National Academies Press: OpenBook

Recognition and Alleviation of Distress in Laboratory Animals (2008)

Chapter: 3 Recognition and Assessment of Stress and Distress

« Previous: 2 Stress and Distress: Definitions
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 25
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 26
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 27
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 28
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 29
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 30
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 31
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 32
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 33
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 34
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 35
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 36
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 37
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 38
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 39
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 40
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 41
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 42
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 43
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 44
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 45
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 46
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 47
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 48
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 49
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 50
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 51
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 52
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 53
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 54
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 55
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 56
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 57
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 58
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 59
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 60
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 61
Suggested Citation:"3 Recognition and Assessment of Stress and Distress." National Research Council. 2008. Recognition and Alleviation of Distress in Laboratory Animals. Washington, DC: The National Academies Press. doi: 10.17226/11931.
×
Page 62

Below is the uncorrected machine-read text of this chapter, intended to provide our own search engines and external engines with highly rich, chapter-representative searchable text of each book. Because it is UNCORRECTED material, please consider the following text as a useful but insufficient proxy for the authoritative book pages.

3 Recognition and Assessment of Stress and Distress Introduction Recognition of distress in laboratory animals requires knowledge of what is normal for the species and strain used. Genetically modified animals should be evaluated in reference to the normality of their genotype. Most vertebrate species routinely experience some type of distress either in natural settings (e.g., during a predator attack) or as part of ­normal development (e.g., following natural maternal separation in rhesus ­monkeys; B ­ erman et al. 1994). The recognition of distress in laboratory animals, how- ever, requires an understanding of what is “normal” for the species being studied. In this chapter we consider the use of behavioral and physiologic variables to recognize stress and distress. Lab animals should behave according to species-specific normal behav- ioral, morphologic, and physiologic values (see Novak and Suomi 1988 and Snowdon and Savage 1989 for a discussion of psychological well-being in captive nonhuman primates). Species-specific normative ranges have been established for many parameters (e.g., hematocrit, blood glucose, body tem- perature, heart rate, blood pressure, respiration rate). Standardized growth curves and weight ranges can be obtained from laboratory animal suppliers for most species. Recognition of stress and distress in laboratory animals requires an understanding of the species-, gender-, and age-specific norms, because the normal range of some of these variables may vary as a function of gender, age, physiological state, or genetic characteristics. Values outside 25

26 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS n ­ ormalcy, therefore, may or may not serve as clinical indicators of a disease state. Various transgenic and knockout mice that exhibit severe behavioral and physiological phenotypes appear abnormal relative to their control littermates, but are normal for their genotype. For example, it is appropri- ate to evaluate Huntington’s disease transgenic mice for signs of stress and distress only relative to their own “normal” behavior, taking into account their particular genetic makeup, their abnormal motor patterns, and reduced weight gain (Mangiarini et al. 1996). Behavioral recognition of stress and distress Normal Behavior Many parameters have an effect on species-specific normal behavior and should be taken into consideration when behavioral characteristics are used to determine normalcy or the presence of stress and distress. Animals exhibit a variety of behavioral changes as part of the normal aging process. Males and females differ in the baseline values of many stress markers. Inbred murine strains differ in almost every behavioral, sensory, motor, and physiological trait studied and each inbred strain may respond to stress differently. Similar behavioral differences in response to stress have been observed in primates. Genetically engineered phenotypes need to be considered when assessing stress and distress in transgenic and knock- out animals. The maternal environment and rearing experiences of the offspring affect their future responses to stress and distress. Special physi- ological states, such as impending parturition, are defined by state-specific behaviors. Housing conditions may also modify species-specific behavioral patterns. Behavioral normalcy is further characterized by the absence of bizarre or atypical patterns of species-specific behavior. The presence of stereotypies usually implies suboptimal environments and ­ possibly poor animal welfare. The identification of species-typical behavior often comes from etho- grams developed by researchers to describe the kinds of behavior that animals display in various settings (Bronson 1979; for more references see Additional References). While the use of species-typical behavior as a normative benchmark has considerable value (Latham and Mason 2004), it does have limitations. First, the full range of species-specific behaviors cannot be recreated (or allowed to be expressed) in the laboratory animal care facilities as some types of behavior observed in natural settings (e.g., severe aggression) are clearly undesirable from a laboratory management perspective. Second, species-typical behaviors are neither invariant nor universal, as both the frequency and the presence of such behaviors vary

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 27 as a function of age, gender, physiological state, and genetic constitution. Third, rearing practices and housing environments may affect expected typical behavior. 1. Age: Many young mammals engage in high levels of social play whereas adults rarely do. Thus, play may be normal for young animals but not necessarily so for adults (Ruppenthal et al. 1974; Vanderschuren et al. 1997). All animals display physiological, behavioral, and cognitive changes as they age. Some of these changes, for example changes in coat/ hair appearance and locomotor ability, are overt and easily recognizable. Many laboratory animals display an age-related decline in exploratory activity, which is sometimes correlated to weight gain (as observed, for example, in mice of various strains; Ingram 2000; Ingram et al. 1981). In addition, a number of neurosensory, cardiovascular, endocrine, gastro- intestinal, musculo­skeletal, and reproductive changes occur with aging, some of which cannot be directly observed in the living animal. Such age- related changes (e.g., hearing and vision deficits) have been documented for a number of murine strains (Hawkins et al. 1985; for more references see Additional References), while cognitive deficits have been reported in aging mice and rats. It is postulated that some of these changes may be gender- and strain-related (Decker et al. 1988; Fischer et al. 1992; Frick et al. 2000). Changes in pain sensitivity and in emotional behavior that may have direct implications for stress and distress have also been reported in aging animals (Berry et al. 2007; Lamberty and Gower 1992). 2. Gender: Female mammals generally care for infants, whereas the extent of male involvement varies across species. Thus, species-­typical behavior may be gender-biased. Moreover, gender-related differences in stress markers can be profound and occur in all vertebrate species. For example, female rats and mice exhibit marked elevations in basal and stress-induced corticosterone release relative to males, although these are buffered by high levels of corticosteroid-binding globulin, thus making free corticosterone levels similar in both sexes (McCormick et al. 1995). Absolute corticosteroid levels in females fluctuate in relation to the stage of estrus, presumably affected by circulating levels of estrogens (Figueiredo et al. 2002). Thus, assessment of HPA activity as a measure of stress (see below) needs to account for the gender of the animal and the type of steroid measurement (i.e., total [plasma] or free [saliva]). Males and females also appear to differ in other aspects of their stress response(s); for example, while females have greater anhedonic and HPA axis responses to chronic mild stress than males, they score lower on tests of behavioral depression caused by chronic stress exposure (Dalla et al. 2005).

28 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS 3. Genetic traits: Genetic variability among many animal species complicates our understanding of the effects of stress and distress in labora- tory animals. Multiple studies in the mouse have shown that generalizations even across a single species can be problematic. Selective breeding has produced hundreds of inbred mouse strains, providing extensive genetic and phenotypic variability (Beck et al. 2000; Silver 1995). A mouse strain is classified as inbred after 20 inbreedings (that is, 20 generations of brother x sister or offspring x parent matings), at which point its members are virtu- ally genetically identical because at the 20th or subsequent generations all animals are traceable to a single breeding pair. One cannot assume that mice from different inbred strains are alike (or even similar), perform identi- cally, or experience and react uniformly to stimuli—stressful or otherwise. In fact, inbred strains of mice differ in almost every behavioral, sensory, motor, and physiological trait studied to date, such as anxiety, learning and memory, brain structure and size, visual acuity, acoustic startle, exploratory behavior, alcohol sensitivity, depression, pain sensitivity, and motor coordi- nation (Crawley et al. 1997; for more references see Additional References). What is typical for one strain—for example, high levels of play behavior or social interaction (Moy et al. 2004) or novelty seeking and exploratory behavior (Bolivar et al. 2000; Kliethermes and Crabbe 2006)—may not be characteristic of another. For these reasons different inbred murine strains respond to stress dif- ferently and thus may well experience distress in different ways. Indeed, a number of behavioral studies provide evidence that strain differences in distress susceptibility are likely. For instance, inbred strains differ in performance on anxiety, depression, and fear learning assays (Balogh and Wehner 2003; for more references see Additional References). Correlating behavioral performance across such matrices can provide some indication of basic genetic differences among strains in response to stressful situations (Ducottet and Belzung 2005). When exposed to a month of unpredictable mild stress (e.g., cage tilting, damp bedding, lights on for a short period dur- ing the dark phase) most strains groom themselves less resulting in poor fur condition, while only a few display heightened aggression levels (Mineur et al. 2003). In general, inbred strain differences appear in the stress-induced hyperthermia model (Bouwknecht and Paylor 2002; van Bogaert et al. 2006) and in stress-invoked autonomic responses (body temperature and heart rate), although the latter are also a function of the intensity of the insult applied (van Bogaert et al. 2006). Behavioral differences have also been observed in primates. High reactor monkeys are much less likely   t is now well established that there are marked individual differences in reactivity among I nonhuman primates when animals are exposed to novel situations or to relatively minor changes in their social or physical environment. Some rhesus monkeys (~20%) respond to

