PROOF COVER SHEET

advertisement
PROOF COVER SHEET
Author(s):
Boyd R. Rorabaugh, Anna Krivenko, Eric D. Eisenmann, Albert D. Bui, Sarah Seeley, Megan E. Fry,
Joseph D. Lawson, Lauren E. Stoner, Brandon L. Johnson, and Phillip R. Zoladz
Article title: Sex-dependent effects of chronic psychosocial stress on myocardial sensitivity to ischemic injury
Article no:
ISTS_A_1087505
Enclosures:
1) Query sheet
2) Article proofs
Dear Author,
Please check these proofs carefully. It is the responsibility of the corresponding author to check against the original
manuscript and approve or amend these proofs. A second proof is not normally provided. Informa Healthcare cannot be
held responsible for uncorrected errors, even if introduced during the composition process. The journal reserves the right
to charge for excessive author alterations, or for changes requested after the proofing stage has concluded.
The following queries have arisen during the editing of your manuscript and are marked in the margins of the proofs.
Unless advised otherwise, submit all corrections using the CATS online correction form. Once you have added all your
corrections, please ensure you press the ‘‘Submit All Corrections’’ button.
Please review the table of contributors below and confirm that the first and last names are structured correctly and that
the authors are listed in the correct order of contribution.
Contrib.
No.
Prefix
Given name(s)
Surname
1
Boyd R.
Rorabaugh
2
Anna
Krivenko
3
Eric D.
Eisenmann
4
Albert D.
Bui
5
Sarah
Seeley
6
Megan E.
Fry
7
Joseph D.
Lawson
8
Lauren E.
Stoner
9
Brandon L.
Johnson
10
Phillip R.
Zoladz
AUTHOR QUERIES
Q1:
Please check if author affiliation as set is correct.
Q2:
Please check whether the section headings are set in the correct hierarchy.
Suffix
Q3:
Please provide the town and state abbreviation (for the USA) or town and country of origin (for other countries)
identifying the headquarters location for ‘‘NIH Image J software’’.
Q4:
Please provide names of all authors (or the first seven and et al., if more than eight) instead of et al., for all the
references.
Please provide last page range.
Q5:
1
2
3
4
5
http://informahealthcare.com/sts
ISSN: 1025-3890 (print), 1607-8888 (electronic)
61
Stress, Early Online: 1–9
! 2015 Taylor & Francis. DOI: 10.3109/10253890.2015.1087505
63
62
64
65
6
7
66
ORIGINAL RESEARCH REPORT
67
8
9
10
11
12
13
14
68
Sex-dependent effects of chronic psychosocial stress on myocardial
sensitivity to ischemic injury
Boyd R. Rorabaugh1, Anna Krivenko2, Eric D. Eisenmann2, Albert D. Bui1, Sarah Seeley1, Megan E. Fry2,
Joseph D. Lawson2, Lauren E. Stoner2, Brandon L. Johnson2, and Phillip R. Zoladz2
15
70
71
72
73
74
75
Department of Pharmaceutical & Biomedical Sciences and 2Department of Psychology, Sociology & Criminal Justice, Ohio Northern University,
Ada,
OH, USA
17
76
18
78
16
Q1
69
1
77
19
Abstract
Keywords
20
21
Individuals with post-traumatic stress disorder (PTSD) experience many debilitating symptoms,
including intrusive memories, persistent anxiety and avoidance of trauma-related cues. PTSD
also results in numerous physiological complications, including increased risk for cardiovascular
disease (CVD). However, characterization of PTSD-induced cardiovascular alterations is lacking,
especially in preclinical models of the disorder. Thus, we examined the impact of a psychosocial
predator-based animal model of PTSD on myocardial sensitivity to ischemic injury. Male and
female Sprague–Dawley rats were exposed to psychosocial stress or control conditions for 31
days. Stressed rats were given two cat exposures, separated by a period of 10 days, and were
subjected to daily social instability throughout the paradigm. Control rats were handled daily
for the duration of the experiment. Rats were tested on the elevated plus maze (EPM) on day
32, and hearts were isolated on day 33 and subjected to 20 min ischemia and 2 h reperfusion on
a Langendorff isolated heart system. Stressed male and female rats gained less body weight
relative to controls, but only stressed males exhibited increased anxiety on the EPM. Male, but
not female, rats exposed to psychosocial stress exhibited significantly larger infarcts and
attenuated post-ischemic recovery of contractile function compared to controls. Our data
demonstrate that predator stress combined with daily social instability sex-dependently
increases myocardial sensitivity to ischemic injury. Thus, this manipulation may be useful for
studying potential mechanisms underlying cardiovascular alterations in PTSD, as well as sex
differences in the cardiovascular stress response.
Animal model, cardiovascular, heart, ischemia,
81
PTSD, stress
22
23
24
25
26
27
28
29
30
31
32
33
34
35
80
82
History
83
Received 18 June 2015
Revised 20 August 2015
Accepted 24 August 2015
Published online 2 2 2
84
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
85
86
87
88
89
90
91
92
93
94
95
96
97
Introduction
Traumatized individuals who develop post-traumatic stress
disorder (PTSD) are at increased risk for developing cardiovascular disease (CVD) (Boscarino, 2011; Buckley et al.,
2013; Coughlin, 2011; Edmondson & Cohen, 2013;
Edmondson et al., 2013). This association appears to be
independent of comorbid depression, genetic influences and
several other potentially confounding psychosocial factors
(Edmondson et al., 2013; Vaccarino et al., 2013). According
to Edmondson and colleagues (2013), the risk for CVD in
PTSD patients could be greater in females, relative to males,
but the limited number of CVD studies involving female
subjects has underpowered such statistical comparisons. The
scientific literature suggests that the link between PTSD and
CVD may be causal in nature and related to the numerous
cardiovascular abnormalities observed in PTSD patients. For
instance, people with PTSD exhibit lower heart rate variability (HRV), reduced baroreflex sensitivity and increased QT
