Content uploaded by Jarrod Bailey
Author content
All content in this area was uploaded by Jarrod Bailey on Dec 05, 2018
Content may be subject to copyright.
Introduction
In the December 2017 issue of ATLA, I published an
Editorial, in which I suggested that stress in ani-
mals in laboratories, resulting from their environ-
ment and experimental procedures, was not limited
to welfare concerns (1). It also adversely affects, sig-
nificantly and unavoidably, multiple biological sys-
tems and therefore affects the resultant
experimental data. This exacerbates inter-species
differences and makes extrapolation to humans even
more difficult and unreliable. In an effort to encour-
age and promote discussion of this issue, which is
underappreciated, I am expanding on that Editorial.
Stress may be thought of as “the sum of the biolog-
ical reactions to any adverse stimulus, physical,
mental, or emotional, internal or external, that
tends to disturb the homeostasis of an organism” (2).
It is increasingly acknowledged that diverse species
experience pain, stress and distress (see below), and
experience depression and anxiety disorders (see
Ferdowsian et al. [3]). This includes fish — notably
the ubiquitous laboratory zebrafish — which show
emotional fever/stress-induced hyperthermia in
response to a variety of stressors, including simple
handling (4, 5). Stress in fish can result in increased
aggressive behaviour (6), elevated anxiety, dimin-
ished weight-gain, and altered levels of dopamine
and serotonin metabolites in the brain (7), as well as
increased brain levels of extracellular adenosine,
which has neuromodulatory effects (8).
While all species experience stressors and stress
in their natural environments, psychological and
physiological problems can arise with exposure to
recurrent stressors, and/or when stress becomes
chronic and too difficult to cope with. This leads to
allostatic overload (excessive wear and tear on the
body), which may manifest in altered physiological
responses, some of which can be harmful (9) — gen-
erally thought of as a state of distress. How ever, the
US National Research Council (NRC) accepts that
there is “confusion in the scientific, regulatory, and
animal welfare communities” concerning the distinc-
tion between stress and distress; and that these
terms are often used interchangeably in animal wel-
Does the Stress of Laboratory Life and Experimentation on
Animals Adversely Affect Research Data? A Critical Review
Jarrod Bailey
Cruelty Free International, London, UK
Summary — Recurrent acute and/or chronic stress can affect all vertebrate species, and can have serious
consequences. It is increasingly and widely appreciated that laboratory animals experience significant and
repeated stress, which is unavoidable and is caused by many aspects of laboratory life, such as captivity,
transport, noise, handling, restraint and other procedures, as well as the experimental procedures
applied to them. Such stress is difficult to mitigate, and lack of significant desensitisation/habituation can
result in considerable psychological and physiological welfare problems, which are mediated by the
activation of various neuroendocrine networks that have numerous and pervasive effects. Psychological
damage can be reflected in stereotypical behaviours, including repetitive pacing and circling, and even
self-harm. Physical consequences include adverse effects on immune function, inflammatory responses,
metabolism, and disease susceptibility and progression. Further, some of these effects are epigenetic,
and are therefore potentially transgenerational: the biology of animals whose parents/grandparents were
wild-caught and/or have experienced chronic stress in laboratories could be altered, as compared to free-
living individuals. It is argued that these effects must have consequences for the reliability of
experimental data and their extrapolation to humans, and this may not be recognised sufficiently among
those who use animals in experiments.
Key words: animal welfare, cost–benefit analysis, data accuracy, glucocorticoids, psychological, stress,
translational medical research.
Address for correspondence: Jarrod Bailey, Cruelty Free International, 16a Crane Grove, London,
N7 8NN, UK.
E-mail: jarrod.bailey@crueltyfreeinternational.org
ATLA 46, 291–305, 2018 291
Comment
fare literature and that relevant available informa-
tion is “far from complete”, with distress remaining
“a complex and still poorly understood phenomenon”
(10). Nevertheless, it is generally understood that
distress manifests when an individual is unable to
cope and adapt successfully to one or more stressors,
resulting in compromised well-being due to the
inability to return to physiological and psychological
homeostasis (11). Notably, an individual can remain
in distress, even when a stressor is removed.
While different species and individuals have dif-
ferent stressors, variable ranges of stress to which
they can adapt, diverse spectra of tolerance, and
dissimilar manifestations and sequelae of exces-
sive stress, they share the same biological path-
ways and mechanisms that are adversely affected
by stress. Briefly, stressors stimulate the hypotha-
lamic–pituitary–adrenal (HPA) axis, the sympa-
thetic adrenal medullary axis, and the sympathetic
and parasympathetic nerve projections that
directly innervate secondary lymphoid organs (12–
14). Among other things, this results in the eleva-
tion of the ‘stress hormones’, cortisol and
corticosterone (CORT). Raised glucocorticoid (GC)
levels, alongside significant elevations of heart
rate, blood pressure and other hormone levels, are
acknowledged indicators of fear, stress and dis-
tress, and are used as biomarkers for stress in ter-
restrial vertebrates in laboratories (e.g. 15–17).
Direct consequences of these neuroendocrine
changes include deleterious effects on innate and
adaptive immunity, central nervous system
pathology, and cardiovascular and reproductive
perturbations (18, 19), leading to extensive and
diverse adverse health effects. Psychological
trauma, for instance, may result in altered health
or damaging patterns of behaviour. In humans,
immune perturbations manifest in poor responses
to vaccines, increased susceptibility to infections,
and accelerated disease progression (see Gurfein et
al. [18]). Further, the various mechanisms under-
lying such effects mean that stress has ramifica-
tions beyond the individuals experiencing it.
Successive generations, and individuals who have
experienced prenatal and/or early-life stress, may
be destined to suffer the consequences in adult-
hood.
Caveats and Framing the Argument
Chronic or long-term stress is not unique to ani-
mals in laboratories — it is part and parcel of life
for all species in whatever environment. I do not
intend to suggest otherwise, and it is accepted that
animals in the wild experience acute stressors reg-
ularly (such as lack of food and shelter, predation,
disease, and so on); not only is this entirely normal,
but it can be beneficial in terms of building allo-
static capacity. This Comment, however, argues
that stresses resulting from life in the laboratory
often have negative consequences — given the
opportunity, the animals would avoid many labo-
ratory stressors, and so they can hardly be consid-
ered benign. In addition, the degree, type,
frequency and duration of laboratory stressors are
different to those in the wild, and together could be
more chronic. This may result in greater adverse
consequences for animal welfare and for scientific
data quality. Experiencing repeated, unnatural
stressors, such as blood draws and gavage, cannot
be compared to the brief elevations in CORT
resulting from the natural diurnal/nocturnal
adrenal cycle. For instance, the former stressors
are very different from simply waking up, or being
hungry. In well-designed experiments, increases in
stress biomarkers are relative to baseline levels in
any case (see, for example, 16, 20).
That laboratory stress can — arguably, frequently
and unavoidably — have effects that compromise
animal welfare and experimental results is accepted
by the US NRC’s Committee on Recognition and
Alleviation of Distress in Laboratory Animals (10):
“In the longer term… a breakdown in an animal’s
ability to cope with its environment is likely to lead
to adverse emotional states and poor welfare. Some
of these cases may be quite minor and not give rise
to significant ethical concerns; but prolonged or
intense circumstances would compromise the ani-
mal’s welfare enough to warrant concern and also
significantly affect the research results.” Further:
“Strong evidence in rodents has shown that mild
stress of 2–3 months duration — a regimen that pro-
duces no signs of overt distress — alters the animals’
performance in tests of anxiety, depression, and
memory… Other findings indicate that rats’ habitu-
ation to a test environment can dramatically affect
their response to a toxic substance”. Stress can also
be beneficial: “Over a longer time frame, glucocorti-
coid production in response to infection helps restrict
the immune system, thus preventing deleterious
effects of inflammatory factors on tissues” (10).
While this latter statement is true, it also supports
the argument made here, that stress — even when
not ‘bad’ — adversely affects biological systems, and
consequently confounds research data.
Therefore, it is not only distress that leads to bio-
logical consequences affecting data reliability,
quality and relevance. It seems clear that the con-
sequences of stress are also an issue for welfare
and data quality. To quote again the US NRC:
“The impact of distress on both animal welfare and
research results is likely even more pronounced
than that of stress. Animals exposed to prolonged
severe stress experience underlying changes in
physiological functions (e.g. gastric lesions or
immunosuppression) that can interfere with exper-
imental manipulations; alter experimental vari-
ables such as behaviour, drug dosing and
clearance; change the progress of a disease; and
292 Comment
contribute to morbidity and mortality. A variety of
stressors can contribute to unintended distress,
from postoperative pain or infection to barren
housing conditions or the solitary confinement of
an individual of a social species. Stereotypies,
abnormal repetitive behaviours indicative of poor
well-being that are often observed in distressed
animals, are thought to reflect defective brain
function and to be a result of poor animal welfare.
Stereotypies are thus likely to interfere with
behavioural, neuroscience, and pharmacological
studies” (10); and “The impact of stress and dis-
tress on the quality of scientific research can result
in the generation of compromised data, which in
turn necessitates the use of more animals… Both
stress and distress represent potential complica-
tions in a wide range of experiments, and should be
proactively addressed by good experimental
design” (10). While I (and others) disagree that the
use of more animals and the careful design of
experiments can have an acceptably positive
impact on welfare, and on the human-relevance of
the data generated, the main points in this
Comment stand. Efforts to address this issue over
many years are acknowledged, but I argue that,
due to fundamental biology and inter-species dif-
ferences, the improvement of husbandry, veteri-
nary care, regulation, oversight, training, etc. can
never be sufficient to significantly address and
overcome these issues.
Stress Resulting from Laboratory Life
and Research, and its Biological
Impact
The issue for animals in laboratories is that life
can be inherently and excessively stressful — per-
haps much more than in their natural environ-
ments, from which laboratory conditions differ
greatly, even when enriched and accounting for
efforts to mitigate stressors. Laboratory conditions
preclude or limit many natural behaviours, hous-
ing tends to be much smaller than the animals’
natural ranges, and the animals are subjected to
frequent manipulations and handling, as well as
other alien factors that they try to resist and avoid
(16, 20). Stressful procedures and environmental
factors are numerous and varied. Briefly, they
include, but are not limited to: general handling
and manipulations, such as weighing and saline
injections (15, 16, 21–23); anaesthesia (15, 24–31);
restraint (21, 32–36); gavage (37–42); blood sam-
pling (15–17, 43–50); food and water restriction
(51, 52); non-natural environment (53–55) and
associated factors, such as noise and light (56–62);
social crowding and/or isolation (63–71); cage con-
ditions/cleaning/changing (18, 58, 72–78); trans-
port (19, 58, 79–83); observing procedures on, and
killing of, other animals (31, 84–87); and even
enrichment itself (17, 88, 89). It has been sug-
gested that captive-bred animals may not know
that these stressors are different to those in the
wild, though there might be some perception that
they are not similar to ‘natural’ stressors such as
limited food, inadequate shelter, predation and so
on. In any case, the point is that they are different,
but more importantly, they are also frequent, reg-
ular and inescapable.