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 29 to explore a novel stimulus than low reactors (Suomi 2004). Moreover, because even within genetically diverse species individual animals will vary on many dimensions, high levels of exploration may be the norm for some but not for others. 4. Transgenic and knockout mouse models: Many genetic mouse models have intentional or incidental behavioral and/or physiological p ­ henotypes relevant to stress. Disturbances in genes associated with brain stress-regulatory systems can elicit stress hyposensitivity (e.g., deletion of the corticotrophin-releasing hormone [CRH] gene; Muglia et al. 1995) or stress hypersensitivity (e.g., overexpression of CRH; Stenzel-Poore et al. 1994). Moreover, there are any number of transgenic/knockout phenotypes that affect behavioral or physiological indices of stress without producing overt stress or distress. For example, deletion of the S6 kinase gene produces a remarkably small animal, not because of the animal’s “failure to thrive” but rather because absence of this powerful cell-size regulator results in a smaller size of otherwise healthy cells (Thomas 2002). Thus, expressed phenotypes need to be considered when assessing stress and/or distress in genetically engineered animal models because their presence may be even more difficult to recognize and diagnose in these animals than in their con- trol littermates. 5. Rearing and postnatal separation: In most mammals, the early environment of the young animal is defined by the presence of its mother; therefore maternal characteristics can have a profound impact on the future behavior of adult offspring. There is ample scientific evidence that maternal environment can be an important epigenetic determinant of physiology and behavior, and should be considered as a variable for assessment of stress and distress. Offspring are generally reared with their mothers and may also be reared in larger social groups that include other offspring as well as adult males and females. Some species- or strain-typical behaviors, such as cross fostering, in which the offspring of one species are reared by the parents of another species or of the same species but a different strain, are more susceptible to parent-related environmental manipulations. The extent to which cross fostering may produce distress in the offspring relatively mild environmental stressors with unusual behavioral disruption and physiological arousal including prolonged activation of the hypothalamic-pituitary-adrenal (HPA) axis, as assessed by plasma cortisol and adrenocorticotropic hormone (ACTH), increased cerebrospinal fluid levels of the norepinephrine metabolite 3-methoxy-4-hydroxyphenylglycol, heightened sympathetic nervous system activity as reflected in altered heart rate rhythms, and abnormal immune system response (Coe et al. 1989; Higley et al. 1991). The same stressors elicit only minor behavioral reactions and transient physiological responses in the remainder of the population (Suomi 2004).

30 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS depends on the degree to which parental care varies across the two species (or strains) in question. In birds, cross fostering can be relatively benign (e.g., rearing of green finches’ offspring by canaries; Guettinger 1979). In other cases, however, cross fostering may complicate the assessment of stress and distress, as cross-fostered rats, mice, and goats frequently exhibit behaviors more similar to the adoptive mother strains (Ahmadiyeh et al. 2004; Anisman et al. 1998; Kendrick et al. 2001). In rats, female offspring of dams bred for high licking and grooming that have been reared with their biological ­mothers will themselves provide extensive maternal care of their own pups. In contrast, if female offspring of high licking and groom- ing dams are instead cross fostered with low licking and grooming (i.e., “poor”) mothers, they will subsequently provide little maternal care to their own offspring, resulting in behavioral and physiological changes that per- sist into adulthood (Francis et al. 1999). Recent research into the effects of maternal behavior on such developmental traits as DNA methylation, an epigenetic mechanism that alters gene expression, has shown that maternal environmental programming (for example, high or low grooming) affects the glucocorticoid receptor gene and possibly the responses of the offspring to stress. Offspring of high grooming mothers (or those cross fostered to them) appeared less responsive to stressful stimuli and had increased expression of these ­ receptors in the hippocampus compared to those raised by low grooming dams (Fish et al. 2004; Weaver et al. 2004). Microarray analysis has shown that more than 900 genes of the hippocampal transcriptome are stably regulated by maternal care (Weaver et al. 2006). In species such as primates, however, infants may be nursery-reared because of the infant’s illness, the mother’s failure to care for the infant, or demands of the experimental protocol. The two most common nursery rear- ing procedures for macaques are peer rearing (i.e., rearing infants together 24 hours a day) and surrogate peer rearing (i.e., rearing infants on inanimate surrogate mothers 24 hours a day with 1-2 hours of daily peer exposure in a playroom setting). Both conditions commence shortly after an animal’s birth before a strong attachment has been formed to the mother, and thus infants show little in the way of separation anxiety. From a developmental perspective, peer rearing appears to confer the greater risk for distress and social maladjustment. Peer-reared monkeys typically show higher levels of mutual clinging and greater fear responses than surrogate-peer-reared monkeys early in life and have difficulty adapt- ing to larger social groups as juveniles (Ruppenthal et al. 1991). Peer rearing has also been associated with impaired immune responses (Coe et al. 1989; Lubach et al. 1995) and, when combined with repeated separa- tions, appears to promote heightened aggressiveness, impaired impulse control, alcohol abuse, and low levels of 5-hydroxyindoleacetic acid (a serotonin metabolite) in cerebrospinal fluid (Ichise et al. 2006). Although

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 31 less ­ studied, surrogate-peer-reared monkeys appear to behave more like normally reared monkeys. Indeed, a large comparison study of surrogate- peer-reared ­monkeys (n=506) to normally reared monkeys (n=1,187) failed to detect any differences in growth, health, survival, reproduction, and maternal abilities between the two groups (Sackett et al. 2002). But because some individuals reared in either condition may be more vulnerable to the development of abnormal behavior than normally reared monkeys, careful observation and ongoing assessments would help guide colony manage- ment decisions regarding group composition and enrichment strategies. A different kind of early rearing experience involves separation from the mother or other attachment figure (e.g., other peers) after a strong attach- ment has been formed. Such separations may occur both for research pur- poses or to facilitate weaning. Depending on such variables as the timing of the separation, the nature of the separation environment, and the primate species, separation can induce high levels of stress in infants expressed by vocalizations and heightened activity (Bayart et al. 1990; Jordan et al. 1985; Laudenslager et al. 1990; Levine et al. 1993). It can also alter HPA activity (Levine 2005; Levine and Mody 2003; Parker et al. 2006; Vogt et al. 1980) and immune responses (Laudenslager et al. 1982). Reactions are often stronger when the infant is separated both from its mother and the environment in which it was raised compared to when only the mother is removed from the infant, but this effect can vary by species (Laudenslager et al. 1990). These signs generally disappear when infants are reunited with their mothers or their attachment figures, although neuroendocrine responses may be altered. 6. Physiological state: Many species (such as dogs, sows, rabbits, and mice) need to build nests just before parturition, whereas others do not engage in such behavior (Arey 1997; Broida and Svare 1982; Crawley 2000; Kunkele 1992). 7. Housing: Environmental conditions can modify species-specific behavioral patterns. Adults housed in same sex groups cannot show some aspects of the species-typical physiological repertoire (e.g., mating or p ­ arental behavior). Housing conditions (such as cage types and environ- mental enrichment) affect the amount of time that mice spend engaging in distinct behavioral patterns, as reported by Olsson and Sherwin who, using videorecording, showed that mice in furnished cages (i.e., cages with nest- ing material, running wheels, nest box, and chew box) “spent less time rest- ing, bar-chewing and bar-circling and more time on exploratory/locomotor behaviors” (Olsson and Sherwin 2006, p. 392). Lack of environmental stimulation or social deprivation adversely impacts normal brain function in rats, such as attenuation of the prepulse inhibition (PPI) behavior ­elicited by

32 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS startling events, and is accompanied by underlying neuro­chemical changes such as enhanced dopamine activity (Würbel 2001). Based on significantly fewer instances of abnormal behaviors (i.e., stereotypies) encountered in wild-caught animals vs. their captive-bred controls, the argument for a neuro­ protective effect of early environmental enrichment against future abnormal behaviors has been made (Lewis et al. 2006). Moreover, studies have shown that dendritic anatomy in young rats was altered in response to a brief 4-day exposure to a complex environment (Wallace et al. 1992) and so was the hippocampus of adult mice in comparison to controls ­(Kempermann et al. 2002; for more discussion on enrichment see Chapter 4). Behavioral normalcy is further characterized by the absence of bizarre or atypical patterns of species-specific behavior. Examples of abnormal behavior include excessive barbering observed in mice (Garner et al. 2004; Morton 2002), regurgitation/rumination and coprophagy seen in apes (Nash et al. 1999), or more serious self-injurious behaviors exhibited by rhesus monkeys (Novak 2003). Sometimes, such behaviors represent normal social patterns. For example, coprophagy associated with mother rearing occurs not only in laboratory-housed apes (Nash et al. 1999) but also in the wild where it is postulated to contribute to the reclaiming of unused resources from the feces (Krief et al. 2004). In other cases, however, such patterns are a sign of well-defined diseases or disorders, as, for example, excessive tremors observed in transgenic mice with Huntington’s disease (Mangiarini et al. 1996). In yet other instances, abnormal behavioral patterns, such as stereotypies, may result from suboptimal housing environments (Bayne et al. 1992, 2002; Hubrecht et al. 1992; Mason 1991). Stereotypic behavior is characterized by highly repetitive and ritual- istic actions, the function of which is largely unknown. Environments that elicit or enhance stereotypies not as part of defined pathophysiology or disease models are typically suboptimal (Berkson and Mason 1964; for more ­references see Additional References). Stereotypies vary across ­species and appear at different times of day and under different conditions (Mason and Mendl 1997). Classic whole-body stereotypies include circling, pacing (dogs, primates), wall bouncing (dogs), and somersaulting and bar chewing (rodents), whereas self- or other-animal-directed stereotypies often involve the limbs or face and include such patterns as digit sucking, paw lick- ing, and overgrooming (Bayne and Novak 1998; for more references see A ­ dditional References). Although there is yet inadequate research on the relationship between distress and stereotypy, a recent meta-analysis of studies linking stereotypy to animal welfare suggests that some stereotypies may function to regulate arousal and possibly reduce distress as “do-it-yourself enrichment” strate- gies to alleviate the effect of a suboptimal environment (see Mason and