57
Correspondence: Phillip R. Zoladz, Department of Psychology,
Sociology, & Criminal Justice, Ohio Northern University, 525 S. Main
59 St., Ada, OH 45810, USA. Tel: +1 419 772 2142.
60 E-mail: p-zoladz@onu.edu
58
79
variability (Cohen et al., 1998, 2000; Rozanski et al., 2005),
each of which has been linked to greater CVD incidence or
cardiovascular-related mortality (Bigger et al., 1992;
Piccirillo et al., 2007; Rozanski et al., 2005). PTSD patients
also demonstrate abnormally elevated heart rate (HR), blood
pressure (BP) and norepinephrine (NE) levels at baseline and
in response to emotional stimuli or events (for a review, see
Zoladz & Diamond, 2013). Coupled with reports of reduced
HRV and parasympathetic nervous system (PNS) activity in
PTSD patients, this exaggerated sympathetic nervous system
(SNS) activity suggests that PTSD causes autonomic
dysregulation.
CVD has been described as a disease of inflammation
(Libby et al., 2009), and PTSD patients exhibit elevated levels
of pro-inflammatory cytokines, such as tumor necrosis factor
a and IL-1b (von Kanel et al., 2007, 2010), as well as
increased incidence of autoimmune diseases (O’Donovan
et al., 2015). These observations may be associated with
baseline alterations of hypothalamus–pituitary–adrenal (HPA)
axis function in PTSD, as a common finding in PTSD patients
is abnormally low levels of baseline cortisol (Daskalakis
et al., 2013; Yehuda, 2009; Yehuda & Seckl, 2011). Because
corticosteroids act as major anti-inflammatory agents,
98
99
100
101
102
103
104
105
106
107
108
109
110
111
112
113
114
115
116
117
118
119
120
2
121
122
123
124
125
126
127
128
129
130
131
132
133
134
135
136
137
138
139
140
141
142
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
161
162
163
164
165
166
167
168
169
170
171
172
173
174
175
176
177
178
179
180
B. R. Rorabaugh et al.
insufficient HPA axis activity, coupled with exaggerated SNS
activity, could promote inflammation and negative health
outcomes.
One of the present authors was previously involved
in developing a psychosocial predator-based animal model
that has been shown to emulate several core symptoms of
PTSD (Roth et al., 2011; Zoladz et al., 2008, 2012, 2013,
2015). In the model, rats are immobilized and placed in close
proximity to a cat (i.e. predator stress) on two separate
occasions. The stressed rats are also exposed to daily
social instability (randomized changing of cage mates)
throughout the duration of the model to increase the
likelihood of producing long-lasting sequelae in the rats.
Thus, the model incorporates elements, such as uncontrollability, unpredictability, a lack of social interaction and a reexperiencing of the stress that, in people, significantly
increases the likelihood of PTSD onset and maintenance
following trauma exposure.
Previous work has shown that exposing rats to this
psychosocial stress manipulation results in a number of
physiological and behavioral abnormalities that are similar to
those observed in people with PTSD. Three weeks after the
second predator exposure, psychosocially stressed rats exhibit
a robust fear memory for the cat exposures (evidenced by
greater immobility in response to cat-associated context and
cues), heightened anxiety on an EPM (evidenced by less time
spent exploring open arms), an exaggerated startle response
and impaired memory for newly learned information (Zoladz
et al., 2008, 2012, 2013). The psychosocially stressed rats also
display physiological changes indicative of elevated SNS
activity and HPA axis abnormalities, such as greater cardiovascular and hormonal reactivity to an acute stressor, an
exaggerated physiological and behavioral response to the a2adrenergic receptor antagonist, yohimbine, abnormally low
levels of baseline corticosterone and enhanced negative
feedback of the HPA axis (Zoladz et al., 2008, 2012, 2013).
Others have replicated and extended the findings from this
model, revealing that the chronic psychosocial stress results in
decreased serotonin, increased norepinephrine and increased
measures of oxidative stress and inflammation in the brain,
adrenal glands and systemic circulation (Wilson et al., 2013,
2014a). More recently, research has shown that some effects
induced by the psychosocial stress manipulation persist for
four months following the initiation of stress (Zoladz et al.,
2015).
There has been limited research investigating the cardiovascular consequences of animal models of PTSD and none
have assessed whether a rodent model of PTSD could
influence myocardial ischemic injury. There have also been
few animal models of PTSD that include females, despite the
fact that females are reportedly more likely to develop the
disorder (Tolin & Foa, 2006). Based on previous work
demonstrating cardiovascular-related abnormalities and
increased inflammatory markers in male rats exposed to the
psychosocial predator-based animal model of PTSD, we
examined whether male and female rats exposed to this
psychosocial stress manipulation would exhibit greater myocardial sensitivity to ischemic injury and whether female rats
would demonstrate the anxiogenic and physiological effects
previously observed in males.
Stress, Early Online: 1–9
Methods and materials
Animals
Experimentally naı̈ve adult male and female Sprague–Dawley
rats (approximately 200–300 g at the beginning of the experiment) part of an established breeding colony (established from
Sprague–Dawley rats that were obtained from Charles River
laboratories) were used in the present experiment. The rats
were housed on a 12-h light/dark schedule (lights on at 07:00)
in standard Plexiglas cages (two per cage) with free access to
food and water. All procedures were approved by the
Institutional Animal Care and Use Committee at Ohio
Northern University and followed those recommended by the
Guide for the Care and Use of Laboratory Animals provided by
the National Institute of Health.