Naturally, this has animal welfare and scientific
implications, acknowledged at least in some quar-
ters. For instance, stress from handling is accepted
as a source of “unexplained variation within and
between animal studies”, as it influences “both the
behaviour and physiology of animals” (23; see also
Balcombe et al. [16] and Meijer et al. [17]), and rel-
atively poor caging conditions “may contribute to
problems in translating murine research into
human studies” (18, 77, 78). However, it is acc -
epted by some that these factors are probably
widely underappreciated (90). It should be noted
that some consider enriched environments simply
as ‘less bad’ rather than ‘better’ than those that are
non-enriched. To illustrate, significant numbers of
animals experiencing enrichment still go on to
develop stereotypies (e.g. 91–98).
The Nature of Stress: Biological
Basis/Mechanisms of Stress and its
Adverse Effects
The physiological consequences of stress are varied
and powerful. This, in itself, is of concern for the
translation of animal data to humans, as they com-
pound and confound existing difficulties in transla-
tion due to species differences. First, however, a
consideration of the underlying mechanisms is
important, to demonstrate their fundamental nature
and potency.
Primary mediators of stress, such as GCs and cat-
echolamines, are released in response to stressors,
with various biological consequences. Such biological
effects generally go beyond species boundaries/limits
for mammalian species, though the mechanisms and
specific effects differ to varying degrees. These pri-
mary mediators are extremely powerful, because
they ultimately modulate the expression of many
genes. Secondary outcomes have been documented
“…in every physiological system, including the car-
diovascular system, metabolism, the central nervous
system, and the immune system”, and are con-
founded by the characteristics of the stressor(s), as
well as the attributes of the affected individual, such
as age, health, status, genetic background, past
experience, etc. (99).
The potency and ubiquity of the stress response
has been demonstrated in a number of in vitro stud-
ies, revealing that it generally blocks every impor-
tant cellular process, including DNA replication,
Comment 293
transcription, pre-mRNA processing, mRNA export,
and translation, until the cells recover (100).
Therefore, stress exerts its effects via varied molec-
ular mechanisms, with far-reaching consequences.
The principal ones are highlighted below.
— Epigenetic mechanisms (histone acetylation and
DNA methylation) (101–107): Psychological
stress alters gene expression via histone acety-
lation and DNA methylation. Much occurs in
response to environmental triggers, e.g. diet,
drugs, toxins, and psychological stress, e.g. fear
conditioning and maternal care (103). Genes
involved in HPA axis function are especially
susceptible (108), e.g. in suicide victims with a
history of child abuse (109), and post-traumatic
stress disorder (PTSD) is strongly associated
with the epigenetic modification of genes
involved in immune function and inflammation
(110).
— Alternative splicing/expression of regulatory
microRNAs: Alternative splicing of gene tran-
scripts and microRNAs (miRNAs), is a powerful
means of altering gene expression that can be
significantly affected by stress (111, 112). For
example, acute stress in humans altered the
splicing of 27 genes in peripheral leukocytes
(100).
— Oxidative damage and ageing: Mental stress con-
tributes to oxidative stress in the body, and
therefore to oxidative damage (113). This effect
has been identified in students undergoing aca-
demic examinations (114), and in the lympho-
cytes of psychologically stressed individuals
(115). Oxidative stress is also associated with
PTSD and depression (116), contributes to the
ageing process (117), and is associated with neu-
rodegenerative disease, ophthalmologic disease,
cancer and cardiovascular disease (including
atherosclerosis, hypertension, cardiomyopathy,
chronic heart failure, myocardial ischaemia and
ventricular arrhythmias; 117, 118). There may be
confounding data on the effects of oxidative stress
from different species/strains of mice, which fur-
ther challenges the translation of data across
species (119).
Direct Physiological Consequences of
Stress
The physiological consequences of stress are
numerous and varied. Table 1 shows how promis-
cuous these consequences, effects and manifesta-
tions are in many species, including humans.
Given that these effects involve so many biological
pathways and systems, the effects on experimental
data must be significant.
Habituation/Desensitisation to Stress
Some argue that animals become habituated (120)
and/or desensitised (121) to stress, so implications
for welfare and experimental results may be over-
come (23, 122, 123). For instance, non-human
primates can be trained to approach test environ-
ments, and to present their arms for blood with-
drawal and so on, seemingly voluntarily. However,
it has been shown that repeat exposure to homo-
typic stressors of greater intensity and/or severity
does not result in habituation, and could actually
result in sensitisation of the CORT response (122).
Furthermore, CORT levels might only decrease for
certain types of stressor, persisting for other types
(35); where desensitisation has been shown, it is
only to a modest degree, in a small proportion of
the animals in the studies, and in small sample
sizes (70). While some studies have suggested that
enrichment might decrease stress, others have
shown a paradoxical increase in stress via CORT
levels (18). Similarly, some studies have suggested
that transferring scent-marked materials from old
to new cages reduces stress-related aggression,
while others found that it increases aggression
(74). Furthermore, mice do not habituate to stress
associated with simple handling, and indeed seem
to become sensitised to it (124–126). In instances
where CORT levels decrease, there is increasing
evidence that this does not necessarily mean an
attendant decrease in stress; other indicators, such
as the neutrophil–lymphocyte ratio, might be ‘bet-
ter’ indicators of chronic stress, which can occur in
the absence of increased serum CORT (127).
Adverse Physiological Sequelae of
Psychological Stress are Initiated
Prenatally or in Early Life, and are
Heritable
Captive-born animals can be exposed to stress pre-
natally via their wild-trapped mothers (128–131),
and as infants in laboratory environments, often
experiencing inadequate maternal contact and
care (132–134; cited by Camus et al. [135], and
Detter et al. [136]). If an animal’s parents or grand-
parents lived in laboratories, and/or were born of
parents that lived in laboratories and/or endured
being wild-caught, then their ancestors experi-
enced highly stressful lives and would have been
affected by the adverse consequences described
herein. Even if such an individual was subse-
quently afforded as stress-free a life as possible
(difficult, if not impossible, in a laboratory), the
consequences of their early lives, and the lives of
their ancestors, would lead to the same adverse
effects as if they had continued to experience
excessive stress as adults.
294 Comment
That early-life experiences affect adult psy-
chopathology is widely accepted. As Jean-Paul
Sartre put it, “Childhood decides” (see Murgatroyd
[102]). “A large body of data shows that stress dur-
ing pregnancy causes an increase of GCs in the
blood of the dams and the foetus, leading to alter-
ations of the structure and function of the develop-
ing brain… These alterations result in the
disturbance of the function of the neuroendocrine
system and different kinds of behaviour through-
out life” (137). Early-life/prenatal exposure to
stress leads to altered adrenocorticotrophic hor-
mone responsiveness, dysfunction of the HPA axis
(138, 139), and altered autonomic modulation of
immune function that may begin in utero (103).
Social isolation in several species leads to neuroen-
docrine changes, increased cortisol, and ensuing
behavioural problems (for references, see Cham -
pagne [101]). Maternal inflammation during preg-
nancy (from infection, or possibly stress) may
increase the risk of neurodevelopmental disorders
such as schizophrenia and cerebral palsy (140).
Physiological sequelae include cardiovascular dis-
ease and metabolic disorders such as diabetes
(141), compromised immune function, including
poor lymphocyte proliferation upon infection and
reduced placental transfer of antibodies during
pregnancy (140), autoimmune disorders, chronic
obstructive lung disease, asthma and obesity (see
Chang [142]). Pivotal to these adverse outcomes
are the aforementioned stress-related epigenetic
processes and oxidative damage. Methylation of
gene regulatory regions is partly involved (143),
the extent of which may be set during prenatal
development (141). Cord blood samples of infants
of mothers with late-pregnancy depression show
altered methylation of the GC receptor promoter,
which also predicts elevated salivary cortisol in
early life (144). Other modifications are inherited
and transgenerational in nature (145); for exam-
ple, poor prenatal nutrition affects GC receptor
methylation, affecting the growth and metabolism
of first and second-generation offspring (146), and
matrilineal transmission of the effects of diethyl-
stilbestrol (DES) occurs via hypomethylation, lead-
ing to increased cancer risk over two generations
(147).
Summary
The inherent, multi-faceted stress of laboratory life
— often excessive, relative to the more transient,
acute and ‘natural’ stresses experienced in the wild
— is evidenced by its often negative impact on the
well-being of the animals involved, both psycholog-
ically and physiologically. Because this harm is
mediated via established trans-species biological
mechanisms involving the HPA axis and the sym-
pathetic nervous system, and effected via oxidative
stress and epigenetic mechanisms, which affect
multiple biological pathways and systems, it can
have adverse and confounding effects on experi-
mental results. This modulation of many biochem-
ical pathways and gene expression can result in
downstream effects such as organ damage, cardio-
vascular diseases, attenuated immune function
and autoimmune disorders, premature ageing and
mortality, developmental abnormalities, elevated
tumour initiation and progression, and muscu-
loskeletal atrophy (16).
The absolute degree of translation of animal stud-
ies to humans is debatable, but undoubtedly,
“Studies using animal models are more translatable
to human disease when the animals’ welfare is
maximised” (127). Arguably, it is difficult to “max-
imise” welfare significantly, given the in herent,
widespread, substantial and largely intractable
nature of the stresses involved in animal research
and laboratory life. Notably, habituation and/or
desensitisation to many of the stressors is often not
possible, or at least not significant, and the impact
of these effects on experimental data — and their
extrapolation to humans — is likely to be signifi-
cant. The many sources of stress have been sum-
marised here, along with their effects on multiple
biological and physiological systems. The literature
warns that: “…animals subjected to the environ-
mental changes that occur during transportation…
react with changes in their physiology, such as body
weight, plasma hormonal levels, heart rate and
blood pressure changes… When measurements of
physiological parameters are performed using con-
ventional measurement techniques, the results
must be interpreted with caution as these conven-
tional techniques also have effects on the animals”
(148). Most importantly, “Suffering in animals can
result in physiological changes which may increase
the variability of experimental data” (149). Many
scientists are well aware of these effects and consid-
erations, and have cautioned against disregarding
them (150–152). Yet, while accepting the negative
effects of pain, stress and distress, and their influ-
ence on study outcome, such effects are often not
reported or are under-reported in scientific publica-
tions (90).