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 33 Latham 2004). Overall, however, the presence of stereotypies should be a cause for concern because animals that exhibit such behavioral patterns may not only have experienced some stress or distress in the past but also live in environments that promote or sustain these abnormal behaviors. Moreover, as a study by Krohn and colleagues has shown, stereotypies are probably underreported as they may occur during the night when staff are not present, or cease when staff enter a room (��������������������������������� Krohn et al. 1999). ������������� If, in fact, the presence of stereotypies is being investigated, then more sophisticated methods such as closed-circuit television or videorecording, or simpler diagnostics such as partially reversed light cycles, would enable staff to observe nocturnal animals during their most active periods in order to docu- ment instances of abnormal behavior (Hubrecht 1997). Abnormal Behavior and Clinical Signs Recognition of distress should be derived from intimate knowledge of the species’ or strain’s normal behavior and may be based on (1) clinical signs and/or (2) significant deviation from the expected behavioral repertoire. Some clinical signs (e.g., changes in temperature, respiration, feeding behavior) indicate an abrupt onset of distress while others (e.g., weight loss) develop over a longer period of time and may serve as warnings. A thorough clinical examination with references to baseline effects of age, gender, genotype, etc., is necessary to establish the presence of distress, while an abrupt and marked change in behavior lasting more than a few days may also indicate a disease state. While the presence of stereotypies is undesirable, the relationship between stereotypic behavior and distress remains largely unknown. Preventing the development of ­ stereotyped behavior by providing species-specific appropriate environments is likely to result in improved welfare. Assuming that an animal’s behavior has been well characterized, indi- cations of distress may include certain clinical signs or marked change from the individual animal’s usual behavioral repertoire (Morton and Griffiths 1985; see score sheet examples in Appendix). An abrupt and marked change in behavior lasting more than a few days may also indicate the presence of a disease state in addition to distress, particularly if these changes occur in conjunction with severe reductions in normal daily activities such as feed- ing behavior, sexual behavior, maternal behavior, or attention to threat. Conversely, animals may exhibit increased activity associated with unusual motions (e.g., head rubbing) or unusually high levels of certain behaviors (e.g., scratching). Even marked changes in behavior, however, must be evaluated in context. For example, females usually exhibit decreased activ- ity the first day following parturition, an expected behavior.

34 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Clinical Signs of Distress Clinical examination to establish the presence of distress should focus on, but not be limited to, the following: signs of abnormal respira- tion ­ (shallow, labored, or rapid); assessment of grooming and hair coat ( ­piloerected or greasy, possibly reflecting reduced grooming); examination of the eyes (runny, glassy, or unfocused); examination of motor postures (hunching or cowering in the corner of the cage, lying on one’s side, lack of movement with loss of muscle tone); absence of alertness or quiescence (inattention to ongoing stimuli); changes in body weight; the ability or failure to produce urine or feces; unusual features of urine (volume, smell, and color) or feces (quantity, consistency, and color); the presence of vomit; the status of the animal’s appetite and water intake; and intense or frequent vocalizations (Bennett et al. 1998; Fortman et al. 2002; Fox et al. 2002). It is appropriate to evaluate some of these signs in context, as, for example, rapid breathing could result from vigorous activities such as playing or r ­ unning on the wheel, lying down may occur as part of social grooming (e.g., among macaques), weight loss is often associated with advanced age, and some mammals raise their hair (piloerection) while eating. In addition, clinical evaluation and diagnosis should consider species, age, gender, physiological state, and genetic variables (Bennett et al. 1998). While some of the clinical signs described above (e.g., respiratory changes, changes in fecal material and/or in urine) are more relevant to the acute onset of a distressful state, other measures may serve as poten- tial early warning signs of distress (e.g., rapid body weight changes in the absence of dietary modifications). Significant and unexpected changes in weight in either direction may be indicators of altered endocrinological, immunological, or neurological parameters. Indeed, the relatively sudden loss of 25% body weight of a nonhuman primate is one of the parameters used to determine humane endpoints in primate research (Association of Primate Veterinarians 2008). This view should not be applied to caloric restriction research protocols where animals may be subject to controlled diets that reduce their weight by as much as 15-20% (Heiderstadt et al. 2000). Such protocols are widely used in gerontology research where diet has been shown to slow aging, extend lifespan, and reduce the incidence of age-related diseases in rodents (Goto et al. 2002; for more references see Additional References), while beneficial effects have also been observed in nonhuman primates (Ingram et al. 2007). Moreover, sensory-motor function and learning studies may use caloric or water restriction as a motivational tool (Heiderstadt et al. 2000; Smith and Metz 2005). In these studies regular monitoring of body weight is essential to ensure that animals either do not fall below an accepted weight

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 35 range or, in the case of young animals, gain the appropriate body weight for their age. Behavioral Signs of Distress It has been suggested that abnormal behavior, such as stereotypies, is a marker for distress (Dawkins 1990). It remains unclear at this time whether any or all abnormal behaviors qualify as indicators of distress. Several alternative (and largely speculative) hypotheses attempt to explain the occurrence of stereotypic behavior in animals (Mason and Latham 2004; Tiefenbacher et al. 2005). Among these, the stimulation hypothesis suggests that when sensory motor input is low, possibly due to existing (i.e., nonstimulating, poor) housing arrangements, animals engage in stereotypic behavior to self-provide increased sensory-motor input (Sherwin 1998). For example, when cage size constrains normal movements, some animals may respond by developing stereotyped pacing in order to satisfy their need for activity (Draper and Bernstein 1963). The habit hypothesis suggests that although stereotypic behavior may have originally arisen in response to stress or distress, it persists as a habit uncoupled from the situation that originally produced it (Dantzer 1986; Mason 1991). Those who favor the arousal reduction hypothesis suggest that stereotypic behavior may serve to calm the animal and thereby avoid distress (reviewed in Mason 1991). Research shows that in some humans and nonhuman primates, even more serious forms of abnormal and self-injurious behavior may function to reduce arousal (Tiefenbacher et al. 2005). The arousal reduction hypothesis is consistent with the view that while an underlying stress or distress state may have initially caused abnormal behavior, eliminating the behavior may be neither desirable nor possible because the stereotypy may sometimes prevent the onset of distress. Preventing the development of stereotyped behavior by providing the animals with species-specific appropriate environments is obviously desirable and likely to result in improved welfare, especially as enrich- ment “therapy” may reduce but will not cure the abnormal behavior (van Praag et al. 2000; Wolfer et al. 2004). Although recent studies suggest that s ­ tereotypical animals may experience psychological distress due to a puta- tive common mechanism between stereotypy, schizophrenia, and autism, the relationship between stereotypic behavior and distress remains largely unknown and is in need of further study (Garner 2006; Garner and Mason 2002; Garner et al. 2003; Mason 2006).

36 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Behavioral Signs of Stress As mentioned in Chapter 2, stress is ubiquitous, it can occur in both pleasurable and aversive situations, and its physiological parameters are well established. Our knowledge of the behavioral correlates of stress, however, is considerably smaller. The behavioral changes observed in a stressed animal (as opposed to a distressed one) may be more subtle and variable, depending on the environmental conditions in which the behavior is being evaluated. In addition to recognizing an animal’s normal patterns of behavior, the observer must be well trained and knowledgeable about the normal species-specific behavior in the context of species, strain, gender, and physiological state. Types of behavior commonly explored to inves- tigate the presence of stress include open-field activity, movements in an elevated plus maze, changes in innate behaviors (e.g., movement, groom- ing, feeding, sexual behavior), defensive behaviors (to external threats), and avoidance/escape (Beck and Luine 2002; for more references see Additional References). Physiologic measures of stress and distress Endocrinological Parameters One of the primary endocrinological systems involved in the stress response is the hypothalamic-pituitary-adrenal (HPA) axis, which reacts to stress by releasing glucocorticoids. Glucocorticoid levels can be used as indicators for the impact and strength of a stressor, with two caveats: (1) they ­cannot inform as to the type of stressor (positive or aversive) that stimulates the HPA and (2) most sampling procedures are themselves stressful to the animals, thereby confounding the measurements. Therefore, the assess- ment of distress based on glucocorticoid levels has limitations, especially under the unproven assumption that a certain glucocorticoid concentra- tion indicates the presence of distress. Furthermore, stress or distress may exist without the concomitant activation of the HPA axis. Glucocorticosteroids The hypothalamic-pituitary-adrenal (HPA) axis, often referred to as the “stress response system”, plays an important role in an organism’s reaction to stressors. In response to a stressful situation the hypothalamic p ­ araventricular nucleus synthesizes corticotrophin-releasing hormone (CRH), which is released into the median eminence and travels to the ante- rior pituitary where it causes the release of adrenocorticotropic hormone (ACTH) into the circulatory system. ACTH then acts selectively on specific

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 37 receptors in the adrenal cortex, resulting in the release of glucocorticoids (cortisol or corticosterone), which mobilize energy stores in response to the perceived stress. When the stressor is removed or otherwise adapted to, glucocorticoids bound to receptors in the hypothalamus and pituitary initi- ate negative feedback that causes a decrease in the production and release of CRH and ACTH, thus terminating the hormonal response and completing the negative feedback loop (Meaney et al. 1996; Miller and O’Callaghan 2002). It should be noted that the HPA axis affects many hormonal and neural systems and plays a role in modulating the immune system. Both positive and negative stimuli activate the HPA axis with short- and long-lasting effects. For example, exposure to novel stimuli may elicit exploratory behavior and brief activation of the HPA axis. In contrast, pro- longed or repeated stressors, such as social separation of young from their mother, generally elicit strong protest reactions and activation of the HPA axis (Levine 2005; Levine and Mody 2003; Vogt et al. 1980). In the first instance, homeostasis is quickly restored, whereas in the latter case animals may be subjected to chronic HPA changes associated with neuroendocrine stress resistance that persists even after animals are returned to their mother (Parker et al. 2006). Glucocorticoid levels (usually corticosterone in rodents, cortisol in other species) are used as indicators of the strength and impact of a stressor. Meaningful interpretation of these values, however, presents significant challenges. Glucocorticoids are typically measured in blood serum or plasma but can also be quantified in saliva, urine, feces, and hair (Abelson et al. 2005; for more references see Additional References). Blood sampling requires venipuncture and possibly other stressful pro- cedures such as handling, transport, capture, restraint, needle stick(s), and sedation. Unless animals are habituated to blood sampling, the method itself can activate the HPA axis thereby confounding assay results. A less stressful sampling method involves measuring glucocorticoid levels in hair. Hair samples are obtained by shaving hair from a particular region ­(usually the nape of the neck) and then shaving the hair once again after a defined period of regrowth (for a discussion of the method in primates see ­Davenport et al. 2006). Although this procedure requires the animals’ habituation, restraint, or sedation, unlike venipuncture the stress caused by hair collec- tion does not confound the measurement. Similarly, saliva collection may impact animals less if they have been habituated to the process (Lutz et al. 2000). Urine and feces are collected after excretion from the body and so probably have the least impact, unless animals are not habituated to the special metabolic cages used for sample collection. The type of sample obtained and the time frame it reflects may also influence results. Blood and saliva yield an index of stress at one brief moment in time (point samples) and are, therefore, influenced by circadian