181
182
183
184
185
186
187
188
189
190
191
192
193
194
195
196
Psychosocial stress procedure
197
Rats were assigned to ‘‘psychosocial stress’’ (males: N ¼ 10;
females: N ¼ 14) or ‘‘no stress’’ (males: N ¼ 11; females:
N ¼ 10) groups. On day 1, rats in the psychosocial stress
groups were immobilized in plastic DecapiCones (Braintree
Scientific; Braintree, MA) and placed in a perforated wedgeshaped Plexiglas enclosure (Braintree Scientific; Braintree,
MA; 20 20 8 cm). This enclosure was then taken to a cat
housing room and placed in a metal cage (61 53 51 cm)
with an adult female cat for 1 h. The Plexiglas enclosure
prevented any contact between the cat and rats, but the rats
were still exposed to all non-tactile sensory stimuli associated
with the cat. Canned cat food was smeared on top of the
enclosure to direct the cat’s attention toward the rats. An hour
later, the rats were returned to their home cages. Rats in the
no stress groups remained in their home cages during the 1-h
stress period.
Rats in the psychosocial stress groups were given two acute
stress sessions, which were separated by 10 days. The first
stress session took place during the light cycle (between 08:00
and 13:00), and the second stress session took place during the
dark cycle (between 19:00 and 21:00). The stress sessions took
place during different times of the day to add an element of
unpredictability as to when the rats might re-experience the
predator exposure, as per previously-employed methodology
(Roth et al., 2011; Zoladz et al., 2008, 2012, 2013, 2015).
198
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
215
216
217
218
219
220
221
222
223
Daily social stress
Beginning on the day of the first stress session (day 1), rats in
the psychosocial stress groups were exposed to unstable housing
conditions until behavioral testing (day 32). Rats in the
psychosocial stress groups were housed two per cage, but
every day, their cohort pair combinations were changed.
Therefore, no rats in the psychosocial stress groups had the
same cage mate on two consecutive days during the 31-day
stress period. Rats in the no stress groups were housed with the
same cohort pair throughout the experiment; these rats were
handled daily to control for handling effects on stressed animals.
Behavioral testing
224
225
226
227
228
229
230
231
232
233
234
235
236
237
Three weeks following the second stress session (day 32), rats 238
were tested on the EPM to assess anxiety-like behavior. Rats 239
were placed on the EPM for 5 min, and their behavior was 240
Cardiac injury in animal model of PTSD
DOI: 10.3109/10253890.2015.1087505
241
242
243
244
245
246
247
videotaped by a JVC hard disk camera hanging above the
EPM and scored offline by two separate investigators who
were blind to the experimental conditions of the animals.
Investigators assessed time spent in the open arms and
number of entries into the open and closed arms. Rats were
removed from data analysis if they fell off of the EPM during
testing.
248
Langendorff heart preparation and measurement of
250 cardiac function
249
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
272
273
274
275
276
277
278
279
280
Q2
281
Measurement of infarct size
282
283
284
285
286
287
288
289
290
291
Q3
On Day 33, rats were anesthetized with an intraperitoneal
injection of pentobarbital (75 mg/kg). Hearts were rapidly
removed and cannulated while bathed in ice-cold Krebs–
Henseleit solution (118 mM NaCl, 4.7 mM KCl, 1.2 mM
MgSO4, 25 mM NaHCO3, 1.2 mM KH2PO4, 0.5 mM
Na2EDTA, 11 mM glucose, 2.5 mM CaCl2, pH 7.4). Krebs
solution was perfused through the aortic cannula at a constant
pressure of 80 mmHg. Contractile function of the left
ventricle was measured using an intraventricular balloon
connected to a pressure transducer. The balloon was inflated
to an end diastolic pressure of 4–8 mmHg, and data were
continually recorded using a Powerlab 4SP data acquisition
system (AD Instruments, Colorado Springs, CO). Hearts were
equilibrated for 25 min prior to the onset of 20 min ischemia
and 2 h reperfusion. Hearts were excluded after the 25-min
equilibration period if developed pressure was less than
100 mmHg, coronary flow rate was greater than 20 ml/min or
if they had a persistent irregular rhythm. Ischemia was
induced by closing a valve which stopped the flow of Krebs
solution through the aortic cannula. Reperfusion was initiated
by reopening the valve and allowing Krebs solution to resume
flow through the cannula (Granville et al., 2004; Waterson
et al., 2011). The heart was submerged in Krebs solution to
maintain the temperature of the heart at 37.5 ± 0.5 C
throughout the experiment, and temperature was continuously
monitored using a thermocouple placed on the surface of the
heart. Pre-ischemic contractile function was measured immediately prior to ischemia. Post-ischemic recovery of contractile function was measured following 1 h of reperfusion.
292
293
294
295
296
297
Following 20 min ischemia and 2 h reperfusion, hearts were
perfused with 1% triphenyltetrazolium chloride for 8 min (at a
rate of 7.5 ml/min) and then submerged in 1% triphenyltetrazolium chloride and incubated at 37 C for 15 min. Hearts
were subsequently frozen at 80 C, sliced into approximately 1 mm sections, soaked in 10% neutral buffered
formalin and then photographed with a Nikon SMZ 800
microscope equipped with a Nikon DS-Fi1 digital camera.
The infarcted surface area and total surface area of each slice
were measured using NIH Image J software. Infarct size was
expressed as a percentage of the area at risk (AAR). The AAR
was defined as the entire ventricular myocardium since hearts
were exposed to global ischemia.
Body and organ weights
Rats were weighed on the days of the first and second stress
299 sessions, as well as on the day of EPM testing. Growth rates
300 (grams per day) were calculated for statistical analysis.
298
3
In females, adrenal and thymus glands were removed and
weighed to compare to previously-reported findings in
psychosocially stressed males. Organ weights were expressed
as mg/100 g body weight for statistical analysis.
301
Statistical analyses
306
Male and female data were analyzed separately. Most data
(body weight, EPM behavior, infarct sizes, adrenal/thymus
weights) were compared via independent samples t tests with
treatment (psychosocial stress, no psychosocial stress) serving
as the between-subjects factor. Parameters of contractile
function were compared with two-way, mixed-model
ANOVAs with treatment (psychosocial stress, no psychosocial stress) serving as the between-subjects factor and time
(pre-ischemic, post-ischemic) serving as the within-subjects
factor. Tukey post-hoc tests were employed when necessary,
and alpha was set at 0.05 for all analyses. Data points more
than three standard deviations beyond exclusive group means
were eliminated from statistical analyses.
