity (Fig. 1). As discussed above, evi- tivity, especially of inflammatory processes, and higher GR density,
dence suggests that immune risk factors, and in particular the if anything patients with the disorder should have ‘‘normal’’ con-
inflammatory system, may play important roles as well. Thus, centrations of circulating inflammatory markers despite low corti-
atypical interactions between immune and neuroendocrine sol. One may even predict that patients with PTSD would have
reduced inflammatory markers. And yet multiple studies suggest
that PTSD involves enhanced inflammatory activation. How can
this apparent paradox be explained?
First, it is possible that enhanced inflammation in patients with
PTSD may not be the direct result of low circulating concentrations
of cortisol. Instead, increased inflammation may be driven by en-
hanced SAM system activity found in PTSD patients. Increased
sympathetic activity in patients with PTSD could be secondary to
reduced cortisol, especially in the context of stress challenge (Yeh-
uda, 2009). Increased SAM system activity may then enhance the
activity of inflammatory pathways, including NF-jB (Bierhaus
et al., 2003). With this possibility, low circulating concentrations
of cortisol would indirectly influence inflammatory function via
the SAM system.
Second, it also may be possible that the low circulating concen-
trations of cortisol found in a portion of patients with PTSD may
fall below critical thresholds needed to maintain normal inflamma-
tory function, even in the presence of enhanced glucocorticoid sen-
sitivity. If this hypothesis is correct, inflammatory excess in these
patients should respond rapidly to glucocorticoid therapy without
altering SAM system activity. Regardless of the exact nature of the
mechanisms involved, it is possible that reduced cortisol, enhanced
sympathetic activity, and enhanced inflammation influence one
another in the context of PTSD (Fig. 1).
It is also important to point out that inflammatory alterations in
patients with PTSD may develop independently of reduced cortisol
levels. As discussed above, increased inflammation may be a pre-
existing risk factor that results from personality traits or genetic
factors. If reduced cortisol is not involved, or if patients with PTSD
have normal circulating concentrations of cortisol (Meewisse et al.,
2007), it is possible that enhanced glucocorticoid sensitivity of im-
mune cells may be unable to overcome this inflammation.

7. From PTSD to medical comorbidities, via immune


Fig. 1. A psychoneuroimmunological model of PTSD. Psychosocial risk factors join
and synergize with biological risk factors for PTSD. Immune system changes dysfunction
identified in PTSD include increased delayed-type hypersensitivity (DTH),
decreased natural killer cell activity (NKCA), and increased in vitro lipopolysac- As discussed in Section 4, PTSD is often comorbid with a num-
charide (LPS)-induced cytokine responses. Major biological alterations already ber of chronic medical illnesses that have immune underpinnings.
known to exist in PTSD engage in a complex interplay, which is hypothesized to
contribute to immune alterations found in the disorder. Immune changes associ-
Could inflammatory and autoimmune changes that appear with
ated with PTSD are hypothesized to increase the risk for developing medical the onset of PTSD encourage later development of the same comor-
illnesses. bid medical illnesses? Although short-lived inflammation may be
12 T.W.W. Pace, C.M. Heim / Brain, Behavior, and Immunity 25 (2011) 6–13

beneficial for combating pathogen exposure or tissue damage, that immune changes subsequent to trauma and the onset of PTSD
long-term inflammation is well known to increase the risk for predict disease development. As mentioned above, one prospective
chronic medical illnesses. In this section, we will briefly illustrate study has already found that PTSD predicts the development of
a psychoneuroimmunological model of PTSD (Fig. 1), by focusing heart disease (Boscarino, 2008).
on two disease states that are often comorbid with the disorder:
CVD and insulin resistance.