38 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS variation (Windle et al. 1998b), making it crucial that samples be collected at the same time each day (e.g., at the nadir of the rhythm). In contrast, urine and feces yield an index of stress reactivity over hours or possibly several days (steady-state samples) and are, therefore, less vulnerable to circadian variation. To date, only hair can provide a chronic index of stress cover- ing a period of several months or more. Assessment of cortisol in hair is presumably unaffected by circadian variation and can be obtained at any time of day. The above information is relevant for understanding and interpreting what might be revealed about stress and distress by examining the activa- tion of the HPA axis. The two most likely ways to assess distress are to (1) examine differences in basal glucocorticoid levels and identify animals outside a “normal range” or (2) obtain glucocorticoid levels before and after the imposition of a stressor. The first approach is problematic because it assumes that a certain concentration of glucocorticoids indicates dis- tress, although there is no scientific evidence to support this assumption. Moreover, as many sampling methods may themselves activate the stress response, there are no standardized ranges for basal glucocorticoid concen- trations. The second approach is also problematic for two reasons: first, the putative relationship between the magnitude of change in glucocorticoid concentrations and distress has not been established; and second, both positive and aversive stimuli activate the HPA axis. Finally, the develop- ment of stress or distress is not necessarily associated with activation of the HPA axis, as hormonal changes are not necessarily present under all clearly stressful conditions. For example, animals that experience chronic neuropathic pain do not exhibit changes in circadian corticosteroid levels or oscillations in HPA responsivity to restraint, despite the presence of neuropathic pain markers (mechanical allodynia and hyperalgesia) and activation of central pain and stress circuits in the amygdala (Bomholt et al. 2005; Ulrich-Lai et al. 2006). Other “Stress” Hormones As is the case with the hormones of the HPA axis, stressors alter the secretion of other endocrine factors (e.g., prolactin, growth hormone, lutein- izing hormone, α-melanocyte stimulating hormone [α-MSH], and ­oxytocin). Serum levels of these hormones can be effectively used to ­ monitor the temporal dynamics of stress responses. While some (prolactin, α-MSH, o ­ xytocin) increase during stress, others decrease (growth hormone, lutein- izing hormone, prolactin), depending on the animal species and the physi­ological state in which stress occurs (Armario et al. 1984; for more references see Additional References). Due to the fact that these hormones are also released in response to other stimuli (e.g., suckling of young, ultra-

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 39 dian or circadian variations), it is necessary to take into consideration and control for their normal patterns of secretion in order to accurately interpret their concentration levels. Moreover, their usefulness is subjected to the same limitations as discussed above, although chronic indwelling vascular c ­ atheters and automated blood collection systems may circumvent this limitation to some degree (Abelson et al. 2005; for more references see Additional References). Neurological Parameters Stressors activate the autonomic nervous system, specific brain areas, and various neurotransmitters, yet a cause and effect relationship has not yet been firmly established. Candidates for the role of a master integrator include the region of the amydgala and the neuropeptide corticotrophin- releasing factor. The Autonomic Nervous System Many different types of stressors cause the rapid activation of the sympa- thetic division of the autonomic nervous system (ANS) (Blanc et al. 1991; for more references see Additional References). This ­activation leads to increased cardiac output via increased heart rate and stroke ­volume; redistribution of blood flow from splanchnic, renal, and cutaneous vascular beds to active muscle; increased mobilization of nutrients; and increased heat production. Some stressors may also increase the activity of the para­sympathetic divi- sion, affecting both core body temperature and the gastrointestinal system (e.g., disturbed intestinal absorption, gastric ulceration, colitis; Johnson et al. 2002; for more references see Additional References). Direct monitoring of autonomic activity to assess the presence of dis- tress in conscious animals is technically challenging, while indirect mea- sures are somewhat easier to acquire (Li et al. 1997; Randall et al. 1994; Zhang and Thoren 1998). For example, telemetry in conscious, unrestrained animals is an effective method for the continuous monitoring of physiologic alterations in heart rate, respiration, blood pressure, ECG, and body tem- perature (Akutsu et al. 2002; for more references see Additional References). Once again, however, changes in these parameters do not necessarily indicate stress as they may result from nonstressful stimuli (e.g., circadian variations). Neurotransmitters Considerable effort has been directed at exploring the neurotransmitter systems and brain areas activated in response to stress as different insults

40 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS activate separate but specific patterns of transmitters, modulators, and brain locations. An active area of investigation has been the identification of a basic core neural circuit that is activated by all stressors. A common approach has been to divide stressors into categories, hypothesizing that each category would activate a particular set of neural structures. The stressors are classified as either “processive” (i.e., stressors that require inter- pretation by higher brain structures) or “systemic” (i.e., stressors evoked by an immediate physiological threat; Herman and Cullinan 1997). However, the neural response has been heterogeneous for both these two as well as more narrowly defined categories. For example, Serrats and Sawchenko administered either lipopolysaccharide (LPS) or staphylococcal enterotoxin B (SEB) to rats in order to study brain activation patterns using c-fos induc- tion as a measure of neural activity. Although both LPS and SEB activated some of the same brain structures, they produced distinctly different patterns of neural activation (Serrats and Sawchenko 2006). Research indicates that there are few, if any, transmitters and ­modulators that are not activated in some region of the brain by some stressor. Mono- aminergic circuits, such as noradrenergic neurons, projecting from the locus coeruleus (Aston-Jones et al. 1996), serotonergic neurons projecting from the dorsal raphe nucleus (Lowry 2002), and dopaminergic neurons projecting from the ventral tegmental area (Pezze and Feldon 2004) are par- ticularly stress-responsive. Research on neuropeptides has focused on CRH (Nemeroff and Vale 2005), endogenous opioids (Ribeiro et al. 2005), and neuropeptide Y (Heilig 2004). CRH may be the key central integrator of the stress response as CRH-containing neurons in the paraventricular nucleus of the hypothalamus are the primary common path in the neural regulation of both the HPA and autonomic responses to stressors. In addition, CRH is found in the central amygdaloid nucleus, an important node in regulating behavioral alterations in response to fear (see below for additional informa- tion on the fear response). A somewhat different experimental approach to elucidate the ­interaction between stressors, neurotransmitters, the brain, and behavioral patterns has been to identify the neural circuit that mediates a specific behavior. For example, the neural circuitry involved in fear conditioning is well known. When a neutral stimulus, such as a light or a tone, is followed closely by an aversive stimulus such as a foot shock, it elicits a fear response. The association between the neutral and aversive stimuli is formed in the basal and lateral nuclei of the amygdala via an N-methyl D-aspartate (NMDA)- d ­ ependent long-term potentiation (LTP)-like process. The information is subsequently transmitted via the central nucleus of the amygdala to the proximate mediators of the particular behavioral and physiological con- stituents of fear (Davis and Whalen 2001) as, for example, to hypothalamic nuclei that regulate respiration. However, many of the fear-modulating

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 41 factors alter the activity in structures that project to the top of the cen- tral nucleus of the amygdala (Sotres-Bayon et al. 2006), underscoring the a ­ mygdala as the key integrative site for fear. In contrast, much remains to be learned about the neural control of defense responses to threat. For example, depending on circumstances, external threats such as the presence of a predator may result in flight, freez- ing, or other defensive behaviors. While the involvement of the midbrain periaqueductal gray is well known (Keay and Bandler 2002), inactivation of the brain areas typically responding to a threat from a predator reduces the defense response(s) elicited by predator odor or exposure (Blanchard et al. 2005; Canteras 2002), which may differentially impact predator defense and shock stimuli responses. Immunological Parameters The relationship between stress, distress, and the immune system is very complex. Acute stress usually activates innate immune responses (i.e., nonspecific immunity), but it may either increase or inhibit adaptive immunity. On the other hand, chronic stressors suppress adaptive immune responses. Activation of various types of immunity-related cells may be used as an indicator of immune system-stress interaction. Signaling pathways link the brain with the immune system thereby allowing stress and distress to influence immune function. Immune system cells such as lymphocytes and macrophages express receptors for a variety of hormones and neurotransmitters, while the spleen and thymus are inner- vated by the autonomic nervous system (Felten et al. 1985; Sanders et al. 2001). The complex nature of these influences, however, does not permit simple generalizations such as “stress/distress suppresses immune function”. The immune responses elicited depend on the type, duration, and intensity of the stressor; the species, strain, age, and gender of the animal(s); and the aspect of immunity examined. Acute Stress/Distress and Immunity The principles that determine whether acute stressors inhibit or poten- tiate adaptive immunity are currently unknown. Nevertheless, adaptive immune responses that involve antigen recognition by T cells are invari- ably affected in acute stress. As Fleshner and colleagues have shown, rats stressed by inescapable tail shock failed to expand a subset of T cells and produced reduced quantities of IgM and IgG antibodies (Fleshner et al. 1995). In contrast, restraint at the time of immunization was shown to facili-