307
Results
302
303
304
305
308
309
310
311
312
313
314
315
316
317
318
319
320
321
322
Preliminary characterization of psychosocial stressed
rats
323
Males
325
Psychosocially stressed males spent significantly less time in
the open arms of the EPM than control males [t(16) ¼ 2.17,
p ¼ 0.046; Figure 1A], indicating a heightened state of
anxiety. This effect was not attributable to reduced locomotor
activity in the psychosocially stressed males, as both groups
exhibited a statistically equivalent number of total arm entries
[t(16) ¼ 0.60, p ¼ 0.56; Figure 1B]. Psychosocial stress also
led to significantly reduced growth rate in male rats over the
31-day period [t(19) ¼ 2.39, p ¼ 0.028; Figure 1C].
Females
Examination of the physiological and behavioral data from
females revealed inconsistent findings. Although, psychosocially stressed females exhibited significantly reduced
growth rate over the 31-day period [t(22) ¼ 2.93, p ¼ 0.008;
Figure 2C], they spent a statistically equivalent amount of
time in the open arms of the EPM as control rats [t(20) ¼ 0.19,
p ¼ 0.86; Figure 2A], suggestive of no anxiogenic effect of
the psychosocial stress manipulation. Adrenal [t(22) ¼ 1.75,
p ¼ 0.09; Figure 2D] and thymus gland [t(20) ¼ 2.04,
p ¼ 0.055; Figure 2E] comparisons were not significant.
324
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
342
343
344
345
346
347
348
Effect of psychosocial stress on cardiac function and
myocardial ischemic injury
Males
Psychosocial stress had no effect on pre-ischemic contractile
function. Pre-ischemic rate pressure product, +dP/dT, dP/
dT and end diastolic pressure were similar in psychosocially
stressed males and controls (Figure 3). However, 20 min of
ischemia induced more myocardial injury in psychosocially
stressed males, relative to controls. Hearts from psychosocially stressed males exhibited significantly larger infarcts
compared to controls [t(15) ¼ 2.54, p ¼ 0.023; Figure 3A].
349
350
351
352
353
354
355
356
357
358
359
360
4
B. R. Rorabaugh et al.
Stress, Early Online: 1–9
361
421
362
422
363
423
364
365
424
425
366
426
367
427
368
428
369
429
370
430
371
431
372
373
432
433
374
434
375
435
376
436
377
437
378
438
379
439
380
381
440
441
382
442
383
443
384
444
385
445
386
446
387
447
388
389
Figure 1. Physiological and behavioral effects of psychosocial stress in male rats. Psychosocially stressed males spent significantly less time in the 448
open arms of the elevated plus maze (A), despite making an equivalent numbers of overall arm entries in the maze (B). The stressed males also 449
390 exhibited significantly reduced growth rates (C) relative to controls. Sample sizes were 9–11 rats per group. Data are presented as means ± SEM. 450
*p50.05 versus no stress.
391
451
392
452
393
394
395
396
397
398
399
400
401
402
403
404
405
406
407
408
409
410
411
412
413
414
415
416
417
In addition, post-ischemic recovery of contractile function
was significantly attenuated in the hearts from psychosocially
stressed males. Recovery of the rate pressure product
[significant Group Time interaction: F(1, 14) ¼ 5.38,
p ¼ 0.036; Figure
3B] and
+dP/dT [significant
Group Time interaction: F(1, 14) ¼ 5.90, p ¼ 0.029;
Figure 3C] was significantly reduced in psychosocially
stressed males, relative to controls, indicating that recovery
of systolic function was worsened by the psychosocial stress.
Recovery of dP/dT was decreased 33% in psychosocially
stressed male hearts compared to control hearts, but this did
not reach statistical significance [Group Time interaction:
F(1, 14) ¼ 3.24, p ¼ 0.094; planned comparison: t(14) ¼ 2.08,
p ¼ 0.056; Figure 3D]. Post-ischemic recovery of end diastolic pressure was significantly elevated in psychosocially
stressed males [significant Group Time interaction:
F(1, 13) ¼ 8.26, p ¼ 0.013; Figure 3E], indicating that recovery of diastolic function was also worsened by psychosocial
stress. Collectively, these data indicate that chronic psychosocial stress occurring prior to the onset of ischemia increases
cell death and decreases post-ischemic recovery of contractile
function in male rats.
Females
Psychosocial stress had no effect on pre-ischemic contractile
419 function in females. In contrast to male rats, infarct size
420 [t(22) ¼ 1.51, p ¼ 0.15); Figure 4A] and post-ischemic
418
recovery of rate pressure product [Group Time interaction:
F(1, 21) ¼ 0.19, p ¼ 0.67; Figure 4B], +dP/dT [Group Time
interaction:
F(1, 21) ¼ 0.04,
p ¼ 0.84],
dP/dT
[Group Time interaction: F(1, 21) ¼ 0.13, p ¼ 0.73] and
end
diastolic
pressure
[Group Time
interaction:
F(1, 22) ¼ 0.07, p ¼ 0.79] were unaffected by psychosocial
stress in females.