The impact of stress on immunological and
inflammatory responses seems particularly preva-
lent, and might be especially critical, seeing as
much animal experimentation involves infectious
agents and/or immune function (for a discussion
and references, see Bailey [153] and Bailey [154]).
Crucially, this impact exacerbates and compounds
existing immune differences between humans and
non-humans due to genetic differences. To illus-
trate, genomic duplications — one of the most sig-
nificant causes of genetic variation among
primates (155) and at the root of many aspects of
intra-species and inter-species diversity — differ-
entially affect many genes involved in immune and
Comment 295
Table 1: A summary of the physiological consequences and manifestations of stress, in many species, including humans
Consequences of general stress Notes
Stereotypies (abnormal, repetitive, invariant These correlate with basal plasma cortisol and corticotrophin-releasing hormone (CRH) in cerebrospinal fluid (166).
behaviours with no obvious function; 160, 161)
and self-harm (e.g. 162–165) in non-humans
Wide-ranging physiological symptoms in humans Anger, depression, anxiety, behavioural changes, food cravings, lack of appetite, frequent crying, difficulty sleeping, tiredness, lack of
concentration, chest pains, constipation, diarrhoea, cramps and muscle spasms, dizziness, fainting, nervous twitches, restlessness, sexual
dysfunctions, breathlessness, and a host of diseases and illnesses with probable associated psychogenic (as well as biological) causes (167).
Long-lasting neurophysiological changes These could “…have direct implications for electrophysiological, behavioural, and molecular studies” (see 69).
— Isolated rats show “…structural and functional changes in the mesocorticolimbic dopaminergic system, exhibited hyperlocomotor
activity and impaired sensorimotor gating” (168).
— Isolated pigs show “…sustained changes in behavioural, neuroendocrine and immune regulation” (169).
— Socially isolated humans show increased risk of death, “…genome-wide transcriptional activity of impaired GC response genes and
increased activity of pro-inflammatory transcription control pathways”, and higher risk of developing “…conduct disorders,
personality disorders, major depression, PTSD, schizophrenia, and anxiety disorders” (see 170).
Multi-faceted modulation of the immune system This occurs in many, if not all, mammalian species (140, 171, 172). It is especially problematic for research involving immune function,
infectious diseases, etc. The expression of several hundred genes (many related to immune function) may be perturbed by simple
handling of animals (173; and see 33), which may be affected by the acute or chronic nature of the stressor (see 173). Chronic stress can:
— shift neutrophil–lymphocyte ratios (127, 174);
— affect expression of cytokines and cytokine receptors (175, 176), such as IL-2 and IL-6 (18);
— alter gene splicing in peripheral leukocytes (10);
— alter global and immune gene specific methylation (177); and
— differentially affect brain activity and neurotransmitter release, macrophage activity and antibody production (33).
The same stressor in acute or chronic forms may have different effects:
— In rodents, acute restraint increases delayed-type hypersensitivity (DTH) and leukocyte redeployment, but can increase or decrease
them when chronic (33, 178).
— In humans, chronic, though not acute, stress increases susceptibility to colds (179).
These inconsistencies show that stress is mediated not only by GCs (180), and that accounting for the effects of stress in animal
research must be difficult, if not impossible.
Observed alterations in human immune function These include:
— “attenuated responses to vaccination, poorer wound healing, exaggerated release of inflammatory mediators, & premature aging of
the immune system” (181);
— increased plasma and CNS cytokine levels, impaired natural killer cell activity, lower T-lymphocyte counts (in PTSD/complex
PTSD patients; 182);
— epigenetic changes exerting lifelong impact on immune and inflammatory function (in PTSD/complex PTSD patients; 182); and
— greater inflammatory responses to vaccinations (in depressed humans; 183).
Neuropeptides involved in stress responses may accentuate pathophysiological sequelae in critically ill individuals (184). In healthy
humans, 49 different genetic pathways are affected by stress, including genes associated with immune function (185). Stressed students
undergoing examinations have significantly increased pro-inflammatory cytokines (186).
Activation of the HPA axis This accelerates ageing generally, with adverse effects on brain/central nervous system, immune system, skeletal muscle and bone tissue
(see 187–189). HPA dysregulation may lead to excessive inflammation via increases in the levels of circulatory inflammatory cytokines,
decreases in anti-inflammatory cytokines, and alterations in the expression of genes involved in immune activation of peripheral blood
cells (see 177).
296 Comment
Table 1: continued
Effects of specific stressors Notes
Disease susceptibility An increase in susceptibility has been noted across several species to:
— general disease and somatic disorders (see 100, 103);
— various cancers (190, 191);
— pancreatitis and pancreatic tumours (192, 193);
— gastrointestinal disorders (194);
— thyroid pathology (195);
— multiple sclerosis (171);
— inflammatory bowel disease (142, 196);
— cardiovascular disease (197–199);
— accelerated ageing and age-related disorders (187); and
— musculoskeletal injury (200).
Stressors can increase oxidative stress, triggering inflammatory pathways associated with type-2 diabetes, cardiovascular disease,
osteoporosis, arthritis, some cancers, and susceptibility to some infections (e.g. 201–205).
Handling Causes biological changes, affecting wellbeing and/or experimental results.
Enrichment intended to mitigate stress can substantially alter brain structure, function and physiology.
Light, noise, cage position/changing etc. all affect physiology, behaviour, anxiety and experimental data (206).
Effects of handling stress may often be “missed” by researchers (23).
Early-life stress in various species Results in:
— abnormal brain development, leading to early-life psychopathologies and adult chronic mental illnesses (e.g. 207, 208);
— epigenetic modifications associated with depression and suicidal behaviour in later life (e.g. 102, 209–211); and
— elevated morbidity and mortality from chronic diseases of ageing, including vascular disease, autoimmune disorders, and premature
mortality (e.g. 141, 212).
Stress from maternal deprivation has negative effects on growth rates in rats, and adversely affects circadian clock and stress
responses (72).
Traumatic stress in humans Studies on PTSD patients have shown that:
— acute stress affects glucose metabolism, inflammation and components of the immune system associated with type-2 diabetes (201);
— serious long-term consequences include hypertension, heart attacks and stroke, as well as increased risk of obesity, Alzheimer’s
disease, and AIDS dementia complex (213).
Comment 297
inflammatory responses (156). Indels (genomic
insertions and deletions) also affect major histo-
compatibility complex (MHC) genes, which are
critical to immune responses, and are associated
with differences in response to infections, as well
as susceptibility to autoimmune diseases. These
are further confounded by sex-related and strain-
related differences. It has also been shown that the
susceptibility and responsiveness of mice to stres-
sors varies with the strain (157, 158). Stress also
affects sleep, and conversely sleep perturbations
exacerbate and sensitise individuals to stress, with
all the attendant consequences. These conse-
quences are also strain-dependent, and intimately
linked to the CRH/HPA system (159).
Additionally, the adverse consequences of stress
are multigenerational, as the associated epigenetic
mechanisms affect the germline. This is likely to
have significant consequences for animals, and
their offspring, in laboratories: if an animal’s par-
ents or grandparents experienced a stressful labo-
ratory life and experimental procedures, and/or if
the offspring experienced significant stress in early
life, then this will compound any further stress
that they experience as adults, in turn compound-
ing species differences and the translation of data
to humans.
Overall, these observations of detrimental phys-
iological effects and the general mechanisms
behind them have been detailed in many species
(including humans), and throughout the evolution-
ary scale from monkeys to rodents. The minutiae of
the genes and biochemical pathways responsible,
and their manifestations, may differ to some
degree, but there are common mechanisms and
adverse effects in all species examined to date. It
must be concluded that laboratory life for animals
used in experiments has serious and intractable
consequences for their welfare, and for the quality
and human relevance of the experimental data
obtained (which, in any case, are already of debat-
able applicability to humans, due to species differ-
ences).
Finally, I believe that this issue should be taken
much more seriously by legislators, regulators,
funders, practitioners and advocates of animal
experiments, and urge all involved to do so. The
information presented here could, and should, be a
valuable resource for project licence applicants,
ethics committees and the Home Office Inspect -
orate, for use in experimental design and harm–
benefit analyses, and to aid data interpretation.
Though it argues that relatively little can be done
to minimise many, if not all, stressors and stress,
it could inform attempts to do so — as well as con-
trolling variable factors and mitigating negative
consequences, etc. The information could also be
factored into existing guidance for the strategic
planning of animal experiments, since stress
impacts all areas of this planning, including study
objectives, species/strain selection, experimental
procedures, analgesia, training of staff, and so on.
Acknowledgements
This review was funded by Cruelty Free
International Trust, London, UK. It was based on
previous (not published) work conducted by the
same author in 2011, funded by the New England
Anti-Vivisection Society (NEAVS), Boston, USA, in
his role as its Science Director.
References
1. Bailey, J. (2017). Does the stress inherent to labora-
tory life and experimentation on animals adversely
affect research data. ATLA 45, 299–301.
2. Anon. (2007). Stress. Saunders Comprehensive
Veterinary Dictionary, 3rd edn. Available at: http://
medical-dictionary.thefreedictionary.com/Stress
(Accessed 19.09.18).
3. Ferdowsian, H.R. & Beck, N. (2011). Ethical and
scientific considerations regarding animal testing
and research. PLoS One 6, e24059.
4. Rey, S., Huntingford, F.A., Boltaña, S., Vargas, R.,
Knowles, T.G. & Mackenzie, S. (2015). Fish can
show emotional fever: Stress-induced hyperthermia
in zebrafish. Proceedings of the Biological Sciences
B282, 20152266.
5. Tran, S., Nowicki, M., Fulcher, N., Chatterjee, D. &
Gerlai, R. (2016). Interaction between handling
induced stress and anxiolytic effects of ethanol in
zebrafish: A behavioral and neurochemical analy-
sis. Behavioural Brain Research 298, 278–285.