42 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS tate immunological memory due to elevated counts of memory and effector helper T cells (Dhabar and Viswanathan 2005). In contrast, the innate immune response is most often enhanced in response to acute stress, including during stress conditions identical to those that interfere with specific immunity (Deak et al. 1999; Fleshner et al. 1998). A variety of stressors have been reported to increase macrophage function and elevate levels of known pro-inflammatory mediators such as ­ interleukin-1 and tumor necrosis factor (O’Connor et al. 2003). Acute phase protein levels are similarly elevated as these cytokines also initiate the acute phase response. In addition, acute stressors potentiate or even directly elicit the sickness response, a set of behavioral and physiological changes (including fever, increased sleep, reduced social interaction and physical activity) that occur during infection (Dantzer 2004; Maier and Watkins 1998). Chronic Stress/Distress and Immunity Chronic or repeated stress has been shown to suppress adaptive immu- nity (Tournier et al. 2001), but not much is known of its effects on innate immunity. Studies looking at the effects of stress on disease outcome rather than on immune responses have shown that stress can either increase or decrease disease severity depending on conditions and variables measured. Disease progression can be either inhibited or facilitated depending on the precise occurrence of the insult, as the timing of stressor exposure relative to disease onset is often critical (Johnson et al. 2006). In addition to assays that measure T-cell proliferation, natural killer cell cytotoxicity, or B-cell activation and antibody production as indicators of adaptive immunity-stress interaction, one can also measure the capacity of polymorphonuclear cells to produce a respiratory burst in vitro. Research has shown that the functional capacity of leukocytes from stressed animals is suppressed, thus diminishing their “coping capacity” (as defined by the production of oxygen free radicals; McLaren et al. 2003). Assessment of Distress Clinical signs interpreted through relevant animal behavior and physiologi- cal states are the most reliable distress measures. Distress evaluation is crucial when research animals are purposefully exposed to stressful condi- tions or when animals appear distressed unexpectedly. The assessment and subsequent interventions should involve researchers, veterinarians, and technicians and the team should continue its collaboration to develop an intervention strategy once the assessment is completed.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 43 Assessment of distress varies in relation to the species, husbandry con- ditions, and experimental protocol employed as well as with each indi- vidual animal, and is most effectively achieved by the collection of multiple behavioral and physiological parameters and the use of a team approach that includes researchers, veterinarians, and animal caretakers/technicians. While the most reliable distress measures are the clinical signs previously described, identification and interpretation of these results depends on a solid foundation of knowledge of animal behavior and may likely require special training of relevant personnel. Distress evaluation becomes crucial in two contexts: (1) when the research protocol calls for the animals’ exposure to stressful situations known to produce distress; and (2) when any animal unexpectedly shows signs of distress. In the first case, the experimental protocol approved by the Institutional Animal Care and Use Committee (IACUC) generally includes procedures and decision algorithms for distress management. The appropri- ate intervention will be informed by the stressor’s duration and intensity as well as some of the animal’s individual characteristics (e.g., species, age, gender). In the second case, additional assessments and monitoring may be necessary. Once an animal has shown initial signs of distress, there should be immediate communication between the primary investigator, the veterinar- ian, and the animal care staff to determine whether the distress is related to the study (whether anticipated or unanticipated) or further investigation into its cause is required. The discussion should also include potential inter- ventions (see Chapter 4) and their effects on the objectives of the research project, as they may introduce unknown variables into the study. Options may include removal of the animal from the study population or euthanasia, depending on the severity and prognosis of the distress insult. It is essential to maintain a collaborative relationship and dialogue between those respon- sible for the care and welfare of the animal throughout the assessment. The next step is to identify the etiology or trigger of the distress episode by performing a thorough examination of the animal and its environment. The investigation should begin by obtaining information regarding the animal’s species, strain, age, gender, and reproductive status. An effective examination should account for species-related differences among natural behaviors, learning abilities, and levels of intelligence, in addition to the ways animals use their senses and communicate. Some species, strains, or breeds are predisposed to certain behavioral problems or have certain behavioral phenotypes, or an individual animal’s characteristics may affect both the development and alleviation of its distress. Physical examination and appropriate diagnostic tests for all distressed animals can help determine whether an underlying medical condition is the primary cause of distress. A review of the medical and investigator records

44 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS is an important part of the process, as background information and history may enable the veterinarian to determine whether preexisting medical conditions were resolved. An examination of colony records and interviews with animal care staff may help pinpoint possible environmental triggers. Other causes for consideration include husbandry and handling procedures, the behavior of other animals in the room, temperature variances, noises, vibrations, and odors, as well as any specific research-related (i.e., protocol specifications) or investigator-related (i.e., disturbance of housing routine) activities. Clinical signs should initially be examined in a relatively undisturbed animal in order to assess the animal’s natural unprovoked behavior (e.g., appearance, behavior, posture, respiratory rate and pattern). The animal showing signs of distress is then observed more closely followed by gentle handling and examination to measure body weight, body condition and temperature, heart rate, dehydration, and alertness. For some parameters, the degree of change from the normal scale is a useful evaluation indica- tor, the assumption being that the greater the deviation from normalcy, the greater the impact. For example, an animal may lose 5, 10, 20, or even 40 percent of its body weight, or its temperature may rise (or fall) by several degrees above (or below) normal. Clinical assessments can also be s ­ upplemented by video records of the animal in the colony room or labora- tory testing environment. A team approach during assessment is crucial. The assessment of dis- tress and subsequent interventions should involve researchers, ­veterinarians, and technicians, as they are often the first to observe signs of distress in individual animals. The team should similarly collaborate to develop an intervention strategy once the assessment is completed. REFERENCES Abelson, K. S., B. Adem, F. Royo, H. E. Carlsson, and J. Hau. 2005. High plasma corticosterone levels persist during frequent automatic blood sampling in rats. In Vivo 19:815-819. Ahmadiyeh, N., J. L. Sklone-Wilcoxon, J. S. Takahashi, and E. E. Redei. 2004. Maternal b ­ ehavior modulates X-linked inheritance of behavioral coping in the defensive burying test. Biol Psychiat 55:1069-1074. Akutsu, H., T. Kikusui, Y. Takeuchi, K. Sano, A. Hatanaka, and Y. Mori. 2002. Alleviating effects of plant-derived fragrances on stress-induced hyperthermia in rats. Physiol Behav 75:355-360. Anisman, H., M. D. Zaharia, M. J. Meaney, and Z. Merali. 1998. Do early-life events per- manently alter behavioral and hormonal responses to stressors? Int J Dev Neurosci 16:149-164. Arey, D. S. 1997. Behavioral observations of peri-parturient sows and the development of alternative farrowing accommodation: A review. Anim Welf 6(3):217-229. Armario, A., J. M. Castellanos, and J. Balasch. 1984. Dissociation between corticosterone and growth hormone adaptation to chronic stress in the rat. Horm Metab Res 16:142-145.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 45 Association of Primate Veterinarians. 2008. APV position statement on humane endpoints of nonhuman primates in research. Aston-Jones G., J. Rajkowski, P. Kubiak, R. J. Valentino, and M. T. Shipley. 1996. Role of the locus coeruleus in emotional activation. Prog Brain Res 107:379-402. Balogh, S. A. and J. M. Wehner. 2003. Inbred mouse strain differences in the establishment of long-term fear memory. Behav Brain Res 140:97-106. Bayart, F., K. T. Hayashi, K. F. Faull, J. D. Barchas, and S. Levine. 1990. Influence of ­maternal proximity on behavioral and physiological responses to separation in infant rhesus m ­ onkeys (Macaca mulatta). Behav Neurosci 104(1):98-107. Bayne, K. A. L. and M. A. Novak. 1998. Psychological disorders. In Nonhuman Primates in Biomedical Research, B. T. Bennett, C. R. Abee, and R. Henrickson, eds. Vol. II: Diseases. New York: Academic Press. 485-500 pp. Bayne, K. A. L., S. Dexter, and S. Suomi. 1992. A preliminary survey of the incidence of a ­ bnormal behavior in rhesus monkeys (Macaca mulatta) relative to housing condition. Lab Anim 22(5):38-44. Bayne, K. A. L., J. A. Mench, B. V. Beaver, and D. B. Morton. 2002. Laboratory Animal Behavior. In Laboratory Animal Science, ACLAM series, 2nd ed. New York: Academic Press. 1240-1264 pp. Beck, K. D. and V. N. Luine. 2002. Sex differences in behavioral and neurochemical profiles after chronic stress: Role of housing conditions. Physiol Behav 75(5):661-673. Beck, J. A., S. Lloyd, M. Hafezparast, M. Lennon-Pierce, J. T. Eppig, M. F. W. Festing, and E. M. C. Fisher. 2000. Genealogies of mouse inbred strains. Nat Genet 24:23-25. Bennett, B., C. Abee, and R. Henrickson, eds. 1998. Nonhuman Primates in Biomedical R ­ esearch: Diseases. New York: Academic Press. Berkson, G. and W. A. Mason. 1964. Stereotyped behaviors of chimpanzees: Relation to gen- eral arousal and other activities. Percept Motor Skill 19:635-652. Berman C. M., K. L. R. Rasmussen, and S. J. Suomi. 1994. Responses of free-ranging rhesus monkeys to a natural form of social separation. I. Parallels with mother-infant separation in captivity. Child Dev 65(4):1028-1041. Berry, A., F. Capone, M. Giorgio, P. G. Pelicci, E. R. de Kloet, E. Alleva, L. Minghetti, and F. Cirulli. 2007 Deletion of the life span determinant p66 (Shc) prevents age-dependent increases in emotionality and pain sensitivity in mice. Exp Gerontol 42(1-2):37-45. Blanc, J., M. L. Grichois, and J. L. Elghozi. 1991. Effects of clonidine on blood pressure and heart rate responses to an emotional stress in the rat: A spectral study. Clin Exp Pharmacol P 18(10):711-717. Blanchard, D. C., N. S. Canteras, C. M. Markham, N. S. Pentkowski, and R. J. Blanchard. 2005. Lesions of structures showing FOS expression to cat presentation: Effects on responsivity to a cat, cat odor, and nonpredator threat. Neurosci Biobehav R 29(8):1243-1253. Bolivar, V. J., B. J. Caldarone, A. A. Reilly, and L. Flaherty. 2000. Habituation of activity in an open field: A survey of inbred strains and F1 hybrids. Behav Genet 30(4):285-293. Bomholt, S. F., J. D. Mikkelsen, and G. Blackburn-Munro. 2005. Normal hypothalamo- p ­ ituitary-adrenal axis function in a rat model of peripheral neuropathic pain. Brain Res 1044(2):216-226. Bouwknecht, J. A. and R. Paylor. 2002. Behavioral and physiological mouse assays for anxiety: A survey in nine mouse strains. Behav Brain Res 136(2):489-501. Broida, J. and B. Svare. 1982. Strain typical patterns of pregnancy-induced nestbuilding in mice: Maternal and experiential influences. Physiol Behav 25:153-157. Bronson, F. H. 1979. The reproductive ecology of the house mouse. Quart Rev Biol 54: 265-299. Canteras, N. S. 2002. The medial hypothalamic defensive system: Hodological organization and functional implications. Pharmacol Biochem Behav 71(3):481-491.