Discussion
Despite extensive evidence of increased risk for CVD in
PTSD patients, few animal models of the disorder have
examined cardiovascular endpoints. In the present study, we
have shown that a psychosocial stress manipulation involving
predator exposure and daily social instability, one that has
previously been shown to emulate several core symptoms of
PTSD, increases myocardial sensitivity to ischemic injury in
male, but not female, rats. Following ischemia, hearts from
psychosocially stressed males exhibited significantly less
recovery of contractile function and demonstrated larger
infarct sizes than hearts from controls. Hearts from psychosocially stressed females, on the other hand, exhibited no
differences in infarct size or recovery of contractile function,
relative to controls. Moreover, psychosocially stressed
females did not show increased anxiety on the EPM, despite
exhibiting a reduced growth rate. Overall, our findings
provide evidence of a sex-dependent increase in myocardial
sensitivity to ischemic injury as a result of predator stress and
453
454
455
456
457
458
459
460
461
462
463
464
465
466
467
468
469
470
471
472
473
474
475
476
477
478
479
480
DOI: 10.3109/10253890.2015.1087505
Cardiac injury in animal model of PTSD
5
481
541
482
542
483
543
484
485
544
545
486
546
487
547
488
548
489
549
490
550
491
551
492
493
552
553
494
554
495
555
496
556
497
557
498
558
499
559
500
501
560
561
502
562
503
563
504
564
505
565
506
566
507
567
508
509
568
569
510
570
511
571
512
572
513
573
514
574
515
575
516
517
576
577
518
578
519
579
520
580
521
581
522
Figure 2. Physiological and behavioral effects of psychosocial stress in female rats. Psychosocially stressed females spent a similar amount of time in 582
the open arms of the elevated plus maze as control rats (A) and made a similar number of overall arm entries in the maze (B), despite exhibiting 583
524 significantly reduced growth rates (C) relative to controls. There were no significant differences between the adrenal (D) or thymus (E) gland weights 584
of stressed and non-stressed female rats. Sample sizes were 10–14 rats per group. Data are presented as means ± SEM. *p50.05 versus no stress.
523
525
585
526
586
chronic social instability, suggesting that this model may be
528 used to explore mechanisms underlying cardiovascular
529 changes that result from intense stress, at least in males.
527
530
531
532
533
534
535
536
537
538
539
540
Cardiovascular effects
PTSD promotes hypertension, atherosclerosis and the production of inflammatory markers that are detrimental to
cardiovascular function (Boscarino, 2011; Buckley et al.,
2013; Coughlin, 2011; Edmondson & Cohen, 2013;
Edmondson et al., 2013; von Kanel et al., 2007, 2010). The
underlying cause of CVD in PTSD patients is thought to be an
acceleration of atherosclerosis and subsequent blockage of
coronary arteries (Ahmadi et al., 2011). It is important to
note, however, that myocardial ischemia in the present study
was not induced by the atherosclerotic occlusion of coronary
arteries but was artificially induced by termination of
coronary flow. The observation that stressed males exhibited
greater ischemic injury than male controls in the absence of
atherosclerotic occlusion of the coronary vasculature suggests
that PTSD-induced atherosclerotic blockade of coronary
arteries may not be the only factor that contributes to the
increased risk of myocardial infarction in patients with PTSD.
The chronic psychosocial stress manipulation utilized in
the present study provides several important advantages over
studying the impact of PTSD on myocardial infarction in a
clinical setting. One advantage is that the psychosocial stress
manipulation avoids factors that are commonly comorbid with
587
588
589
590
591
592
593
594
595
596
597
598
599
600
6
B. R. Rorabaugh et al.
Stress, Early Online: 1–9
601
661
602
662
603
663
604
605
664
665
606
666
607
667
608
668
609
669
610
670
611
671
612
613
672
673
614
674
615
675
616
676
617
677
618
678
619
679
620
621
680
681
622
682
623
683
624
684
625
685
626
686
627
687
628
629
688
689
630
690
631
691
632
692
633
693
634
694
635
695
636
637
696
697
638
698
639
699
640
700
641
701
642
643
644
645
646
647
Figure 3. Myocardial infarct sizes, pre-ischemic contractile function and post-ischemic recovery of contractile function in male hearts exposed to
myocardial ischemia. Hearts from psychosocially stressed males exhibited significantly larger infarcts than control hearts (A; insets depict
representative myocardial slices with white areas indicative of infarction). There were no differences in pre-ischemic parameters of contractile function
between hearts from psychosocially stressed and control males. Recovery of rate pressure product (B) and +dP/dT (C) were significantly attenuated in
hearts from psychosocially stressed males, relative to those from controls. Recovery of dP/dT (D) was borderline significant in hearts from
psychosocially stressed males. Recovery of end diastolic pressure (E) was significantly elevated in psychosocially stressed males relative to controls.
Samples sizes were 7–9 hearts per group. Data are presented as means ± SEM. *p50.05 relative to no stress.
648
651
652
653
654
655
656
657
658
659
660
703
704
705
706
707
708
649
650
702
709
PTSD (depression, smoking, drug and alcohol abuse, etc.) and
can independently alter cardiovascular function. It also
provides a mechanism to compare the extent of myocardial
injury in stressed and non-stressed hearts under conditions
where the ischemic insult is identical between subjects.
Clinical data (death rates, hospitalization rates, rates of
coronary revascularization, etc.) have been important for
establishing that PTSD increases the likelihood of experiencing a myocardial infarction. However, clinical studies have
been unable to establish whether or not PTSD increases the
extent of myocardial injury. In addition to the common
comorbid factors mentioned above, ischemic conditions in
patients who are experiencing heart attacks are heterogeneous. For example, occlusion of a large coronary artery that
delivers blood to a wide region of myocardial tissue induces
ischemia over a much larger region of the heart than occlusion
of a smaller vessel that delivers blood to a more localized
area. In contrast, the psychosocial stress paradigm used here
enables stressed and non-stressed hearts to be made uniformly
ischemic so that the anatomical location and amount of
myocardial tissue that is subjected to ischemia is consistent
between individual hearts. One fundamental difference
710
711
712
713
714
715
716
717
718
719
720
DOI: 10.3109/10253890.2015.1087505
Cardiac injury in animal model of PTSD
7
721
781
722
782
723
783
724
725
784
785
726
786
727
787
728
788
729
789
730
790
731
791
732
733
792
793
734
794
735
795
736
796
737
797
738
798
739
799
740
741
800
801
742
802
743
803
744
804
745
805
746
806
747
807
748
749
808
809
750
810
751
811
752
812
753
813
754
814
755
815
756
757
816
817
758
818
Figure 4. Myocardial infarct sizes, pre-ischemic contractile function and post-ischemic recovery of contractile function in female hearts exposed to
myocardial ischemia. Psychosocial stress had no effect on infarct sizes (A; insets depict representative myocardial slices with white areas indicative of 819
760 infarction) or pre-ischemic contractile function. In addition, post-ischemic recovery of rate pressure product (B), +dp/dT (C), dP/dT (D) and end 820
761 diastolic pressure (E) were unaffected by psychosocial stress. Sample sizes were 10–14 hearts per group. Data are presented as means ± SEM.