Connections between PTSD, immune function and CVD may be- 8. Conclusions
gin with increased concentrations of proinflammatory cytokines in
the circulation that appear with the onset of PTSD. Circulating PTSD is a debilitating disorder that strikes some, but not all peo-
cytokine changes then persist for months, and perhaps years, along ple after exposure to extreme stress. The pathophysiological
with ongoing PTSD symptoms. Although prospective studies are underpinnings of the disorder are complicated. Moreover, as with
yet to demonstrate that the onset of PTSD symptoms is associated other psychiatric disorders, it is very likely that not all cases of
with increased circulating inflammatory markers, as noted above PTSD share the same underlying mechanistic ‘‘nuts and bolts’’. This
increased plasma levels of IL-6 at the time of trauma have been complicates attempts to understand the disorder, as well as efforts
associated with later development of PTSD (Pervanidou et al., to develop new treatment approaches. Although research on the
2007). In addition, acute and chronic stressors are well known to biology of PTSD has been historically dominated by studies with
increase inflammatory markers (Segerstrom and Miller, 2004). Of neuroendocrine endpoints, it is now clear that PTSD pathophysiol-
note, PTSD has already been prospectively associated with the ogy may also involve the immune system. Given the close relation-
development of heart disease 15 years later in veterans who were ship between the immune and neuroendocrine systems, as well as
free of heart disease at baseline (Boscarino, 2008). how immune alterations are thought to play a central role in other
Along with increased lipoproteins, inflammation within the neuropsychiatric disorders, it is reasonable to hypothesize that the
wall of blood vessels fosters the atherosclerotic process (Sprague immune system plays a central role in PTSD pathophysiology as
and Khalil, 2009). Increased circulating concentrations of proin- well as in medical illnesses found along with PTSD.
flammatory cytokines associated with PTSD may eventually acti-
vate inflammatory signaling pathways (e.g. NF-kB) in vascular
References
cells that make up the vessel wall. Inflammatory pathway activa-
tion in these cells encourages increased cell adhesion, increased Altemus, M., Dhabhar, F.S., Yang, R., 2006. Immune function in PTSD. Ann. NY Acad.
vessel permeability, and apoptosis. Monocytes are then able to at- Sci. 1071, 167–183.
Baker, D.G., Ekhator, N.N., Kasckow, J.W., Hill, K.K., Zoumakis, E., Dashevsky, B.A.,
tach more readily to vessel walls, and eventually mature into mac-
Chrousos, G.P., Geracioti, T.D., 2001. Plasma and cerebrospinal fluid interleukin-
rophages. These macrophages then amplify the proinflammatory 6 concentrations in posttraumatic stress disorder. NeuroImmunoModulation 9,
message in the vascular wall by secreting additional IL-1b and 209–217.
TNF-alpha, as well as other factors, which furthers expression of Bedi, U.S., Arora, R., 2007. Cardiovascular manifestations of posttraumatic stress
disorder. J. Natl. Med. Assoc. 99, 642–649.
adhesion molecules such as vascular cell adhesion molecule-1. Bichescu, D., Schauer, M., Saleptsi, E., Neculau, A., Elbert, T., Neuner, F., 2005. Long-
Macrophages eventually become large foam cells, forming ‘‘fatty term consequences of traumatic experiences: an assessment of former political
streaks’’ inside vessels that can develop into atherosclerotic le- detainees in Romania. Clin. Pract. Epidemiol. Ment. Health 1, 17.
Bierhaus, A., Wolf, J., Andrassy, M., Rohleder, N., Humpert, P.M., Petrov, D., Ferstl, R.,
sions. Proinflammatory cytokines would also draw smooth muscle von Eynatten, M., Wendt, T., Rudofsky, G., Joswig, M., Morcos, M., Schwaninger,
cells to the lesion, forming fibrous capsules over these lesions. Fi- M., McEwen, B., Kirschbaum, C., Nawroth, P.P., 2003. A mechanism converting
nally, circulating proinflammatory cytokines have been shown to psychosocial stress into mononuclear cell activation. Proc. Natl Acad. Sci. USA
100, 1920–1925.
potentiate thromboses, or rupture of atherosclerotic lesions, lead- Boscarino, J.A., 2004. Posttraumatic stress disorder and physical illness: results from
ing to MI or ischemic stroke (Tousoulis et al., 2006). Although clinical and epidemiologic studies. Ann. NY Acad. Sci. 1032, 141–153.