46 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Coe, C. L., G. R. Lubach, W. B. Ershler, and R. G. Klopp. 1989. Influence of early rearing on lymphocyte proliferation response s in juvenile rhesus monkeys. Brain Behav Immun 3(1):47-60. Crawley, J. N. 2000. What’s Wrong with My Mouse? Behavioral Phenotyping of Transgenic and Knockout Mice. New York: Wiley-Liss, John Wiley & Sons, Inc. Crawley, J. N., J. K. Belknap, A. Collins, J. C. Crabbe, W. Frankel, N. Henderson, R. J. H ­ itzemann, S. C. Maxson, L. L. Miner, A. J. Silva, J. M. Wehner, A. Wynshaw-Boris, and R. Paylor. 1997. Behavioral phenotypes of inbred mouse strains: Implications and recom- mendations for molecular studies. Psychopharmacology 132(2):107-124. Dalla, C., K. Antoniou, G. Drossopoulou, M. Xagoraris, N. Kokras, A. Sfikakis, and Z. P ­ apadopoulou-Daifoti. 2005. Chronic mild stress impact: Are females more vulnerable? Neuroscience 135(3):703-714. Dantzer, R. 1986. Behavioral, physiological, and functional aspects of stereotyped behavior: A review and reinterpretation. J Anim Sci 62(6):1776-1786. Dantzer, R. 2004. Cytokine-induced sickness behaviour: A neuroimmune response to activa- tion of innate immunity. Euro J Pharmacol 500(1-3):399-411. Davenport, M. D., S. Tiefenbacher, C. K. Lutz, M. A. Novak, and J. S. Meyer. 2006. Analysis ��������������� of endogenous cortisol concentrations in the hair of rhesus macaques. Gen Comp Endocr 147(3):255-261. Davis, M. and P. J. Whalen. 2001. The amygdala: Vigilance and emotion. Mol Psychiat 6(1):13-24. Dawkins, M. S. 1990. From an animal’s point of view: Motivation, fitness, and animal welfare. Behav Brain Sci 13(1):1-61. Deak, T., K. T. Nguyen, M. Fleshner, L. R. Watkins, and S. F. Maier. 1999. Acute stress may facilitate recovery from a subcutaneous bacterial challenge. Neuroimmunomodulation 6(5):344-354. Decker, M. W., M. A. Pelleymounter, and M. Gallagher. 1988. Effects of training on a spatial memory task on high affinity choline uptake in hippocampus and cortex in young adult and aged rats. J Neurosci 8(1):90-99. Dhabar, F. S. and K. Viswanathan. 2005. Short-term stress experienced at time of immuniza- tion induces a long lasting increase in immunological memory. Am J Physiol Reg-I 289: R738-R744. Draper, W. A. and I. S. Bernstein. 1963. Stereotyped behavior and cage size. Percept Motor Skill 16:231-234. Ducottet, C. and C. Belzung. 2005. Correlations between behaviours in the elevated plus-maze and sensitivity to unpredictable subchronic mild stress: Evidence from inbred strains of mice. Behav Brain Res 156(1):153-162. Felten, D. L., S. Y. Felten, S. L. Carlson, J. A. Olschowka, and S. Livnat. 1985. Noradrenergic and peptidergic innervation of lymphoid tissue. J Immunol 135(2 Suppl):755-765. Figueiredo, H. F., C. M. Dolgas, and J. P. Herman. 2002. Stress activation of cortex and hippo- campus is modulated by sex and stage of estrus. Endocrinology 143(7):2534-2540. Fischer, W., K. S. Chen, F. H. Gage, and A. Bjorklund. 1992. Progressive decline in spatial learning and integrity of forebrain cholinergic neurons in rats during aging. Neurobiol Aging 13(1):9-23. Fish, E. W., D. Shahrokh, R. Bagot, C. Caldji, T. Bredy, M. Szyf, and M. J. Meaney. 2004. Epigenetic programming of stress responses through variations in maternal care. Ann NY Acad Sci 1036:167-180. Fleshner, M., J. Nermann, L. Lockwood, M. L. Laudenslager, L. R. Watkins, and S. F. Maier. 1995. Stressed rats fail to expand the CD45RC+CD4+ (th1-like) T cell subset in response to KLH: Possible involvement of interferon gamma. Brain Behav Immun 9(2):101-112.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 47 Fleshner, M., K. T. Nguyen, C. S. Cotter, L. R. Watkins, and S. F. Maier. 1998. Acute stressor exposure both suppresses acquired immunity and potentiates innate immunity. Am J Physiol 275(3 Pt 2):R870-R878. Fortman, J. D., T. A. Newett, and R. T. Bennett. 2002. The Laboratory Nonhuman Primate. Boca Raton, FL: CRC Press. Fox, J. G., L. C. Anderson, F. M. Loew, and F. W. Quimby, eds. 2002. Laboratory Animal Medicine, 2nd ed. San Diego: Academic Press. Francis, D. D., F. A. Champagne, D. Liu, and M. J. Meaney. 1999. Maternal care, gene expres- sion, and the development of individual differences in stress reactivity. Ann NY Acad Sci 896:66-84. Frick, K. M., L. A. Burlingame, J. A. Arters, and J. Berger-Sweeney. 2000. Reference memory, anxiety and estrous cyclicity in C57BL/6NIA mice are affected by age and sex. Neuro­ science 95(1):293-307. Garner, J. P. 2006. Perseveration and stereotypy: Systems-level insights from clinical p ­ sychology. In Stereotypic Animal Behavior: Fundamentals and Applications to Welfare, 2nd ed, G. Mason and J. Rushen, eds. Wallingford, UK: CAB International. 121-153 pp. Garner, J. P. and G. Mason. 2002. Evidence for a relationship between cage stereotypies and behavioural disinhibition in laboratory rodents. Behav Brain Res 136(1):83-92. Garner, J. P., C. L. Meehan, and J. A. Mench. 2003. Stereotypies in caged parrots, schizophrenia and autism: Evidence for a common mechanism. Behav Brain Res 145(1-2):125-134. Garner, J. P., S. M. Weisker, B. Dufour, and J. A. Mench. 2004. Barbering (fur and whisker trimming) by laboratory mice as a model of human trichotillomania and obsessive- c ­ ompulsive spectrum disorder. Comparative Med 54(2):216-224. Goto, S., R. Takahashi, S. Araki, and H. Nakamoto. 2002. Dietary restriction initiated in late adulthood can reverse age-related alterations of protein and protein metabolism. Ann NY Acad Sci 959:50-56. Guettinger, H. R. 1979. The integration of learnt and genetically programmed behaviour: A study of hierarchical organization in songs of canaries, greenfinches and their hybrids. Z Tierpsychol 49:285-303. Hawkins, R. K., H. G. Jansen, and S. Sanyal. 1985. Development and degeneration of retina in rds mutant mice: Photoreceptor abnormalities in the heterozygotes. Exp Eye Res 41(6):701-720. Heiderstadt, K. M., R. M. McLaughlin, D. C. Wright, S. E. Walker, and C. E. Gomez-Sanchez. 2000. The effect of chronic food and water restriction on open-filed behaviour and serum corticosterone levels in rats. Lab Anim 34(1):20-28. Heilig, M. 2004. The NPY system in stress, anxiety, and depression. Neuropeptides 38(4):213-224. Herman, J. P. and W. E. Cullinan. 1997. Neurocircuitry of stress: Central control of the h ­ ypothalamo-pituitary-adrenocortical axis. Trends Neurosci 20(2):78-84. Higley, J. D., S. J. Suomi, and M. Linnoila. 1991. CSF monoamine metabolite concentrations vary according to age, rearing, and sex, and are influenced by the stressor of social sepa- ration in rhesus monkeys. Psychopharmacology (Berl) 103(4):551-556. Hubrecht, R. C. 1997. Comfortable quarters for laboratory dogs. In: Comfortable Quarters for Laboratory Animals, Viktor Reinhardt, ed. Washington: Animal Welfare Institute. 63-74 pp. Hubrecht, R. C., J. A. Serpell, and T. B. Poole. 1992. Correlates of pen size and housing condi- tions on the behaviour of kennelled dogs. Appl Anim Behav Sci 34:365-383. Ichise, M., D. C. Vines, T. Gura, G. M. Anderson, S. J. Suomi, J. D. Higley, and R. B. Innis. 2006. Effects of early life stress on [11C]DASB positron emission tomography imaging of serotonin transporters in adolescent peer- and mother-reared rhesus monkeys. J Neurosci 26(17):4638-4643.