6. Rambo, C.L., Mocelin, R., Marcon, M., Villanova,
D., Koakoski, G., de Abreu, M.S., Oliveira, T.A.,
Barcellos, L.J.G., Piato, A.L. & Bonan, C.D. (2017).
Gender differences in aggression and cortisol levels
in zebrafish subjected to unpredictable chronic stress.
Physiology & Behavior 171, 50–54.
7. Fulcher, N., Tran, S., Shams, S., Chatterjee, D. &
Gerlai, R. (2017). Neurochemical and behavioral
responses to unpredictable chronic mild stress fol-
lowing developmental isolation: The zebrafish as a
model for major depression. Zebrafish 14, 23–34.
8. Zimmermann, F.F., Altenhofen, S., Kist, L.W., Leite,
C.E., Bogo, M.R., Cognato, G.P. & Bonan, C.D.
(2016). Unpredictable chronic stress alters adenosine
metabolism in zebrafish brain. Molecular Neuro -
biology 53, 2518–2528.
9. Maestripieri, D. & Hoffman, C.L. (2011). Chronic
stress, allostatic load, and aging in nonhuman pri-
mates. Development & Psychopathology 23, 1187–
1195.
10. National Research Council (US) Committee on
Recognition and Alleviation of Distress in Labor -
atory Animals (2008). Recognition and Alleviation of
Pain and Distress in Laboratory Animals, 198pp.
Wash ing ton, DC, USA: National Academies Press.
Avail able at: https://www.ncbi.nlm.nih.gov/books/
NBK4032/ (Accessed 13.09.17).
11. Moberg, G.P. & Mench, J.A. (eds) (2000). The Biology
of Animal Stress: Basic Principles and Implications
for Animal Welfare, 377pp. Wallingford, UK: CABI
Publishing.
298 Comment
12. Tracey, K.J. (2009). Reflex control of immunity.
Nature Reviews Immunology 9, 418–428.
13. Sternberg, E.M. (2006). Neural regulation of innate
immunity: A coordinated nonspecific host response
to pathogens. Nature Reviews Immunology 6, 318–
328.
14. Glaser, R. & Kiecolt-Glaser, J.K. (2005). Stress-
induced immune dysfunction: Implications for
health. Nature Reviews Immunology 5, 243–251.
15. Altholtz, L.Y., Fowler, K.A., Badura, L.L. & Kovacs,
M.S. (2006). Comparison of the stress response in
rats to repeated isoflurane or CO2:O2anesthesia
used for restraint during serial blood collection via
the jugular vein. Journal of the American Assoc -
iation for Laboratory Animal Science 45(3), 17–22.
16. Balcombe, J.P., Barnard, N.D. & Sandusky, C.
(2004). Laboratory routines cause animal stress.
Con temporary Topics in Laboratory Animal Science
43, 42–51.
17. Meijer, M.K., Sommer, R., Spruijt, B.M., van
Zutphen, L.F. & Baumans, V. (2007). Influence of
environmental enrichment and handling on the
acute stress response in individually housed mice.
Laboratory Animals 41, 161–173.
18. Gurfein, B.T., Stamm, A.W., Bacchetti, P., Dall man,
M.F., Nadkarni, N.A., Milush, J.M., Touma, C.,
Palme, R., Di Borgo, C.P., Fromentin, G., Lown-
Hecht, R., Konsman, J.P., Acree, M., Premenko-
Lanier, M., Darcel, N., Hecht, F.M. & Nixon, D.F.
(2012). The calm mouse: An animal model of stress
reduction. Molecular Medicine 18, 606–617.
19. Obernier, J.A. & Baldwin, R.L. (2006). Establishing
an appropriate period of acclimatization following
transportation of laboratory animals. ILAR Journal
47, 364–369.
20. Balcombe, J.P. (2006). Laboratory environments and
rodents’ behavioural needs: A review. Laboratory
Animals 40, 217–235.
21. Meijer, M.K., Spruijt, B.M., van Zutphen, L.F.M. &
Baumans, V. (2006). Effect of restraint and injec-
tion methods on heart rate and body temperature in
mice. Laboratory Animals 40, 382–391.
22. Novak, M.A., Hamel, A.F., Kelly, B.J., Dettmer,
A.M. & Meyer, J.S. (2013). Stress, the HPA axis,
and nonhuman primate well-being: A review.
Applied Animal Behaviour Science 143, 135–149.
23. Gouveia, K. & Hurst, J.L. (2013). Reducing mouse
anxiety during handling: Effect of experience with
handling tunnels. PLoS One 8, e66401.
24. Conlee, K.M., Stephens, M.L., Rowan, A.N. & King,
L.A. (2005). Carbon dioxide for euthanasia: Con -
cerns regarding pain and distress, with special ref-
erence to mice and rats. Laboratory Animals 39,
137–161.
25. Wong, D., Makowska, I.J. & Weary, D.M. (2013).
Rat aversion to isoflurane versus carbon dioxide.
Biology Letters 9, 20121000.
26. Moody, C.M. & Weary, D.M. (2014). Mouse aversion
to isoflurane versus carbon dioxide gas. Applied
Animal Behaviour Science 158, 95–101.
27. Fyer, M.R., Uy, J., Martinez, J., Goetz, R., Klein,
D.F., Fyer, A., Liebowitz, M.R. & Gorman, J. (1987).
CO2challenge of patients with panic disorder.
American Journal of Psychiatry 144, 1080–1082.
28. Danneman, P.J., Stein, S. & Walshaw, S.O. (1997).
Humane and practical implications of using carbon
dioxide mixed with oxygen for anesthesia or
euthanasia of rats. Laboratory Animal Science 47,
376–385.
29. Anestis, S.F. (2009). Urinary cortisol responses to
unusual events in captive chimpanzees (Pan trog -
lodytes). Stress 12, 49–57.
30. Whitten, P.L., Stavisky, R., Aureli, F. & Russell, E.
(1998). Response of fecal cortisol to stress in captive
chimpanzees (Pan troglodytes). American Journal
of Primatology 44, 57–69.
31. Springer, D.A. & Baker, K.C. (2007). Effect of keta-
mine anesthesia on daily food intake in Macaca
mulatta and Cercopithecus aethiops. American
Journal of Primatology 69, 1080–1092.
32. Martin, C. (2014). Contributions and complexities
from the use of in vivo animal models to improve
understanding of human neuroimaging signals.
Frontiers in Neuroscience 8, 211.
33. Bowers, S.L., Bilbo, S.D., Dhabhar, F.S. & Nelson,
R.J. (2008). Stressor-specific alterations in corticos-
terone and immune responses in mice. Brain,
Behavior, & Immunity 22, 105–113.
34. Maninger, N., Mason, W., Ruys, J., Mendoza, S. &
Maninger, C. (2010). Acute and chronic stress
increase DHEAS concentrations in rhesus monkeys.
Psychoneuroendocrinology 35, 1055–1062.
35. Lee, J.I., Shin, J.S., Lee, J.E., Jung, W.Y., Lee, G.,
Kim, M.S., Park, C.G. & Kim, S.J. (2013). Changes
of N/L ratio and cortisol levels associated with
experimental training in untrained rhesus mac -
aques. Journal of Medical Primatology 42, 10–14.
36. Reinhardt, V., Liss, C. & Stevens, C. (1995).
Restraint methods of laboratory non-human pri-
mates: A critical review. Animal Welfare 4, 221–
238.
37. de Meijer, V.E., Le, H.D., Meisel, J.A. & Puder, M.
(2010). Repetitive orogastric gavage affects the phe-
notype of diet-induced obese mice. Physiology &
Behavior 100, 387–393.
38. Õkva, K., Tamoseviciute, E., Ciziute, A., Pokk, P.,
Ruksenas, O. & Nevalainen, T. (2006). Refinements
for intragastric gavage in rats. Scandinavian Jour -
nal of Laboratory Animal Sciences 33, 243–252.
39. Gonzales, C., Zaleska, M.M., Riddell, D.R., Atchison,
K.P., Robshaw, A., Zhou, H. & Sukoff Rizzo, S.J.
(2014). Alternative method of oral administration by
peanut butter pellet formulation results in target
engagement of BACE1 and attenuation of gavage-
induced stress responses in mice. Pharmacology,
Biochemistry & Behavior 126, 28–35.
40. Bonnichsen, M., Dragsted, N. & Hansen, A.K.
(2005). The welfare impact of gavaging laboratory
rats. Animal Welfare 14, 223–227.
41. Brown, A.P., Dinger, N. & Levine, B.S. (2000).
Stress produced by gavage administration in the
rat. Contemporary Topics in Laboratory Animal
Science 39, 17–21.
42. Walker, M.K., Boberg, J.R., Walsh, M.T., Wolf, V.,
Trujillo, A., Duke, M.S., Palme, R. & Felton, L.A.
(2012). A less stressful alternative to oral gavage for
pharmacological and toxicological studies in mice.
Toxicology & Applied Pharmacology 260, 65–69.
43. Morton, D.B., Abbot, D., Barclay, R., Close, B.S.,
Ewbank, R., Gask, D., Heath, M., Mattic, S., Poole,
T., Seamer, J., Southee, J., Thompson, A., Trussell,
B., West, C. & Jennings, M. (1993). Removal of
blood from laboratory mammals and birds. First
report of the BVA/FRAME/RSPCA/UFAW Joint
Working Group on Refinement. Laboratory Ani -
mals 27, 1–22.
44. Sarlis, N.J. (1991). Chronic blood sampling tech-
niques in stress experiments in the rat — A mini
Comment 299
review. Animal Technology 42, 51–59.
45. Capitanio, J.P., Mendoza, S.P. & McChesney, M.
(1996). Influences of blood sampling procedures on
basal hypothalamic-pituitary-adrenal hormone lev-
els and leukocyte values in rhesus macaques
(Macaca mulatta). Journal of Medical Primatology
25, 26–33.
46. Teilmann, A.C., Kalliokoski, O., Sorensen, D.B.,
Hau, J. & Abelson, K.S. (2014). Manual versus
automated blood sampling: Impact of repeated
blood sampling on stress parameters and behavior
in male NMRI mice. Laboratory Animals 48, 278–
291.