CVD depends on a number of factors, including dietary habits, Boscarino, J.A., 2008. A prospective study of PTSD and early-age heart disease
mortality among Vietnam veterans: implications for surveillance and
smoking, and exercise, it is conceivable that increased inflamma- prevention. Psychosom. Med. 70, 668–676.
tion begun by trauma and PTSD may eventually push PTSD pa- Boscarino, J.A., Chang, J., 1999. Higher abnormal leukocyte and lymphocyte counts
tients into comorbid CVD. 20 years after exposure to severe stress: research and clinical implications.
Psychosom. Med. 61, 378–386.
Inflammation has also been implicated in the pathophysiology
Boscarino, J.A., Forsberg, C.W., Goldberg, J., 2010. A twin study of the association
of insulin resistance and metabolic syndrome. Multiple studies between PTSD symptoms and rheumatoid arthritis. Psychosom. Med. 72, 481–
have found that chronic activation of inflammatory signaling 486.
pathways can lead to obesity-related insulin resistance. In the psy- Bremner, J.D., Licinio, J., Darnell, A., Krystal, J.H., Owens, M.J., Southwick, S.M.,
Nemeroff, C.B., Charney, D.S., 1997. Elevated CSF corticotropin-releasing factor
choneuroimmunological model of PTSD, elevated circulating con- concentrations in posttraumatic stress disorder. Am. J. Psychiatry 154, 624–629.
centrations of proinflammatory cytokines associated with PTSD Bremner, J.D., Elzinga, B., Schmahl, C., Vermetten, E., 2008. Structural and functional
would potentiate activation of macrophages in adipose tissue and plasticity of the human brain in posttraumatic stress disorder. Prog. Brain Res.
167, 171–186.
the liver that are central to insulin resistance. These activated mac- Danese, A., Caspi, A., Williams, B., Ambler, A., Sugden, K., Mika, J., Werts, H.,
rophages are the main generators of inflammatory products, such Freeman, J., Pariante, C.M., Moffitt, T.E., Arseneault, L., 2010. Biological
as TNF-alpha, that go onto induce inflammatory signaling path- embedding of stress through inflammation processes in childhood. Mol.
Psychiatry, in press.
ways within insulin target cells. Increased inflammatory signaling Danik, J.S., Ridker, P.M., 2007. Genetic determinants of C-reactive protein. Curr.
activity within insulin target cells is then able to antagonize nor- Atheroscler. Rep. 9, 195–203.
mal insulin receptor signaling (Olefsky and Glass, 2010). Although de Kloet, C.S., Vermetten, E., Bikker, A., Meulman, E., Geuze, E., Kavelaars, A.,
Westenberg, H.G., Heijnen, C.J., 2007. Leukocyte glucocorticoid receptor
increased inflammation as a result of trauma and PTSD may not be expression and immunoregulation in veterans with and without post-
sufficient to induce insulin resistance alone, the model predicts traumatic stress disorder. Mol. Psychiatry 12, 443–453.
that inflammatory changes induced by PTSD would synergize with Garcia-Linares, M.I., Sanchez-Lorente, S., Coe, C.L., Martinez, M., 2004. Intimate male
partner violence impairs immune control over herpes simplex virus type 1 in
other factors, including waist-to-hip ratio and exercise habits, to
physically and psychologically abused women. Psychosom. Med. 66, 965–972.
encourage development of insulin resistance and metabolic Geracioti, T.D., Baker, D.G., Ekhator, N.N., West, S.A., Hill, K.K., Bruce, A.B., Schmidt,
syndrome. D., Rounds-Kugler, B., Yehuda, R., Keck Jr., P.E., Kasckow, J.W., 2001. CSF
Although work to date supports an immune component in norepinephrine concentrations in posttraumatic stress disorder. Am. J.