48 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Ingram, D. K. 2000. Age-related decline in physical activity: Generalization to nonhumans. Med Sci Sports Exerc 32(9):1623-1629. Ingram, D. K., E. D. London, M. A. Reynolds, S. B. Waller, and C. L. Goodrick. 1981. Dif- ferential effects of age on motor performance in two mouse strains. Neurobiol Aging 2(3):221-227. Ingram, D. K., J. Young, and J. A. Mattison. 2007. Calorie restriction in nonhuman primates: Assessing effects on brain and behavioral aging. Neuroscience 145(4):1359-1364. Johnson, R. P., Y. L. Yang, and C. J. Gordon. 2002. Peripheral cholinergic pathway modu- lates hyperthermia induced by stress in rats exposed to open-field stress. J Appl Physiol 92(1):789-794. Johnson, R. R., T. W. Prentice, P. Bridegam, C. R. Young, A. J. Steelman, T. H. Welsh, C. J. Welsh, and M. W. Meagher. 2006. Social stress alters the severity and onset of the chronic phase of Theiler’s virus infection. J Neuroimmunol 175(1-2):39-51. Jordan, T. C., M. B. Hennessy, C. A. Gonzalez, and S. Levine. 1985. Social and environmental factors influencing mother-infant separation-reunion in squirrel monkeys. Physiol Behav 34(4):489-493. Keay, K. A. and R. Bandler. 2002. Parallel circuits mediating distinct emotional coping r ­ eactions to different types of stress. Neurosci Biobehav R 25(7-8):669-678. Kempermann, G., D. Gast, and F. H. Gage. 2002. Neuroplasticity in old age: Sustained five- fold induction of hippocampal neurogenesis by long-term environmental enrichment. Ann Neurol 52(2):135-143. Kendrick, K. M., M. A. Haupt, M. R. Hinton, K. D. Broad, and J. D. Skinner. 2001. Sex differ- ences in the influence of mothers on the sociosexual preferences of their offspring. Horm Behav 40(2):322-338. Kliethermes, C. L. and J. C. Crabbe. 2006. Genetic independence of mouse measures of some aspects of novelty seeking. Proc Natl Acad Sci USA 103(13):5018-5023. Krief, S., A. Jamart, and C. M. Hladik. 2004. On the possible adaptive value of coprophagy in free-ranging chimpanzees. Primates 45(2):141-145. Krohn, T.C., J. Ritskes-Hoitinga, and P. Svendsen. 1999. The effects of feeding and housing on the behaviour of the laboratory rabbit. Lab Anim 33(2):101-107. Kunkele, J. 1992. Infanticide in wild rabbits (Oryctolagus cuniculus). J Mammal 73(2):317-320. Lamberty, Y. and A. J. Gower. 1992. Age-related changes in spontaneous behavior and learn- ing in NMRI mice from middle to old age. Physiol Behav 51(1):81-88. Latham, N. and G. Mason. 2004. From house mouse to mouse house: The behavioural ­biology of free-living Mus musculus and its implications in the laboratory. Appl Anim Behav 86(3-4):261-289. Laudenslager, M. L., M. Reite, and R. J. Harbeck. 1982. Suppressed immune response in infant monkeys associated with maternal separation. Behav Neural Biol 36(1):40-48. Laudenslager, M. L., P. E. Held, M. L. Boccia, M. L. Reite, and J. J. Cohen. 1990. Behavioral and immunological consequences of brief mother-infant separation: A species compari- son. Dev Psychobiol 23(3):247-264. Levine, S. 2005. Developmental determinants of sensitivity and resistance to stress. Psycho- neuroendocrinology 30(10):939-946. Levine, S. and T. Mody. 2003. The long-term psychobiological consequences of intermittent postnatal separation in the squirrel monkey. Neuroscience Biobehav R 27(1-2):83-89. Levine, S., S. G. Wiener, and C. L. Coe. 1993. Temporal and social factors influencing b ­ ehavioral and hormonal responses to separation in mother and infant squirrel monkeys. Psychoneuroendocrinology 18(4):297-306.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 49 Lewis, M. H., M. F. Presti, J. B. Lewis, and C. A. Turner. The neurobiology of stereotypy I: Environ­mental complexity. In Stereotypic Animal Behavior: Fundamentals and Applica- tions to Welfare, 2nd ed, G. Mason and J. Rushen, eds. Wallingford, UK: CAB Interna- tional. 196 pp. Lewis, M. H., Y. Tanimura, L. W. Lee, and J. W. Bodfish. 2007. Animal models of restricted repetitive behavior in autism. Behav Brain Res 176(1):66-74. Li, S. G., J. E. Lawler, D. C. Randall, and D. R. Brown. 1997. Sympathetic nervous activity and arterial pressure responses during rest and acute behavioral stress in SHR versus WKY rats. J Autonom Nerv Syst 62(3):147-154. Lowry, C. A. 2002. Functional subsets of serotonergic neurons: Implications for control of the HPA axis. J Neuroendocrinol 14(11):911-923. Lubach, G. R., C. L. Coe, and W. B. Ershler. 1995. Effects of early rearing environment on immune responses on infant rhesus monkeys. Brain Behav Imm 9(1):31-46. Lutz, C., S. Tiefenbacher, M. Jorgensen, J. Meyer, and M. A. Novak. 2000. Techniques for collecting saliva from awake, unrestrained, adult macaque monkeys for cortisol assay. Am J Primatol 52(2):93-99. Maier, S. F. and L. R. Watkins. 1998. Cytokines for psychologists: Implications of bi-­directional immune-to-brain communication for understanding behavior, mood, and thought. ­Psychol Rev 105(1):83-107. Mangiarini, L., K. Sathasivam, M. Seller, B. Cozens, A. Harper, C. Hetherington, M. Lawton, Y. Trottier, H. Lehrach, S. W. Davies, and G. P. Bates. 1996. Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87(3):493-506. Mason, G. 1991. Stereotypies: A critical review. Anim Behav 41(6): 1015-1037. Mason, G. 2006. Stereotypic behavior in captive animals: Fundamentals and implications for welfare and beyond. In Stereotypic Animal Behavior: Fundamentals and Applications to Welfare, 2nd ed, G. Mason and J. Rushen, eds. Wallingford, UK: CAB International. 325-357 pp. Mason, G. and M. Mendl. 1997. Do the stereotypies of pigs, chickens and mink reflect adap- tive species differences in the control of foraging? Appl Anim Behav Sci: 53(1-2):45-58. Mason, G. J. and N. R. Latham. 2004. Can’t stop, won’t stop: Is stereotypy a reliable animal welfare indicator? Anim Welf 13(Suppl):557-569. McCormick, C. M., J. W. Smythe, S. Sharma, and M. J. Meaney. 1995. Sex-specific effects of prenatal stress on hypothalamic-pituitary-adrenal responses to stress and brain gluco­ corticoid receptor density in adult rats. Dev Brain Res 84(1):55-61. McLaren, G. W., D. W. Macdonald, C. Georgiou, F. Mathews, C. Newman, and R. Mian. 2003. Leukocyte coping capacity: A novel technique for measuring the stress response in vertebrates. Exp Physiol 88(4):541-546. Meaney, M. J., J. Diorio, D. Francis, J. Widdowson, P. LaPlante, C. Caldji, S. Sharma, J. R. Seckl, and P. M. Plotsky. 1996. Early environmental regulation of forebrain gluco­ corticoid ­ receptor gene expression: Implications for adrenocortical responses to stress. Dev ­Neurosci 18(1-2):49-72. Miller, J. B. and J. P. O’Callaghan. 2002. Neuroendocrine aspects of the response to stress. Metabolism 51(6 Suppl):5-10. Mineur, Y. S., D. J. Prasol, C. Belzung, and W. E. Crusio. 2003. Agonistic behavior and unpre- dictable chronic mild stress in mice. Behav Genet 33(5):513-519. Morton, D. B. 2002. Behaviour of rabbits and rodents. In The Ethology of Domestic Animals— An Introductory Text, P. Jensen, ed. Oxford: CABI Wallingford Oxford. 193-209 pp. Morton, D. B. and P. H. M. Griffiths. 1985. Guidelines on the recognition of pain, distress, and discomfort in experimental animals and an hypothesis for assessment. Vet Record 116(16):431-436.