821
759
762
763
764
765
766
767
768
769
770
771
772
773
774
775
776
777
778
779
780
822
between the psychosocial stress paradigm used here and a
clinical heart attack is that atherosclerotic blockage of
coronary arteries develops over a long period of time and
may be accompanied by the formation of collateral vessels or
other myocardial compensatory changes as ischemic heart
disease progresses. In contrast, the fact that the present
psychosocial stress manipulation induces myocardial ischemia through a mechanism that is independent of atherosclerotic coronary occlusion represents an important distinction
(and perhaps a limitation) from the clinical scenario.
Our finding that chronic psychosocial stress increases
myocardial sensitivity to ischemic injury is consistent with
previous work exploring the impact of chronic social,
psychological or emotional stress on myocardial sensitivity
to ischemic injury. For instance, Mercanoglu et al. (2008)
reported that consecutive exposure to a series of multiple
stressors (e.g. cage crowding, isolation, changes in light/dark
cycle, changes in cage mates, cage tilting, food/water
restriction) increased infarct size in the rat heart. Chronic
restraint stress produces a similar effect (Scheuer & Mifflin,
1998). In contrast, acute stress immediately prior to the onset
of ischemia protects the heart from ischemic injury
(Moghimian et al., 2012). Thus, the duration and timing of
stress appear to play a critical role in how ischemia impacts
the heart.
The present psychosocial stress paradigm induces multiple
physiological changes that may underlie the increased sensitivity of the male heart to ischemic injury. Wilson and
colleagues reported that male rats exposed to this psychosocial stress exhibit increased expression of inflammatory
cytokines and other protein markers of inflammation in the
brain (Wilson et al., 2013, 2014b,c). These rats also have
elevated concentrations of reactive oxygen species in blood,
brain and adrenal glands (Wilson et al., 2013). In addition,
male rats exposed to the psychosocial stress exhibit increased
norepinephrine release in the hippocampus and prefrontal
823
824
825
826
827
828
829
830
831
832
833
834
835
836
837
838
839
840
8
841
842
843
844
845
846
847
848
849
850
851
852
853
854
855
856
857
858
859
860
861
862
863
864
865
866
867
868
869
870
871
872
873
874
875
876
877
878
879
880
881
882
883
884
885
886
887
888
889
890
891
892
893
894
895
896
897
898
899
900
B. R. Rorabaugh et al.
cortex (Wilson et al., 2014a). Elevated blood pressure and
heart rate in psychosocial stressed rats are suggestive of
increased SNS stimulation (Zoladz et al., 2008, 2013).
Inflammation and oxidative stress play prominent roles in
modulating ischemia/reperfusion injury (Frangogiannis,
2014; Ling et al., 2013; Sandanger et al., 2013). Chronic
b-adrenergic receptor stimulation also worsens ischemic
injury (Hu et al., 2006). Thus, pro-inflammatory conditions,
increased oxidative stress and chronic b-adrenergic receptor
stimulation that pre-exist prior to the onset of ischemia may
all contribute to the psychosocial stress-induced increase in
myocardial sensitivity to ischemic injury.
Sex differences
Investigators have reported that traumatized females are twice
as likely as traumatized males to develop PTSD (Tolin & Foa,
2006). However, in a recent review paper, Zoladz & Diamond
(2013) discussed issues with this position, noting that for
several types of trauma, the incidence of PTSD is statistically
equivalent across the sexes. According to Zoladz & Diamond
(2013), most research reporting a greater prevalence of PTSD
in females, relative to males, has involved individuals whose
trauma included some element of assault. Indeed, for nonassault trauma, there appears to be no difference in the
prevalence of PTSD across men and women (e.g. Kessler
et al., 1995). Moreover, the authors raise the issue of female
hormone influences on emotional memory and, thus, traumatic memory formation. Basic and applied research studies
have shown that the levels of progesterone and estradiol
significantly influence emotional memories and their extinction (Bryant et al., 2011; Felmingham et al., 2012a,b). Thus,
the point at which the trauma occurs during the menstrual
cycle can significantly influence a single female’s likelihood
of PTSD development. To suggest that being female, per se,
presents an inherent vulnerability to PTSD may be an
oversimplification of a much more complicated process. It
is likely that the female sex interacts with genetics, trauma
type, personality characteristics and other environmental
factors to affect PTSD susceptibility.
Most basic science research on stress has been conducted
in males, and an established animal model of PTSD including
females is non-existent. The present study included an initial
attempt to extend the psychosocial, predator-based model to
females. However, we observed no anxiogenic or cardiovascular effects of psychosocial stress in female rats, despite
stressed females exhibiting reduced growth rate compared to
non-stressed females. One possibility for the lack of
anxiogenic or cardiovascular effects in stressed females is
that the psychosocial stress manipulation is less ‘‘stressful’’ to
females. Additionally, as suggested in the previous paragraph,
extensive work has shown that female hormone levels
significantly influence the stress response and emotional
memory formation. Thus, it is possible that the random times
of predator stress and physiological/behavioral testing in the
present study masked any estrous-dependency of the observed
effects. On the other hand, a recent meta-analysis showed that,
across thousands of behavioral, morphological, physiological
and molecular traits, there was no greater variability in the
data obtained from female rats that were examined without
Stress, Early Online: 1–9
estrous stage monitoring than was observed in data obtained
from male rats (Prendergast et al., 2014). In fact, the authors
of this meta-analysis observed significantly more variability
in the data from male rats for certain dependent variables.