Psychiatry 158, 1227–1230.
mechanisms linking PTSD and chronic medical illness, more infor- Gill, J.M., Saligan, L., Woods, S., Page, G., 2009. PTSD is associated with an excess of
mation is needed. Prospective studies will need to demonstrate inflammatory immune activities. Perspect. Psychiatr. Care 45, 262–277.
T.W.W. Pace, C.M. Heim / Brain, Behavior, and Immunity 25 (2011) 6–13 13

Goshen, I., Kreisel, T., Ben-Menachem-Zidon, O., Licht, T., Weidenfeld, J., Ben-Hur, T., Rohleder, N., Wolf, J.M., Wolf, O.T., 2010. Glucocorticoid sensitivity of cognitive and
Yirmiya, R., 2008. Brain interleukin-1 mediates chronic stress-induced inflammatory processes in depression and posttraumatic stress disorder.
depression in mice via adrenocortical activation and hippocampal Neurosci. Biobehav. Rev. 35, 104–114.
neurogenesis suppression. Mol. Psychiatry 13, 717–728. Sabioncello, A., Kocijan-Hercigonja, D., Rabatic, S., Tomasic, J., Jeren, T., Matijevic, L.,
Gotovac, K., Vidovic, A., Vukusic, H., Krcmar, T., Sabioncello, A., Rabatic, S., Dekaris, Rijavec, M., Dekaris, D., 2000. Immune, endocrine, and psychological responses
D., 2010. Natural killer cell cytotoxicity and lymphocyte perforin expression in in civilians displaced by war. Psychosom. Med. 62, 502–508.
veterans with posttraumatic stress disorder. Prog. Neuropsychopharmacol. Biol. Segerstrom, S.C., Miller, G.E., 2004. Psychological stress and the human immune
Psychiatry 34, 597–604. system: a meta-analytic study of 30 years of inquiry. Psychol. Bull. 130, 601–630.
Graham, J.E., Robles, T.F., Kiecolt-Glaser, J.K., Malarkey, W.B., Bissell, M.G., Glaser, R., Skarpa, I., Rubesa, G., Moro, L., Manestar, D., Petrovecki, M., Rukavina, D., 2001.
2006. Hostility and pain are related to inflammation in older adults. Brain Changes of cytolytic cells and perforin expression in patients with
Behav. Immun. 20, 389–400. posttraumatic stress disorder. Croat. Med. J. 42, 551–555.
Heim, C., Mletzko, T., Purselle, D., Musselman, D.L., Nemeroff, C.B., 2008. The Sommershof, A., Aichinger, H., Engler, H., Adenauer, H., Catani, C., Boneberg, E.M.,
dexamethasone/corticotropin-releasing factor test in men with major Elbert, T., Groettrup, M., Kolassa, I.T., 2009. Substantial reduction of naive and
depression: role of childhood trauma. Biol. Psychiatry 63, 398–405. regulatory T cells following traumatic stress. Brain Behav. Immun. 23, 1117–
Heim, C., Nemeroff, C.B., 2009. Neurobiology of posttraumatic stress disorder. CNS 1124.
Spectr. 14, 13–24. Sondergaard, H.P., Hansson, L.O., Theorell, T., 2004. The inflammatory markers C-
Heppner, P.S., Crawford, E.F., Haji, U.A., Afari, N., Hauger, R.L., Dashevsky, B.A., Horn, reactive protein and serum amyloid A in refugees with and without
P.S., Nunnink, S.E., Baker, D.G., 2009. The association of posttraumatic stress posttraumatic stress disorder. Clin. Chim. Acta 342, 93–98.
disorder and metabolic syndrome: a study of increased health risk in veterans. Spitzer, C., Barnow, S., Volzke, H., Wallaschofski, H., John, U., Freyberger, H.J., Lowe,
BMC Med. 7, 1. B., Grabe, H.J., 2010. Association of posttraumatic stress disorder with low-
Inoue-Sakurai, C., Maruyama, S., Morimoto, K., 2000. Posttraumatic stress and grade elevation of C-reactive protein: evidence from the general population. J.
lifestyles are associated with natural killer cell activity in victims of the Psychiatr. Res. 44, 15–21.