47. Teilmann, A.C., Nygaard Madsen, A., Holst, B., Hau,
J., Rozell, B. & Abelson, K.S. (2014). Physiological
and pathological impact of blood sampling by retro-
bulbar sinus puncture and facial vein phlebotomy in
laboratory mice. PLoS One 9, e113225.
48. Burnett, J.E. (2011). Dried blood spot sampling:
Practical considerations and recommendation for
use with preclinical studies. Bioanalysis 3, 1099–
1107.
49. Wickremsinhe, E.R. & Perkins, E.J. (2015). Using
dried blood spot sampling to improve data quality
and reduce animal use in mouse pharmacokinetic
studies. Journal of the American Association for Lab -
oratory Animal Science 54(2), 139–144.
50. Langkilde, T. & Shine, R. (2006). How much stress
do researchers inflict on their study animals? A
case study using a scincid lizard, Eulamprus heat-
wolei. Journal of Experimental Biology 209, 1035–
1043.
51. Jennings, M., Prescott, M.J., Buchanan-Smith, H.M.,
Gamble, M.R., Gore, M., Hawkins, P., Hubrecht, R.,
Hudson, S., Jennings, M., Keeley, J.R., Morris, K.,
Morton, D.B., Owen, S., Pearce, P.C., Prescott, M.J.,
Robb, D., Rumble, R.J., Wolfensohn, S. & Buist, D.
(2009). Refinements in husbandry, care and common
procedures for non-human primates: Ninth report of
the BVAAWF/FRAME/RSPCA/UFAW Joint Working
Group on Refinement. Laboratory Animals 43, Suppl.
1, 1–47.
52. Willems, R.A. (2009). Regulatory issues regarding
the use of food and water restriction in laboratory
animals. Lab Animal 38, 325–328.
53. Calisi, R.M. & Bentley, G.E. (2009). Lab and field
experiments: Are they the same animal? Hormones
& Behavior 56, 1–10.
54. Meijer, T. & Schwabl, H. (1989). Hormonal patterns
in breeding and nonbreeding kestrels, Falco tinnun-
culus: Field and laboratory studies. General &
Comparative Endocrinology 74, 148–160.
55. Marra, P.P., Kevin, T.L. & Bruce, L.T. (1995).
Plasma corticosterone levels in two species of
Zonotrichia sparrows under captive and free-living
conditions. Wilson Bulletin 107, 305–296.
56. Barrett, A.M. & Stockham, M.A. (1963). The effect
of housing conditions and simple experimental pro-
cedures upon the corticosterone level in the plasma
of rats. Journal of Endocrinology 26, 97–105.
57. Cavigelli, S.A., Guhad, F.A., Ceballos, R.M., Whetzel,
C.A., Nevalainen, T., Lang, C.M. & Klein, L.C. (2006).
Fecal corticoid metabolites in aged male and female
rats after husbandry-related disturbances in the
colony room. Journal of the American Association for
Laboratory Animal Science 45(6), 17–21.
58. Castelhano-Carlos, M.J. & Baumans, V. (2009). The
impact of light, noise, cage cleaning and in-house
transport on welfare and stress of laboratory rats.
Laboratory Animals 43, 311–327.
59. Baldwin, A.L. (2007). Effects of noise on rodent
physiology. International Journal of Comparative
Psychology 20, 134–144.
60. Pines, M.K., Kaplan, G. & Rogers, L.J. (2004).
Stressors of common marmosets (Callithrix jacchus)
in the captive environment: Effects on behaviour and
cortisol levels. Folia Primatologica 75, 317–318.
61. Schreuder, M.F., Fodor, M., van Wijk, J.A. &
Delemarre-van de Waal, H.A. (2007). Weekend ver-
sus working day: Differences in telemetric blood
pressure in male Wistar rats. Laboratory Animals
41, 86–91.
62. Patterson-Kane, E.G. & Farnworth, M.J. (2006).
Noise exposure, music, and animals in the labora-
tory: A commentary based on Laboratory Animal
Refinement and Enrichment Forum (LAREF) dis-
cussions. Journal of Applied Animal Welfare Science
9, 327–332.
63. Kirillov, O.I., Khasina, E.I. & Durkina, V.B. (2003).
[Effect of stress on postnatal growth in weight of rat
body and adrenal gland]. Ontogenez 34, 371–376.
64. Bernátová, I., Púzserová, A., Navarová, J., Csizmad -
iová, Z. & Zeman, M. (2007). Crowding-induced alter-
ations in vascular system of Wistar-Kyoto rats: Role
of nitric oxide. Physiological Research 56, 667–669.
65. Armario, A., Castellanos, J.M. & Balasch, J. (1984).
Effect of crowding on emotional reactivity in male
rats. Neuroendocrinology 39, 330–333.
66. Cyr, N.E. & Romero, L.M. (2008). Fecal glucocorti-
coid metabolites of experimentally stressed captive
and free-living starlings: Implications for conserva-
tion research. General & Comparative Endocrin -
ology 158, 20–28.
67. Wingfield, J.C. & Kitaysky, A.S. (2002). Endocrine
responses to unpredictable environmental events:
Stress or anti-stress hormones. Integrative & Com -
parative Biology 42, 600–609.
68. Romero, L.M. & Wingfield, J.C. (1999). Alterations
in hypothalamic-pituitary-adrenal function associ-
ated with captivity in Gambel’s white-crowned
sparrows (Zonotrichia leucophrys gambelii). Com -
parative Biochemistry & Physiology. Part B, Bio -
chemistry & Molecular Biology 122, 13–20.
69. Ferland, C.L. & Schrader, L.A. (2011). Cage mate
separation in pair-housed male rats evokes an
acute stress corticosterone response. Neuroscience
Letters 489, 154–158.
70. Clay, A.W., Bloomsmith, M.A., Marr, M.J. & Maple,
T.L. (2009). Habituation and desensitization as
methods for reducing fearful behavior in singly
housed rhesus macaques. American Journal of
Primatology 71, 30–39.
71. Lilly, A.A., Mehlman, P.T. & Higley, J.D. (1999).
Trait-like immunological and hematological meas-
ures in female rhesus across varied environmental
conditions. American Journal of Primatology 48,
197–223.
72. Allen, K.P., Dwinell, M.R., Zappa, A., Temple, A. &
Thulin, J. (2013). Comparison of 2 rat breeding
schemes using conventional caging. Journal of the
American Association of Laboratory Animal Science
52(2), 142–145.
73. Burn, C.C., Peters, A., Day, M.J. & Mason, G.J.
(2006). Long-term effects of cage-cleaning frequency
and bedding type on laboratory rat health, welfare,
and handleability: A cross-laboratory study.
Laboratory Animals 40, 353–370.
74. Meller, A., Kasanen, I., Ruksenas, O., Apanaviciene,
300 Comment
N., Baturaite, Z., Voipio, H.M. & Nevalainen, T.
(2011). Refining cage change routines: Comparison of
cardiovascular responses to three different ways of
cage change in rats. Laboratory Animals 45, 167–
173.
75. Line, S.W., Markowitz, H., Morgan, K.N. & Strong,
S. (1991). Effects of cage size and environmental
enrichment on behavioral and physiological res -
ponses of rhesus macaques to the stress of daily
events. In Through the Looking Glass (ed. M.A.
Novak & A.J. Petto), pp. 160–179. Washington, DC,
USA: American Psychological Association.
76. Clarke, A.S., Mason, W.A. & Mendoza, S.P. (1994).
Heart rate patterns under stress in three species of
macaques. American Journal of Primatology 33,
133–148.
77. Wurbel, H. (2001). Ideal homes? Housing effects on
rodent brain and behaviour. Trends in Neuro -
sciences 24, 207–211.
78. Wolfer, D.P., Litvin, O., Morf, S., Nitsch, R.M.,
Lipp, H.P. & Würbel, H. (2004). Laboratory animal
welfare: Cage enrichment and mouse behaviour.
Nature, London 432, 821–822.
79. Burn, C.C., Deacon, R.M. & Mason, G.J. (2008).
Marked for life? Effects of early cage-cleaning
frequency, delivery batch, and identification tail-
marking on rat anxiety profiles. Developmental
Psych o biology 50, 266–277.
80. Schapiro, S.J., Lambeth, S.P., Jacobsen, K.R.,
Williams, L.E., Nehete, B.N. & Nehete, P.N. (2012).
Physiological and welfare consequences of trans-
port, relocation, and acclimatization of chimpanzees
(Pan troglodytes). Applied Animal Behaviour Science
137, 183–193.
81. Wolfensohn, S.E. (1997). Brief review of scientific
studies of the welfare implications of transporting
primates. Laboratory Animals 31, 303–305.
82. Honess, P.E., Johnson, P.J. & Wolfensohn, S.E.
(2004). A study of behavioural responses of non-
human primates to air transport and re-housing.
Laboratory Animals 38, 119–132.
83. Ferreira, C.S., Vasconcellos, R.S., Pedreira, R.S.,
Silva, F.L., Sá, F.C., Kroll, F.S., Maria, A.P., Vent -
urini, K.S. & Carciofi, A.C. (2014). Alterations to
oxidative stress markers in dogs after a short-term
stress during transport. Journal of Nutritional
Science 3, e27.
84. Line, S.W., Morgan, K.N., Markowitz, H. & Strong,
S. (1989). Heart rate and activity of rhesus monkeys
in response to routine events. Laboratory Primate
Newsletter 28, 9–12.
85 Sharp, J., Zammit, T., Azar, T. & Lawson, D. (2003).
Are “by-stander” female Sprague-Dawley rats
affected by experimental procedures. Contemporary
Topics in Laboratory Animal Science 42, 19–27.
86. Gilmore, A.J., Billing, R.L. & Einstein, R. (2008).
The effects on heart rate and temperature of mice
and vas deferens responses to noradrenaline when
their cage mates are subjected to daily restraint
stress. Laboratory Animals 42, 140–148.
87. Bowers, C.L., Crockett, C.M. & Bowden, D.M.
(1998). Differences in stress reactivity of laboratory
macaques measured by heart period and respira-
tory sinus arrhythmia. American Journal of Prima -
tology 45, 245–261.
88. Baumans, V. (2005). Environmental enrichment for
laboratory rodents and rabbits: Requirements of
rodents, rabbits, and research. ILAR Journal 46,
162–170.
89. Hutchinson, E., Avery, A. & Vandewoude, S.
(2005). Environmental enrichment for laboratory
rodents. ILAR Journal 46, 148–161.