Thus, it is also possible that, in the present study, estrous stage
did not introduce anymore variability in the behavioral or
myocardial measures than was observed in males. We did not
assess the estrous cycle in females because we did not want
the stress associated with estrous testing to confound our
results. To conclusively determine whether females are less
stressed by this psychosocial stress manipulation or if estrous
stage influences female responses to the psychosocial stress,
future research is necessary.
901
902
903
904
905
906
907
908
909
910
911
912
913
914
Conclusions
The psychosocial predator-based model used here provides a
useful tool to study the cardiac impact of intense stress. We
anticipate that future work will further our understanding of
the mechanisms by which PTSD-like sequelae in rats increase
myocardial ischemic injury. This could provide new therapeutic strategies to protect the hearts of PTSD patients from
myocardial infarction and other ischemic heart diseases.
915
916
917
918
919
920
921
922
923
924
Declaration of interest
925
The authors report no conflicts of interest.
926
927
References
928
929
Ahmadi N, Hajsadeghi F, Mirshkarlo HB, et al. (2011). Post-traumatic
stress disorder, coronary atherosclerosis, and mortality. Am J Cardiol
108(1):29–33.
Bigger Jr JT, Fleiss JL, Steinman RC, et al. (1992). Frequency domain
measures of heart period variability and mortality after myocardial
infarction. Circulation 85(1):164–71.
Boscarino JA. (2011). Post-traumatic stress disorder and cardiovascular
disease link: time to identify specific pathways and interventions. Am
J Cardiol 108(7):1052–3.
Bryant RA, Felmingham KL, Silove D, et al. (2011). The association
between menstrual cycle and traumatic memories. J Affect Disord
131(1–3):398–401.
Buckley T, Tofler G, Prigerson HG. (2013). Posttraumatic stress disorder
as a risk factor for cardiovascular disease: a literature review and
proposed mechanisms. Curr Cardiovasc Risk Rep 7:506–13.
Cohen H, Benjamin J, Geva AB, et al. (2000). Autonomic dysregulation
in panic disorder and in post-traumatic stress disorder: application of
power spectrum analysis of heart rate variability at rest and in
response to recollection of trauma or panic attacks. Psychiatry Res
96(1):1–13.
Cohen H, Kotler M, Matar MA, et al. (1998). Analysis of heart rate
variability in posttraumatic stress disorder patients in response to a
trauma-related reminder. Biol Psychiatry 44(10):1054–9.
Coughlin SS. (2011). Post-traumatic stress disorder and cardiovascular
disease. Open Cardiovasc Med J 5:164–70.
Daskalakis NP, Lehrner A, Yehuda R. (2013). Endocrine aspects of posttraumatic stress disorder and implications for diagnosis and treatment.
Endocrinol Metab Clin North Am 42(3):503–13.
Edmondson D, Cohen BE. (2013). Posttraumatic stress disorder and
cardiovascular disease. Prog Cardiovasc Dis 55(6):548–56.
Edmondson D, Kronish IM, Shaffer JA, et al. (2013). Posttraumatic
stress disorder and risk for coronary heart disease: a meta-analytic
review. Am Heart J 166(5):806–14.
Felmingham KL, Fong WC, Bryant RA. (2012a). The impact of
progesterone on memory consolidation of threatening images in
women. Psychoneuroendocrinology 37:1896–900.
Felmingham KL, Tran TP, Fong WC, et al. (2012b). Sex differences in
emotional memory consolidation: the effect of stress-induced salivary
alpha-amylase and cortisol. Biol Psychol 89:539–44.
930
931
932
933
934
935
936
937
938
939
940
941
942
943
944
945
946
947
948
949
950
951
952
953
954
955
956
957
958
959
960
Q4
DOI: 10.3109/10253890.2015.1087505
961
962
963
964
965
966
967
968
969
970
971
972
973
974
975
976
977
978
979
980
981
982
983
984
985
986
987
988
989
990
991
992
993
994
995
996
997
998
999
1000
1001
1002
1003
Frangogiannis NG. (2014). The inflammatory response in myocardial
injury, repair, and remodelling. Nat Rev Cardiol 11(5):255–65.
Granville DJ, Tashakkor B, Takeuchi C, et al. (2004). Reduction of
ischemia and reperfusion-induced myocardial damage by cytochrome
P450 inhibitors. Proc Natl Acad Sci USA 101(5):1321–6.
Hu A, Jiao X, Gao E, et al. (2006). Chronic beta-adrenergic receptor
stimulation induces cardiac apoptosis and aggravates myocardial
ischemia/reperfusion injury by provoking inducible nitric-oxide
synthase-mediated nitrative stress. J Pharmacol Exp Ther 318(2):
469–75.
Kessler RC, Sonnega A, Bromet E, et al. (1995). Posttraumatic stress
disorder in the National Comorbidity Survey. Arch Gen Psychiatry
52(12):1048–60.
Libby P, Ridker PM, Hansson GK. (2009). Inflammation in atherosclerosis: from pathophysiology to practice. J Am Coll Cardiol 54(23):
2129–38.
Ling H, Gray CB, Zambon AC, et al. (2013). Ca2+/Calmodulindependent protein kinase II delta mediates myocardial ischemia/
reperfusion injury through nuclear factor-kappaB. Circ Res 112(6):
935–44.
Mercanoglu G, Safran N, Uzun H, et al. (2008). Chronic emotional stress
exposure increases infarct size in rats: the role of oxidative and
nitrosative damage in response to sympathetic hyperactivity. Methods
Find Exp Clin Pharmacol 30(10):745–52.
Moghimian M, Faghihi M, Karimian SM, et al. (2012). The effect of
acute stress exposure on ischemia and reperfusion injury in rat heart:
role of oxytocin. Stress 15(4):385–92.