Hanshin–Awaji earthquake in Japan. Prev. Med. 31, 467–473. Spivak, B., Shohat, B., Mester, R., Avraham, S., Gil-Ad, I., Bleich, A., Valevski, A.,
Ironson, G., Wynings, C., Schneiderman, N., Baum, A., Rodriguez, M., Greenwood, D., Weizman, A., 1997. Elevated levels of serum interleukin-1 beta in combat-
Benight, C., Antoni, M., LaPerriere, A., Huang, H.S., Klimas, N., Fletcher, M.A., related posttraumatic stress disorder. Biol. Psychiatry 42, 345–348.
1997. Posttraumatic stress symptoms, intrusive thoughts, loss, and immune Sprague, A.H., Khalil, R.A., 2009. Inflammatory cytokines in vascular dysfunction
function after Hurricane Andrew. Psychosom. Med. 59, 128–141. and vascular disease. Biochem. Pharmacol. 78, 539–552.
Kawamura, N., Kim, Y., Asukai, N., 2001. Suppression of cellular immunity in men with Stein, M.B., Koverola, C., Hanna, C., Torchia, M.G., McClarty, B., 1997. Hippocampal
a past history of posttraumatic stress disorder. Am. J. Psychiatry 158, 484–486. volume in women victimized by childhood sexual abuse. Psychol. Med. 27,
Keane, T.M., Marshall, A.D., Taft, C.T., 2006. Posttraumatic stress disorder: etiology, 951–959.
epidemiology, and treatment outcome. Annu. Rev. Clin. Psychol. 2, 161–197. Sundin, J., Fear, N.T., Iversen, A., Rona, R.J., Wessely, S., 2010. PTSD after deployment
Kessler, R.C., 2000. Posttraumatic stress disorder: the burden to the individual and to Iraq: conflicting rates, conflicting claims. Psychol. Med. 40, 367–382.
to society. J. Clin. Psychiatry 61 (Suppl. 5), 4–12. Discussion 13–14. Tousoulis, D., Antoniades, C., Koumallos, N., Stefanadis, C., 2006. Pro-inflammatory
Kessler, R.C., Sonnega, A., Bromet, E., Hughes, M., Nelson, C.B., 1995. Posttraumatic cytokines in acute coronary syndromes: from bench to bedside. Cytokine
stress disorder in the National Comorbidity Survey. Arch. Gen. Psychiatry 52, Growth Factor Rev. 17, 225–233.
1048–1060. Tucker, P., Jeon-Slaughter, H., Pfefferbaum, B., Khan, Q., Davis, N.J., 2010. Emotional
Kibler, J.L., 2009. Posttraumatic stress and cardiovascular disease risk. J. Trauma and biological stress measures in Katrina survivors relocated to Oklahoma. Am.
Dissociation 10, 135–150. J. Disaster. Med. 5, 113–125.
Koo, J.W., Russo, S.J., Ferguson, D., Nestler, E.J., Duman, R.S., 2010. Nuclear factor- Uddin, M., Aiello, A.E., Wildman, D.E., Koenen, K.C., Pawelec, G., de Los Santos, R.,
kappaB is a critical mediator of stress-impaired neurogenesis and depressive Goldmann, E., Galea, S., 2010. Epigenetic and immune function profiles
behavior. Proc. Natl. Acad. Sci. USA 107, 2669–2674. associated with posttraumatic stress disorder. Proc. Natl. Acad. Sci. USA 107,
Kosor Krnic, E., Gagro, A., Kozaric-Kovacic, D., Vilibic, M., Grubisic-Ilic, M., 9470–9475.