90. Reinhardt, V. & Reinhardt, A. (2000). Blood collec-
tion procedure of laboratory primates: A neglected
variable in biomedical research. Journal of Applied
Animal Welfare Science 3, 321–333.
91. Olsson, A. & Dahlborn, K. (2002). Improving hous-
ing conditions for laboratory mice: A review of
‘environmental enrichment’. Laboratory Animals
36, 243–270.
92. Zimmermann, A., Stauffacher, M., Langhans, W. &
Würbel, H. (2001). Enrichment-dependent differ-
ences in novelty exploration in rats can be explained
by habituation. Behavioural Brain Research 121,
11–20.
93. Würbel, H., Chapman, R. & Rutland, C. (1998).
Effect of feed and environmental enrichment on
development of stereotypic wire-gnawing in labora-
tory mice. Applied Animal Behaviour Science 60,
69–81.
94. Powell, S.B., Newman, H.A., McDonald, T.A.,
Bugenhagen, P. & Lewis, M.H. (2000). Develop -
ment of spontaneous stereotyped behavior in deer
mice: Effects of early and late exposure to a more
complex environment. Developmental Psychobiol -
ogy 37, 100–108.
95. Callard, M.D., Bursten, S.N. & Price, E.O. (2000).
Repetitive backflipping behaviour in captive roof
rats (Rattus rattus) and the effects of cage enrich-
ment. Animal Welfare 9, 139–152.
96. Balcombe, J. (2010). Laboratory rodent welfare:
Thinking outside the cage. Journal of Applied Ani -
mal Welfare Science 13, 77–88.
97. Leach, M.C., Ambrose, N., Bowell, V.J. & Morton,
D.B. (2000). The development of a novel form of
mouse cage enrichment. Journal of Applied
Animal Wel fare Science 3, 81–91.
98. Würbel, H., Freire, R. & Nicol, C.J. (1998). Pre -
vention of stereotypic wire-gnawing in laboratory
mice: Effects on behaviour and implications for
stereotypy as a coping response. Behavioural Pro -
cesses 42, 61–72.
99. McEwen, B.S. & Seeman, T. (1999). Protective and
damaging effects of mediators of stress. Elabor -
ating and testing the concepts of allostasis and allo-
static load. Annals of the New York Academy of
Sciences 896, 30–47.
100. Kurokawa, K., Kuwano, Y., Tominaga, K., Kawai,
T., Katsuura, S., Yamagishi, N., Satake, Y., Kajita,
K., Tanahashi, T. & Rokutan, K. (2010). Brief nat-
uralistic stress induces an alternative splice vari-
ant of SMG-1 lacking exon 63 in peripheral
leukocytes. Neuroscience Letters 484, 128–132.
101. Champagne, F.A. (2010). Epigenetic influence of
social experiences across the lifespan. Develop mental
Psychobiology 52, 299–311.
102. Murgatroyd, C. & Spengler, D. (2011). Epigenetic
programming of the HPA axis: Early life decides.
Stress 14, 581–589.
103. Wright, R. (2011). Epidemiology of stress and
asthma: From constricting communities and fragile
families to epigenetics. Immunology & Allergy
Clinics of North America 31, 19–39.
104. Hoffmann, A., Zimmermann, C.A. & Spengler, D.
(2015). Molecular epigenetic switches in neurode-
velopment in health and disease. Frontiers in
Behavioral Neuroscience 9, 120.
105. Grace, C.E., Kim, S.J. & Rogers, J.M. (2011). Mat -
Comment 301
ernal influences on epigenetic programming of the
developing hypothalamic–pituitary–adrenal axis.
Birth Defects Research. Part A, Clinical & Mol -
ecular Teratology 91, 797–805.
106. Gudsnuk, K. & Champagne, F.A. (2012). Epi -
genetic influence of stress and the social environ-
ment. ILAR Journal 53, 279–288.
107. Champagne, F.A. (2012). Interplay between social
experiences and the genome: Epigenetic conse-
quences for behavior. Advances in Genetics 77, 33–
57.
108. Wright, R.J. & Enlow, M.B. (2008). Maternal stress
and perinatal programming in the expression of
atopy. Expert Review of Clinical Immunology 4, 535–
538.
109. McGowan, P.O., Sasaki, A., D’Alessio, A.C., Dymov,
S., Labonte, B., Szyf, M., Turecki, G. & Meaney, M.J.
(2009). Epigenetic regulation of the glucocorticoid
receptor in human brain associates with childhood
abuse. Nature Neuroscience 12, 342–348.
110. Uddin, M., Aiello, A.E., Wildman, D.E., Koenen,
K.C., Pawelec, G., de los Santos, R., Goldmann, E.
& Galea, S. (2010). Epigenetic and immune func-
tion profiles associated with posttraumatic stress
disorder. Proceedings of the National Academy of
Sciences of the USA 107, 9470–9475.
111. Biamonti, G. & Caceres, J.F. (2009). Cellular stress
and RNA splicing. Trends in Biochemical Sciences
34, 146–153.
112. Katsuura, S., Kuwano, Y., Yamagishi, N., Kuro -
kawa, K., Kajita, K., Akaike, Y., Nishida, K.,
Masuda, K., Tanahashi, T. & Rokutan, K. (2012).
MicroRNAs miR-144/144* and miR-16 in peripheral
blood are potential biomarkers for naturalistic stress
in healthy Japanese medical students. Neuroscience
Letters 516, 79–84.
113. Hapuarachchi, J.R., Chalmers, A.H., Winefield,
A.H. & Blake-Mortimer, J.S. (2003). Changes in
clinically relevant metabolites with psychological
stress parameters. Behavioral Medicine 29, 52–59.
114. Sivonova, M., Zitnanova, I., Hlincikova, L., Skod -
acek, I., Trebaticka, J. & Durackova, Z. (2004).
Oxidative stress in university students during
examinations. Stress 7, 183–188.
115. Knickelbein, K.Z., Flint, M., Jenkins, F. & Baum,
A. (2008). Psychological stress and oxidative
damage in lymphocytes of aerobically fit and
unfit individuals. Journal of Applied Biobehavioral
Research 13, 1–19.
116. Maes, M. (2001). Psychological stress and the
inflam matory response system. Clinical Science 101,
193–194.
117. Videan, E.N., Heward, C.B., Chowdhury, K., Plum -
mer, J., Su, Y. & Cutler, R.G. (2009). Com parison of
biomarkers of oxidative stress and cardiovascular
disease in humans and chimpanzees (Pan
troglodytes). Comparative Medicine 59, 287–296.
118. Wang, L., Muxin, G., Nishida, H., Shirakawa, C.,
Sato, S. & Konishi, T. (2007). Psychological stress-
induced oxidative stress as a model of sub-healthy
condition and the effect of TCM. Evidence-Based
Complementary & Alternative Medicine 4, 195–
202.
119. Lewis, K.N., Andziak, B., Yang, T. & Buffenstein,
R. (2013). The naked mole-rat response to oxidative
stress: Just deal with it. Antioxidants & Redox
Signaling 19, 1388–1399.
120. Webster, J. (2002). Animal Welfare: A Cool Eye To -
wards Eden, 284pp. Oxford, UK: Blackwell Science.
121. Chance, P. (2003). Learning and Behavior (5th
Edition). Belmont, CA, USA: Thomson Wadworth.
122. Barnum, C.J., Blandino, P.J. & Deak, T. (2007).
Adaptation in the corticosterone and hyperthermic
responses to stress following repeated stressor
exposure. Journal of Neuroendocrinology 19, 632–
642.
123. Gruen, M.E., Thomson, A.E., Clary, G.P., Hamilton,
A.K., Hudson, L.C., Meeker, R.B. & Sherman, B.L.
(2013). Conditioning laboratory cats to handling and
transport. Lab Animal 42, 385–389.
124. Longordo, F., Fan, J., Steimer, T., Kopp, C. &
Luthi, A. (2011). Do mice habituate to “gentle han-
dling?” A comparison of resting behavior, corticos-
terone levels and synaptic function in handled and
undisturbed C57BL/6J mice. Sleep 34, 679–681.
125. Kramer, K., van de Weerd, H., Mulder, A., Van
Heijningen, C., Baumans, V., Remie, R., Voss, H.P.
& van Zutphen, B.F. (2004). Effect of conditioning
on the increase of heart rate and body temperature
provoked by handling in the mouse. ATLA 32,
Suppl. 1A, 177–181.
126. Clement, J.G., Mills, P. & Brockway, B. (1989). Use
of telemetry to record body temperature and activ-
ity in mice. Journal of Pharmacological Methods
21, 129–140.
127. Swan, M.P. & Hickman, D.L. (2014). Evaluation of
the neutrophil–lymphocyte ratio as a measure of
distress in rats. Lab Animal 43, 276–282.
128. Anon. (2009). The Revision of the UK Directive on
the Protection of Animals Used for Scientific Pur -
poses, Volume II: Evidence, 252pp. London, UK:
The Stationery Office. Available at: http://www.
publications.parliament.uk/pa/ld200809/ldselect/
ldeucom/164/164ii.pdf (Accessed 14.09.18).
129. Anon. (2006). Action to be Taken Against Laos,
Vietnam and Cambodia Monkey Business. London,
UK: Cruelty Free International. Available at:
https://www.crueltyfreeinternational.org/breaking-
news/action-be-taken-against-laos-vietnam-and-
cambodia-monkey-business (Accessed 19.09.18).
130. Anon. (2008). Cambodia: The Trade in Primates for
Research. London, UK: Cruelty Free International.
131. Anon. (2010). Mauritius: The Trade in Primates for
Research. London, UK: Cruelty Free International.
132. Suomi, S.J. (1991). Primate separation models of
affective disorders. In Neurobiology of Learning,
Emotion and Affect (ed. J. Madden), pp. 195–214.
New York, NY, USA: Lippincott Williams and Wil -
kins.
133. Nakamichi, M., Cho, F. & Minami, T. (1990).
Mother–infant interactions of wild-born, individu-
ally-caged cynomolgus monkeys (Macaca fascicu-
laris) during the first 14 weeks of infant life.
Primates 31, 213–224.
134. Maestripieri, D., Martel, F.L., Nevison, C.M.,
Simpson, M.J. & Keverne, E.B. (1991). Anxiety in
rhesus monkey infants in relation to interactions
with their mother and other social companions.
Developmental Psychobiology 24, 571–581.