O’Donovan A, Cohen BE, Seal KH, et al. (2015). Elevated risk for
autoimmune disorders in Iraq and Afghanistan veterans with
posttraumatic stress disorder. Biol Psychiatry 77(4):365–74.
Piccirillo G, Magri D, Matera S, et al. (2007). QT variability strongly
predicts sudden cardiac death in asymptomatic subjects with mild or
moderate left ventricular systolic dysfunction: a prospective study. Eur
Heart J 28(11):1344–50.
Prendergast BJ, Onishi KG, Zucker I. (2014). Female mice liberated for
inclusion in neuroscience and biomedical research. Neurosci
Biobehav Rev 40:1–5.
Roth TL, Zoladz PR, Sweatt JD, et al. (2011). Epigenetic modification of
hippocampal Bdnf DNA in adult rats in an animal model of posttraumatic stress disorder. J Psychiatr Res 45:919–26.
Rozanski A, Blumenthal JA, Davidson KW, et al. (2005). The
epidemiology, pathophysiology, and management of psychosocial
risk factors in cardiac practice: the emerging field of behavioral
cardiology. J Am Coll Cardiol 45(5):637–51.
Sandanger O, Ranheim T, Vinge LE, et al. (2013). The NLRP3
inflammasome is up-regulated in cardiac fibroblasts and mediates
myocardial ischaemia-reperfusion injury. Cardiovasc Res 99(1):
164–74.
Scheuer DA, Mifflin SW. (1998). Repeated intermittent stress
exacerbates myocardial ischemia–reperfusion injury. Am J Physiol
274(2 Pt 2):R470–5.
Tolin DF, Foa EB. (2006). Sex differences in trauma and posttraumatic
stress disorder: a quantitative review of 25 years of research. Psychol
Bull 132(6):959–92.
Cardiac injury in animal model of PTSD
9
Vaccarino V, Goldberg J, Rooks C, et al. (2013). Post-traumatic stress
disorder and incidence of coronary heart disease: a twin study. J Am
Coll Cardiol 62(11):970–8.
von Kanel R, Begre S, Abbas CC, et al. (2010). Inflammatory
biomarkers in patients with posttraumatic stress disorder caused by
myocardial infarction and the role of depressive symptoms.
Neuroimmunomodulation 17(1):39–46.
von Kanel R, Hepp U, Kraemer B, et al. (2007). Evidence for low-grade
systemic proinflammatory activity in patients with posttraumatic
stress disorder. J Psychiatr Res 41(9):744–52.
Waterson RE, Thompson CG, Mabe NW, et al. (2011). Galpha(i2)mediated protection from ischaemic injury is modulated by
endogenous RGS proteins in the mouse heart. Cardiovasc Res
91(1):45–52.
Wilson CB, Ebenezer PJ, McLaughlin LD, et al. (2014a). Predator
exposure/psychosocial stress animal model of post-traumatic stress
disorder modulates neurotransmitters in the rat hippocampus and
prefrontal cortex. PLoS One 9(2):e89104.
Wilson CB, McLaughlin LD, Ebenezer PJ, et al. (2014b). Differential
effects of sertraline in a predator exposure animal model of posttraumatic stress disorder. Front Behav Neurosci 8:256.
Wilson CB, McLaughlin LD, Ebenezer PJ, et al. (2014c). Valproic acid
effects in the hippocampus and prefrontal cortex in an animal model
of post-traumatic stress disorder. Behav Brain Res 268:72–80.
Wilson CB, McLaughlin LD, Nair A, et al. (2013). Inflammation and
oxidative stress are elevated in the brain, blood, and adrenal glands
during the progression of post-traumatic stress disorder in a predator
exposure animal model. PLoS One 8(10):e76146.
Yehuda R. (2009). Stress hormones and PTSD. In: Shiromani PJ, Keane
TM, LeDoux JE, editors. Post-traumatic stress disorder: basic science
and clinical practice. New York, NY: Humana Press. p. 257–75.
Yehuda R, Seckl J. (2011). Minireview: stress-related psychiatric
disorders with low cortisol levels: a metabolic hypothesis.
Endocrinology 152(12):4496–503.
Zoladz PR, Conrad CD, Fleshner M, et al. (2008). Acute episodes
of predator exposure in conjunction with chronic social instability as
an animal model of post-traumatic stress disorder. Stress 11(4):
259–81.
Zoladz PR, Diamond DM. (2013). Current status on behavioral and
biological markers of PTSD: a search for clarity in a conflicting
literature. Neurosci Biobehav Rev 37(5):860–95.
Zoladz PR, Fleshner M, Diamond DM. (2013). Differential effectiveness
of tianeptine, clonidine and amitriptyline in blocking traumatic
memory expression, anxiety and hypertension in an animal model
of PTSD. Prog Neuropsychopharmacol Biol Psychiatry 44:1–16.
Zoladz PR, Fleshner M, Diamond DM. (2012). Psychosocial animal
model of PTSD produces a long-lasting traumatic memory, an
increase in general anxiety and PTSD-like glucocorticoid abnormalities. Psychoneuroendocrinology 37(9):1531–45.
Zoladz PR, Park CR, Fleshner M, et al. (2015). Psychosocial predatorbased animal model of PTSD produces physiological and behavioral
sequelae and a traumatic memory four months following stress onset.
Physiol Behav 147:183–92.
1021
1022
1023
1024
1025
1026
1027
1028
1029
1030
1031
1032
1033
1034
1035
1036
1037
1038
1039
1040
1041
1042
1043
1044
1045
1046
1047
1048
1049
1050
1051
1052
1053
1054
1055
1056
1057
1058
1059
1060
1061
1062
1063
1004
1005
1064
1065
1006
1066
1007
1067
1008
1068
1009
1069
1010
1070
1011
1071
1012
1013
1072
1073
1014
1074
1015
1075
1016
1076
1017
1077
1018
1078
1019
1079
1020
1080
Q5
Download