Folnegovic-Smalc, V., Drazenovic, V., Cecuk-Jelicic, E., Gjenero-Margan, I., van Zuiden, M., Heijnen, C.J., Geuze, E., Willemen, H., Maas, M., Vermetten, E.,
Kuzman, I., Jeren, T., Sabioncello, A., Kerhin-Brkljacic, V., Kaic, B., Markotic, A., Kavelaars, A., 2010. Increased glucocorticoid binding to peripheral blood
Gotovac, K., Rabatic, S., Mlinaric-Galinovic, G., Dekaris, D., 2007. Outcome of mononuclear cells before military deployment in individuals developing PTSD
influenza vaccination in combat-related post-traumatic stress disorder (PTSD) symptoms 6 months after deployment. Brain Behav. Immun. 24, S9–S10.
patients. Clin. Exp. Immunol. 149, 303–310. Vidovic, A., Vilibic, M., Sabioncello, A., Gotovac, K., Rabatic, S., Folnegovic-Smalc, V.,
Kunz-Ebrecht, S.R., Mohamed-Ali, V., Feldman, P.J., Kirschbaum, C., Steptoe, A., 2003. Dekaris, D., 2007. Circulating lymphocyte subsets, natural killer cell
Cortisol responses to mild psychological stress are inversely associated with cytotoxicity, and components of hypothalamic–pituitary–adrenal axis in
proinflammatory cytokines. Brain Behav. Immun. 17, 373–383. Croatian war veterans with posttraumatic stress disorder: cross-sectional
Laudenslager, M.L., Aasal, R., Adler, L., Berger, C.L., Montgomery, P.T., Sandberg, E., study. Croat. Med. J. 48, 198–206.
Wahlberg, L.J., Wilkins, R.T., Zweig, L., Reite, M.L., 1998. Elevated cytotoxicity in Vieweg, W.V., Julius, D.A., Fernandez, A., Beatty-Brooks, M., Hettema, J.M.,
combat veterans with long-term post-traumatic stress disorder: preliminary Pandurangi, A.K., 2006. Posttraumatic stress disorder: clinical features,
observations. Brain Behav. Immun. 12, 74–79. pathophysiology, and treatment. Am. J. Med. 119, 383–390.
Maes, M., Lin, A.H., Delmeire, L., Van Gastel, A., Kenis, G., De Jongh, R., Bosmans, E., von Känel, R., Begre, S., Abbas, C.C., Saner, H., Gander, M.L., Schmid, J.P., 2010.
1999. Elevated serum interleukin-6 (IL-6) and IL-6 receptor concentrations in Inflammatory biomarkers in patients with posttraumatic stress disorder caused
posttraumatic stress disorder following accidental man-made traumatic events. by myocardial infarction and the role of depressive symptoms.
Biol. Psychiatry 45, 833–839. NeuroImmunoModulation 17, 39–46.
Meewisse, M.L., Reitsma, J.B., de Vries, G.J., Gersons, B.P., Olff, M., 2007. Cortisol and von Känel, R., Hepp, U., Kraemer, B., Traber, R., Keel, M., Mica, L., Schnyder, U., 2007.
post-traumatic stress disorder in adults: systematic review and meta-analysis. Evidence for low-grade systemic proinflammatory activity in patients with
Br. J. Psychiatry 191, 387–392. posttraumatic stress disorder. J. Psychiatr. Res. 41, 744–752.