135. Camus, S.M., Rochais, C., Blois-Heulin, C., Li, Q.,
Hausberger, M. & Bezard, E. (2013). Birth origin
differentially affects depressive-like behaviours:
Are captive-born cynomolgus monkeys more vul-
nerable to depression than their wild-born counter-
parts? PLoS One 8, e67711.
136. Dettmer, A.M., Novak, M.A., Suomi, S.J. & Meyer,
J.S. (2012). Physiological and behavioral adapta-
tion to relocation stress in differentially reared rhe-
302 Comment
sus monkeys: Hair cortisol as a biomarker for anx-
iety-related responses. Psychoneuroendocrinol ogy
37, 191–199.
137. Butkevich, I., Mikhailenko, V., Semionov, P.,
Bagaeva, T., Otellin, V. & Aloisi, A.M. (2009). Eff -
ects of maternal corticosterone and stress on
behavioral and hormonal indices of formalin pain
in male and female offspring of different ages.
Hormones & Behavior 55, 149–157.
138. Heim, C., Newport, D.J., Wagner, D., Wilcox, M.M.,
Miller, A.H. & Nemeroff, C.B. (2002). The role of
early adverse experience and adulthood stress in the
prediction of neuroendocrine stress reactivity in
women: A multiple regression analysis. Depression &
Anxiety 15, 117–125.
139. Heim, C., Mletzko, T., Purselle, D., Musselman,
D.L. & Nemeroff, C.B. (2008). The dexamethasone/
corticotropin-releasing factor test in men with
major depression: Role of childhood trauma.
Biological Psychiatry 63, 398–405.
140. Christian, L.M. (2012). Psychoneuroimmunology
in pregnancy: Immune pathways linking stress
with maternal health, adverse birth outcomes, and
fetal development. Neuroscience & Biobehavioral
Reviews 36, 350–361.
141. Kinnally, E.L., Feinberg, C., Kim, D., Ferguson, K.,
Leibel, R., Coplan, J.D. & John, M.J. (2011). DNA
methylation as a risk factor in the effects of early
life stress. Brain, Behavior, & Immunity 25, 1548–
1553.
142. Chang, L. (2011). The role of stress on physiologic
responses and clinical symptoms in irritable bowel
syndrome. Gastroenterology 140, 761–765.
143. Bird, A.P. (1986). CpG-rich islands and the func-
tion of DNA methylation. Nature, London 321,
209–213.
144. Oberlander, T.F., Weinberg, J., Papsdorf, M.,
Grunau, R., Misri, S. & Devlin, A.M. (2008). Pre -
natal exposure to maternal depression, neonatal
methylation of human glucocorticoid receptor gene
(NR3C1) and infant cortisol stress responses.
Epigenetics 3, 97–106.
145. Fukuda, S. & Taga, T. (2005). Cell fate determina-
tion regulated by a transcriptional signal network
in the developing mouse brain. Anatomical Science
International 80, 12–18.
146. Zambrano, E., Martinez-Samayoa, P.M., Bautista,
C.J., Deas, M., Guillen, L., Rodriguez-Gonzalez,
G.L., Guzman, C., Larrea, F. & Nathanielsz, P.W.
(2005). Sex differences in transgenerational alter-
ations of growth and metabolism in progeny (F2) of
female offspring (F1) of rats fed a low protein diet
during pregnancy and lactation. Journal of Phys -
iology 566, 225–236.
147. Newbold, R.R., Padilla-Banks, E. & Jefferson, W.N.
(2006). Adverse effects of the model environmental
estrogen diethylstilbestrol are transmitted to sub-
sequent generations. Endocrinology 147, S11–S17.
148. Capdevila, S., Giral, M., Ruiz de la Torre, J.L.,
Russell, R.J. & Kramer, K. (2007). Acclimatization
of rats after ground transportation to a new animal
facility. Laboratory Animals 41, 255–261.
149. Anon. (2007). NC3Rs blood sampling microsite
launched. Laboratory Animals 41, 407.
150. Mason, J.W., Wool, M.S., Wherry, F.E., Pennington,
L.L., Brady, J.V. & Beer, B. (1968). Plasma growth
hormone response to avoidance sessions in the mon-
key. Psychosomatic Medicine 30, Suppl., 760–773.
151. Roberts, R.A., Soames, A.R., James, N.H., Gill, J.H.
& Wheeldon, E.B. (1995). Dosing-induced stress
causes hepatocyte apoptosis in rats primed by the
rodent nongenotoxic hepatocarcinogen cyproterone
acetate. Toxicology & Applied Pharmacology 135,
192–199.
152. Brenner, G.J., Cohen, N., Ader, R. & Moynihan,
J.A. (1990). Increased pulmonary metastases and
natural killer cell activity in mice following han-
dling. Life Sciences 47, 1813–1819.
153. Bailey, J. (2011). Lessons from chimpanzee-based
research on human disease: The implications of
genetic differences. ATLA 39, 527–540.
154. Bailey, J. (2014). Monkey-based research on human
disease: The implications of genetic differences.
ATLA 42, 287–317.
155. Armengol, G., Knuutila, S., Lozano, J.J., Madrigal,
I. & Caballin, M.R. (2010). Identification of human
specific gene duplications relative to other pri-
mates by array CGH and quantitative PCR.
Genomics 95, 203–209.
156. Perry, G.H., Yang, F., Marques-Bonet, T., Murphy,
C., Fitzgerald, T., Lee, A.S., Hyland, C., Stone, A.C.,
Hurles, M.E., Tyler-Smith, C., Eichler, E.E., Carter,
N.P., Lee, C. & Redon, R. (2008). Copy number vari-
ation and evolution in humans and chimpanzees.
Genome Research 18, 1698–1710.
157. Tang, X., Orchard, S.M. & Sanford, L.D. (2002).
Home cage activity and behavioral performance in
inbred and hybrid mice. Behavioural Brain
Research 136, 555–569.
158. Tang, X. & Sanford, L.D. (2002). Telemetric record-
ing of sleep and home cage activity in mice. Sleep
25, 691–699.
159. Sanford, L.D., Yang, L., Wellman, L.L., Dong, E. &
Tang, X. (2008). Mouse strain differences in the
effects of corticotropin releasing hormone (CRH) on
sleep and wakefulness. Brain Research 1190, 94–
104.
160. Mason, G.J. (1991). Stereotypies and suffering. Be -
hav ioural Processes 25, 103–115.
161. Prescott, M.J., Morton, D.B., Anderson, D., Buck -
well, A., Heath, M.S., Hubrecht, R., Jennings, M.,
Robb, M.D., Ruane, M.B. & Swallow, M.J. (2004).
Refining dog husbandry and care. Laboratory Ani -
mals 38, 1–94.
162. Hubrecht, R. (1995). The welfare of dogs in human
care. In The Domestic Dog: Its Evolution, Behav -
iour, and Interactions with People (ed. J. Serpell),
pp. 179–198. Cambridge, UK: Cambridge Univers -
ity Press.
163. Bourgeois, S.R., Vazquez, M. & Brasky, K. (2007).
Combination therapy reduces self-injurious behav-
ior in a chimpanzee (Pan troglodytes troglodytes): A
case report. Journal of Applied Animal Welfare
Science 10, 123–140.
164. Brüne, M., Brüne-Cohrs, U., McGrew, W.C. & Preu -
schoft, S. (2006). Psychopathology in great apes:
Concepts, treatment options and possible homologies
to human psychiatric disorders. Neuro science &
Biobehavioral Reviews 30, 1246–1259.
165. Lutz, C., Well, A. & Novak, M. (2003). Stereotypic
and self-injurious behavior in rhesus macaques: A
survey and retrospective analysis of environment
and early experience. American Journal of Prima -
tology 60, 1–15.
166. Novak, M.A., Meyer, J.S., Lutz, C. & S., T. (2007).
Stress and the performance of primate stereotyp-
ies. In Stereotypic Animal Behaviour: Fundamen -
tals and Applications for Welfare (ed. G. Mason &
Comment 303
J. Rushen), p. 248. Wallingford, UK: CAB Inter -
national.
167. Anon. (2009). Stress. [Nursing Times, 23.02.09].
Available at: https://www.nursingtimes.net/stress/
1995960.article (Accessed 17.09.18).
168. Wang, Y.C., Ho, U.C., Ko, M.C., Liao, C.C. & Lee,
L.J. (2012). Differential neuronal changes in medial
prefrontal cortex, basolateral amygdala and nucleus
accumbens after postweaning social isolation. Brain
Structure & Function 217, 337–351.
169. Tuchscherer, M., Kanitz, E., Puppe, B., Tuch sch -
erer, A. & Viergutz, T. (2009). Changes in endo -
crine and immune responses of neonatal pigs
exposed to a psychosocial stressor. Research in
Veterinary Science 87, 380–388.
170. Kaushal, N., Nair, D., Gozal, D. & Ramesh, V.
(2012). Socially isolated mice exhibit a blunted
homeostatic sleep response to acute sleep depriva-
tion compared to socially paired mice. Brain
Research 1454, 65–79.
171. Sorenson, M., Janusek, L. & Mathews, H. (2011).
Psychological stress and cytokine production in
multiple sclerosis: Correlation with disease symp-
tomatology. Biological Research for Nursing 15,
226–233.
172. Dhabhar, F.S. (2009). Enhancing versus suppres-
sive effects of stress on immune function: Implica -
tions for immunoprotection and immunopathology.
Neuroimmunomodulation 16, 300–317.
173. He, Y.D., Karbowski, C.M., Werner, J., Everds, N.,
Di Palma, C., Chen, Y., Higgins-Garn, M., Tran, S.,
Afshari, C.A. & Hamadeh, H.K. (2014). Common
handling procedures conducted in preclinical safety
studies result in minimal hepatic gene expression
changes in Sprague-Dawley rats. PLoS One 9,
e88750.
174. Li, H., Chen, L., Zhang, Y., Lesage, G., Zhang, Y.,
Wu, Y., Hanley, G., Sun, S. & Yin, D. (2011). Chronic
stress promotes lymphocyte reduction through TLR2
mediated PI3K signaling in a beta-arrestin 2
dependent manner. Journal of Neuro immunology
233, 73–79.
175. Xiang, L., Rehm, K., Marshall, G. & Xiang, D.B.
(2011). Effects of acute stress-induced immuno -
modulation on Th1/Th2 cytokine and catechol -
amine receptor expression in human peripheral
blood cells. Neuropsychobiology 65, 12–19.