Miller, A.H., 2010. Depression and immunity: a role for T cells? Brain Behav. Immun. Vythilingam, M., Heim, C., Newport, J., Miller, A.H., Anderson, E., Bronen, R.,
24, 1–8. Brummer, M., Staib, L., Vermetten, E., Charney, D.S., Nemeroff, C.B., Bremner,
Nater, U.M., Youngblood, L.S., Jones, J.F., Unger, E.R., Miller, A.H., Reeves, W.C., Heim, J.D., 2002. Childhood trauma associated with smaller hippocampal volume in
C., 2008. Alterations in diurnal salivary cortisol rhythm in a population-based women with major depression. Am. J. Psychiatry 159, 2072–2080.
sample of cases with chronic fatigue syndrome. Psychosom. Med. 70, 298–305. Wessa, M., Rohleder, N., 2007. Endocrine and inflammatory alterations in post-
Olefsky, J.M., Glass, C.K., 2010. Macrophages, inflammation, and insulin resistance. traumatic stress disorder. Expert Rev. Endocrinol. Metab. 2, 91–122.
Annu. Rev. Physiol. 72, 219–246. Wilson, S.N., van der Kolk, B., Burbridge, J., Fisler, R., Kradin, R., 1999. Phenotype of
Pervanidou, P., Kolaitis, G., Charitaki, S., Margeli, A., Ferentinos, S., Bakoula, C., blood lymphocytes in PTSD suggests chronic immune activation.
Lazaropoulou, C., Papassotiriou, I., Tsiantis, J., Chrousos, G.P., 2007. Elevated Psychosomatics 40, 222–225.
morning serum interleukin (IL)-6 or evening salivary cortisol concentrations Woods, A.B., Page, G.G., O’Campo, P., Pugh, L.C., Ford, D., Campbell, J.C., 2005. The
predict posttraumatic stress disorder in children and adolescents six months mediation effect of posttraumatic stress disorder symptoms on the relationship
after a motor vehicle accident. Psychoneuroendocrinology 32, 991–999. of intimate partner violence and IFN-gamma levels. Am. J. Community Psychol.
Pole, N., Neylan, T.C., Otte, C., Henn-Hasse, C., Metzler, T.J., Marmar, C.R., 2009. 36, 159–175.
Prospective prediction of posttraumatic stress disorder symptoms using fear Yehuda, R., 2009. Status of glucocorticoid alterations in post-traumatic stress
potentiated auditory startle responses. Biol. Psychiatry 65, 235–240. disorder. Ann. NY Acad. Sci. 1179, 56–69.
Qureshi, S.U., Pyne, J.M., Magruder, K.M., Schulz, P.E., Kunik, M.E., 2009. The link Yehuda, R., LeDoux, J., 2007. Response variation following trauma: a translational
between post-traumatic stress disorder and physical comorbidities: a neuroscience approach to understanding PTSD. Neuron 56, 19–32.
systematic review. Psychiatr. Q. 80, 87–97. Yehuda, R., Siever, L.J., Teicher, M.H., Levengood, R.A., Gerber, D.K., Schmeidler, J.,
Reichenberg, A., Yirmiya, R., Schuld, A., Kraus, T., Haack, M., Morag, A., Pollmacher, Yang, R.K., 1998. Plasma norepinephrine and 3-methoxy-4-
T., 2001. Cytokine-associated emotional and cognitive disturbances in humans. hydroxyphenylglycol concentrations and severity of depression in combat
Arch. Gen. Psychiatry 58, 445–452. posttraumatic stress disorder and major depressive disorder. Biol. Psychiatry
Rohleder, N., Joksimovic, L., Wolf, J.M., Kirschbaum, C., 2004. Hypocortisolism and 44, 56–63.
increased glucocorticoid sensitivity of pro-Inflammatory cytokine production in Zalcman, S., Green-Johnson, J.M., Murray, L., Nance, D.M., Dyck, D., Anisman, H.,
Bosnian war refugees with posttraumatic stress disorder. Biol. Psychiatry 55, Greenberg, A.H., 1994. Cytokine-specific central monoamine alterations
745–751. induced by interleukin-1, -2 and -6. Brain Res. 643, 40–49.

You might also like