176. Huang, C.J., Stewart, J.K., Franco, R.L., Evans,
R.K., Lee, Z.P., Cruz, T.D., Webb, H.E. & Acevedo,
E.O. (2011). LPS-stimulated tumor necrosis factor-
alpha and interleukin-6 mRNA and cytokine
responses following acute psychological stress.
Psychoneuroendocrinology 36, 1553–1561.
177. Smith, A., Conneely, K., Kilaru, V., Weiss, T.,
Bradley, B., Cubells, J., Ressler, K., Kilaru, M. &
Bradley, T. (2011). Differential immune system DNA
methylation and cytokine regulation in post-trau-
matic stress disorder. American Journal of Medical
Genetics. Part B, Neuropsychiatric Gen etics 156,
700–708.
178. Dhabhar, F.S. & McEwen, B.S. (1997). Acute stress
enhances while chronic stress suppresses cell-
mediated immunity in vivo: A potential role for
leukocyte trafficking. Brain, Behavior, & Imm unity
11, 286–306.
179. Cohen, S., Frank, E., Doyle, W.J., Skoner, D.P.,
Rabin, B.S. & Gwaltney, J.M. (1998). Types of
stressors that increase susceptibility to the com-
mon cold in healthy adults. Health Psychology 17,
214–223.
180. Blecha, F., Kelley, K.W. & Satterlee, D.G. (1982).
Adrenal involvement in the expression of delayed-
type hypersensitivity to SRBC and contact sensitiv-
ity to DNFB in stressed mice. Proceedings of the
Society for Experimental Biology & Medicine 169,
247–252.
181. Gouin, J.P., Hantsoo, L. & Kiecolt-Glaser, J.K.
(2008). Immune dysregulation and chronic stress
among older adults: A review. Neuroimmuno
-
modulation 15, 251–259.
182. Pace, T.W. & Heim, C.M. (2011). A short review on
the psychoneuroimmunology of posttraumatic
stress disorder: From risk factors to medical comor-
bidities. Brain, Behavior, & Immunity 25, 6–13.
183. Glaser, R., Robles, T.F., Sheridan, J., Malarkey,
W.B. & Kiecolt-Glaser, J.K. (2003). Mild depressive
symptoms are associated with amplified and pro-
longed inflammatory responses after influenza
virus vaccination in older adults. Archive of
General Psychiatry 60, 1009–1014.
184. Papathanassoglou, E.D., Giannakopoulou, M.,
Mpouzika, M., Bozas, E. & Karabinis, A. (2010).
Potential effects of stress in critical illness through
the role of stress neuropeptides. Nursing in Crit ical
Care 15, 204–216.
185. Nater, U.M., Whistler, T., Lonergan, W., Mletzko,
T., Vernon, S.D. & Heim, C. (2009). Impact of acute
psychosocial stress on peripheral blood gene
expression pathways in healthy men. Biological
Psychology 82, 125–132.
186. Kamezaki, Y., Katsuura, S., Kuwano, Y., Tana -
hashi, T. & Rokutan, K. (2012). Circulating cyto -
kine signatures in healthy medical students
exposed to academic examination stress. Psycho -
physiology 49, 991–997.
187. Hasan, K.M., Rahman, M.S., Arif, K.M. & Sobhani,
M.E. (2011). Psychological stress and aging: Role of
glucocorticoids (GCs). Age 34, 1421–1433.
188. Kitajima, T., Ariizumi, K., Bergstresser, P.R. &
Takashima, A. (1996). A novel mechanism of gluco-
corticoid-induced immune suppression: The
inhibiton of T cell-mediated terminal maturation of
a murine dendritic cell line. Journal of Clinical
Investigation 98, 142–147.
189. Porter, N.M. & Landfield, P.W. (1998). Stress hor-
mones and brain aging: Adding injury to insult?
Nature Neuroscience 1, 3–4.
190. Sun, X., Zhong, X., Liu, Z., Cai, H., Fan, Q., Wang,
Q., Liu, Q., Song, Y., He, S., Zhang, X. & Lu, Z.
(2010). Proteomic analysis of chronic restraint
stress-induced Gan (肝)-stagnancy syndrome in rats.
Chinese Journal of Integrative Medicine 16, 510–517.
191. Gidron, Y. & De, Z. (2010). Influence of stress and
health-behaviour on miRNA expression. Molecular
Medicine Reports 3, 455–457.
192. Binker, M.G., Binker-Cosen, A.A., Richards, D.,
Gaisano, H.Y., de Cosen, R.H. & Cosen-Binker, L.I.
(2010). Chronic stress sensitizes rats to pancreati-
tis induced by cerulein: Role of TNF-a. World
Journal of Gastroenterology 16, 5565–5581.
193. Schuller, H.M., Al-Wadei, H.A., Ullah, M.F. &
Plummer, H.K. (2012). Regulation of pancreatic can-
cer by neuropsychological stress responses: A novel
target for intervention. Carcinogenesis 33, 191–196.
194. Hong, S., Owyang, C., Hong, Z. & Owyang, W.
(2011). Corticosterone mediates reciprocal changes
in CB1 and TRPV1 receptors in primary sensory
neurons in the chronically stressed rat. Gastro -
304 Comment
enterology 140, 627–637.
195. Nadol’nik, L.I. (2010). [Stress and the thyroid
gland]. Biomeditsinskaia Khimiia 56, 443–456.
196. Rampton, D. (2011). The influence of stress on the
development and severity of immune-mediated dis-
eases. Journal of Rheumatology Supplement 88,
43–47.
197. Menezes, A., Lavie, C., Milani, R., O’Keefe, J. &
O’Keefe, L. (2011). Psychological risk factors and
cardiovascular disease: Is it all in your head?
Postgraduate Medicine 123, 165–176.
198. Gavrilovic, L., Spasojevic, N. & Dronjak, S. (2010).
Subsequent stress increases gene expression of cat-
echolamine synthetic enzymes in cardiac ventricles
of chronic-stressed rats. Endocrine 37, 425–429.
199. Eitel, I., von Knobelsdorff-Brenkenhoff, F., Bern -
hardt, P., Carbone, I., Muellerleile, K., Aldrovandi,
A., Francone, M., Desch, S., Gutberlet, M., Strohm,
O., Schuler, G., Schulz-Menger, J., Thiele, H. &
Friedrich, M.G. (2011). Clinical characteristics and
cardiovascular magnetic resonance findings in
stress (takotsubo) cardiomyopathy. JAMA 306,
277–286.
200. Allen, D., McCall, G., Loh, A., Madden, M. &
Mehan, R. (2010). Acute daily psychological stress
causes increased atrophic gene expression and
myostatin-dependent muscle atrophy. American
Journal of Physiology. Regulatory, Integrative &
Comparative Physiology 299, R889–R898.
201. Nowotny, B., Cavka, M., Herder, C., Loffler, H.,
Poschen, U., Joksimovic, L., Kempf, K., Krug, A.W.,
Koenig, W., Martin, S. & Kruse, J. (2010). Effects of
acute psychological stress on glucose metabolism
and subclinical inflammation in patients with post-
traumatic stress disorder. Hormone & Metabolic
Research 42, 746–753.
202. Marotta, F., Naito, Y., Padrini, F., Xuewei, X., Jain,
S., Soresi, V., Zhou, L., Catanzaro, R., Zhong, K.,
Polimeni, A. & Chui, D.H. (2011). Redox balance sig-
nalling in occupational stress: Modification by
nutraceutical intervention. Journal of Biological
Regulators & Homeostatic Agents 25, 221–229.
203. Jankord, R., Zhang, R., Flak, J.N., Solomon, M.B.,
Albertz, J. & Herman, J.P. (2010). Stress activation
of IL-6 neurons in the hypothalamus. Ameri can
Journal of Physiology. Regulatory, Integrative &
Comparative Physiology 299, R343–R351.
204. Videan, E.N., Fritz, J. & Murphy, J. (2008). Effects
of aging on hematology and serum clinical chem-
istry in chimpanzees (Pan troglodytes). American
Journal of Primatology 70, 327–338.
205. Lammey, M., Baskin, G., Gigliotti, A., Lee, D.R.,
Ely, J. & Sleeper, M. (2008). Interstitial myocardial
fibrosis in a captive chimpanzee (Pan troglodytes)
population. Comparative Medicine 58, 389–394.
206. Rasmussen, S., Miller, M.M., Filipski, S.B. & Tol -
wani, R.J. (2011). Cage change influences serum
corticosterone and anxiety-like behaviors in the
mouse. Journal of the American Association for
Laboratory Animal Science 50(4), 479–483.
207. Wei, L., Simen, A., Mane, S. & Kaffman, A. (2012).
Early life stress inhibits expression of a novel
innate immune pathway in the developing hip-
pocampus. Neuropsychopharmacology 37, 567–580.
208. Jackowski, A., Perera, T.D., Abdallah, C.G., Garrido,
G., Tang, C.Y., Martinez, J., Mathew, S.J., Gorman,
J.M., Rosenblum, L.A., Smith, E.L., Dwork, A.J.,
Shungu, D.C., Kaffman, A., Gelernter, J., Coplan,
J.D. & Kaufman, J. (2011). Early-life stress, corpus
callosum development, hippocampal volumetrics,
and anxious behavior in male nonhuman primates.
Psychiatry Research 192, 37–44.
209. Mehta, M. & Schmauss, C. (2011). Strain-specific
cognitive deficits in adult mice exposed to early life
stress. Behavioral Neuroscience 125, 29–36.
210. Labonte, B. & Turecki, G. (2010). The epigenetics of
suicide: Explaining the biological effects of early
life environmental adversity. Archives of Suicide
Research 14, 291–310.
211. Schroeder, M., Krebs, M.O., Bleich, S. & Frieling,
H. (2010). Epigenetics and depression: Current
challenges and new therapeutic options. Current
Opinion in Psychiatry 23, 588–592.
212. Miller, G., Chen, E. & Chen, P. (2011). Psych -
ological stress in childhood and susceptibility to the
chronic diseases of aging: Moving toward a model
of behavioral and biological mechanisms. Psychol -
ogical Bulletin 137, 959–997.
213. Raber, J. (1998). Detrimental effects of chronic
hypothalamic–pituitary–adrenal axis activation.
From obesity to memory deficits. Molecular
Neurobiology 18, 1–22.
Comment 305