50 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Moy, S. S., J. J. Nadler, A. Perez, R. P. Barbaro, J. M. Johns, T. R. Magnuson, J. Piven, and J. N. Crawley. 2004. Sociability and preference for social novelty in five inbred strains: An approach to assess autistic-like behavior in mice. Genes Brain Behav 3(5):287-302. Muglia, L., L. Jacobson, P. Dikkes, and J. A. Majzoub. 1995. Corticotropin-releasing hor- mone deficiency reveals major fetal but not adult glucocorticoid need. Nature 373(6513):427-432. Nash, L. T., J. Fritz, P. A. Alford, and L. Brent. 1999. Variables influencing the origins of diverse abnormal behaviors in a large sample of captive chimpanzees (Pan troglodytes). Am J Primatol 48(1):15-29. Nemeroff, C.B. and W. W. Vale. 2005. The neurobiology of depression: Inroads to treatment and new drug discovery. J Clin Psychiat 66(Suppl 7):5-13. Novak, M. A. 2003. Self-injurious behavior in rhesus monkeys: New insights into its etiology, physiology, and treatment. Am J Primatol 59(1):3-19. Novak, M. A. and S. J. Suomi. 1988. Psychological well-being of primates in captivity. Am Psychol 43(10):765-73. O’Connor, K. A., J. D. Johnson, M. K. Hansen, J. L. Wiesler-Frank, L. R. Watkins, and S. F. Maier. 2003. Peripheral and central proinflammatory cytokine response to a severe acute stressor. Brain Res 991:123-132. Olsson, I. A. S. and C. M. Sherwin. 2006. Behaviour of laboratory mice in different housing conditions when allowed to self-administer an anxiolytic. Lab Anim 40(4):392-399. Parker, K. J., C. L. Buckmaster, K. Sundlass, A. F. Schatzberg, and D. M. Lyons. 2006. Maternal mediation, stress inoculation, and the development of neuroendocrine stress resistance in primates. Proc Nat Acad Sci USA 103(8):3000-3005. Pezze, M. A. and J. Feldon. 2004. Mesolimbic dopaminergic pathways in fear conditioning. Prog Neurobiol 74(5):301-320. Randall, D. C., D. R. Brown, L. V. Brown, and J. M. Kilgore. 1994. Sympathetic nervous activity and arterial blood pressure control in conscious rat during rest and behavioral stress. Am J Physiol 267(5 Part 2):R1241-R1249. Ribeiro, S. C., S. E. Kennedy, Y. R. Smith, C. S. Stohler, and J. K. Zubieta. 2005. Interface of physical and emotional stress regulation through the endogenous opioid system and mu- opioid receptors. Prog Neuropsychopharmacol Biol Psychiatry 29(8):1264-1280. Ruppenthal, G. C., C. G. Walker, and G. P. Sackett. 1991. Rearing infant monkeys (Macaca nemestrina) in pairs produces deficient social development compared with rearing in single cages. Am J Primatol 25(2):103-113. Ruppenthal, G. C., M. K. Harlow, C. D. Eisele, H. F. Harlow, and S. J. Suomi. 1974. Develop- ment of peer interactions of monkeys reared in a nuclear-family environment. Child Dev 45(3):670-682. Sackett, G. P., G. C. Ruppenthal, and A. E. Davis. 2002. Survival, growth, health, and repro- duction following nursery rearing compared with mother rearing in pigtailed monkeys (Macaca nemestrina). Am J Primatol 56(3):165-183. Sanders, V. M., D. J. Kasprowicz, A. P. Kohm, and M. A. Swanson. 2001. Neurotransmitter receptors on lymphocytes and other lymphoid cells. In Psychoneuroimmunology, 3rd ed. R. Ader, D. L. Felten, and N. Cohen, eds. New York: Academic Press. 161-197 pp. Serrats, J. and P. E. Sawchenko. 2006. CNS activational responses to staphylococcal ­enterotoxin B: T-lymphocyte-dependent immune challenge effects on stress-related circuitry. J Comp Neurol 495(2):236-254. Sherwin, C. 1998. Voluntary wheel running: A review and novel interpretation. Anim Behav 56(1):11-27. Silver, L. M. 1995. Mouse Genetics: Concepts and Applications. New York: Oxford University Press.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 51 Smith, L. K. and G. A. Metz. 2005. Dietary restriction alters fine motor function in rats. Physiol Behav 85(5):581-592. Snowdon, C. T. and A. Savage. 1989. Psychological well-being of captive primates: General considerations and examples from callitrichids. In Housing, Care and Psychological Wellbeing of Captive and Laboratory Primates, E. F. Segal, ed. Park Ridge, NJ: Noyes Publications. 75-88 pp. Sotres-Bayon, F., C. F. Cain, and J. E. LeDoux. 2006. Brain mechanisms of fear extinc- tion: Historical perspectives on the contributions of prefrontal cortex. Biol Psychiat 60(4):329-336. Stenzel-Poore, M., S. C. Heinrichs, S. Rivest, G. F. Koob, and W. W. Vale. 1994. Over­ production of corticotrophin-releasing factor in transgenic mice: A genetic model of anxiogenic behavior. J Neurosci 14(5 Pt 1):2579-2584. Suomi, S. J. 2004. How gene-environment interactions shape biobehavioral development: L ­ essons from studies with rhesus monkeys. Res Hum 1:205-222. Thomas, G. 2002. The S6 kinase signaling pathway in the control of development and growth. Biol Res 35(2):305-313. Tiefenbacher, S. T., M. A. Novak, C. K. Lutz, and J. S. Meyer. 2005. The physiology and neurochemistry of self-injurious behavior: A nonhuman primate model. Front Biosci 10(1):1-11. Tournier, J. N., J. Mathieu, Y. Mailfert, E. Multon, C. Drouet, A. Jouan, and E. Drouet. 2001. Chronic restraint stress induces severe disruption of the T-cell specific response to tetanus toxin vaccine. Immunology 102(1):515-523. Ulrich-Lai, Y. M, W. Xie, J. T. Meij, C. M. Dolgas, L. Yu, and J. P. Herman. 2006. Limbic and HPA axis function in an animal model of chronic neuropathic pain. Physiol Behav 88(1-2):67-76. van Bogaert, M. J., L. Groenink, R. S. Oosting, K. G. Westphal, J. van der Gugten, and B. Olivier. 2006. Mouse strain differences in autonomic responses to stress. Genes Brain Behav 5(2):139-149. van Praag, H., G. Kempermann, and F. H. Gage. 2000. Neural consequences of environmental enrichment. Nat Rev Neurosci 1(3):191-198. Vanderschuren, L. J., R. J. Niesink, and J. M. Van Ree. 1997. The neurobiology of social play behavior in rats. Neurosci Biobehav R 21(3):309-326. Vogt, J. L., C. L. Coe, E. Lowe, and S. Levine. 1980. Behavioral and pituitary-adrenal ­response of adult squirrel monkeys to mother-infant separation. Psychoneuroendocrinology 5(3):181-190. Wallace, C. S., V. L. Kilman, G. S. Withers, and W. T. Greenough. 1992. Increases in dendritic length in occipital cortex after 4 days of differential housing in weanling rats. Behav Neural Biol 58(1):64-68. Weaver, I. C. G., M. J. Meaney, and M. Szyf. 2006. Maternal care effects on the hippocampal transcriptome and anxiety-mediated behaviors in the offspring that are reversible in adult- hood. Proc Natl Acad Sci USA 103(9):3480-3485. Weaver, I. C. G., N. Cervoni, F. A. Champagne, A. C. D’Alessio, S. Sharma, J. R. Seckl, S. Dymov, M. Szyf, and M. J. Meaney. 2004. Epigenetic programming by maternal behavior. Nat Neurosci 7:847-854. Windle, R. J., S. A. Wood, N. Shanks, S. L. Lightman, and C. D. Ingram. 1998b. Ultradian rhythm of basal corticosterone release in the female rat: Dynamic interaction with the response to acute stress. Endocrinology 139(2):443-450. Wolfer, D. P., O. Litvin, S. Morf, R. M. Nitsch, H. P. Lipp, and H. Würbel. 2004. Cage enrich- ment and mouse behavior. Nature 432(7019):821-822. Würbel, H. 2001. Ideal homes? Housing effects on rodent brain and behaviour. Trends Neu- rosci 24(4):207-211.

52 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Zhang, W. and P. Thoren. 1998. Hyper-responsiveness of adrenal sympathetic nerve activity in spontaneously hypertensive rats to ganglionic blockade, mental stress and neuron­ glucopenia. Pflugers Arch 437(1):56-60. Additional References Page 26  The identification of species-typical behavior often comes from ethograms devel- oped by researchers to describe the kinds of behavior that animals display in various settings (Bronson 1979). Chopra, P. K., P. K. Seth, and S. Seth. 1992. Behavioural profile of free-ranging rhesus m ­ onkeys. Primate Rep 32:75-105. Latham, N. and G. Mason. 2004. From house mouse to mouse house: The behavioural biol- ogy of free-living Mus musculus and its implications in the laboratory. Appl Anim Behav 86(3-4):261-289. Mayer, J. 2007. Use of behavior analysis to recognize pain in small animals. Lab Anim 36(6):43-48. Morton, D. B. 2002. Behaviour of rabbits and rodents. In The Ethology of Domestic Animals— An Introductory Text, Per Jensen, ed. Oxford: CABI Wallingford Oxford. 193-209 pp. Morton, D. B. and P. H. M. Griffiths. 1985. Guidelines on the recognition of pain, distress and discomfort in experimental animals and an hypothesis for assessment. Vet Record 116(16):431-436. Olivier B. and D. van Dalen. 1982. Social behaviour in rats and mice: An ethologically based model for differentiating psychoactive drugs. Aggress Behav 8(2):163-168. Page 27  Such age-related changes (e.g., hearing and vision deficits) have been documented for a number of murine strains (Hawkins et al. 1985). Johnson, K. R., Q. Y. Zheng, and K. Noben-Trauth. 2006. Strain background effects and genetic modifiers of hearing in mice. Brain Res 1091(1):79-88. Jones, S. M., T. A. Jones, K. R. Johnson, H. Yu, L. C. Erway, and Q. Y. Zheng. 2006. A comparison of vestibular and auditory phenotypes in inbred mouse strains. Brain Res 1091(1):40-46. Malek, G., L. V. Johnson, B. E. Mace, P. Saloupis, D. E. Schmechel, D. W. Rickman, C. A. Toth, P. M. Sullivan, and C. Bowes Rickman. 2005. Apolipoprotein E allele-dependent pathogenesis: A model for age-related retinal degeneration. Proc Natl Acad Sci USA 102(33):11900-11905. Ohlemiller, K. K. 2006. Contributions of mouse models to understanding of age- and noise- related hearing loss. Brain Res 1091(1):89-102. Smith, R. S., S. W. John, A. Zabeleta, M. T. Davisson, N. L. Hawes, and B. Chang. 2000. The bst locus on mouse chromosome 16 is associated with age-related subretinal neovascu- larization. Proc Natl Acad Sci USA 97(5):2191-2195. Page 28  In fact, inbred strains of mice differ in almost every behavioral, sensory, motor, and physiological trait studied to date, such as anxiety, learning and memory, brain structure and size, visual acuity, acoustic startle, exploratory behavior, alcohol sensitivity, depression, pain sensitivity, and motor coordination (Crawley et al. 2000). Bolivar, V. J., B. J. Caldarone, A. A. Reilly, and L. Flaherty. 2000. Habituation of activity in an open field: A survey of inbred strains and F1 hybrids. Behav Genet 30(4):285-293.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 53 Bolivar, V. J., O. Pooler, and L. Flaherty. 2001. Inbred strain variation in contextual and cued fear conditioning behavior. Mamm Genome 12(8):651-656. Bothe, G. W. M., V. J. Bolivar, M. J. Vedder, and J. G. Geistfeld. 2005. Behavioral differences among fourteen inbred mouse strains commonly used as disease models. Comparative Med 55(4):326-334. Bouwknecht, J. A. and R. Paylor. 2002. Behavioral and physiological mouse assays for anxiety: A survey in nine mouse strains. Behav Brain Res 136(2):489-501. Cook, M. N., R. W. Williams, and L. Flaherty. 2001. Anxiety-related behaviors in the elevated zero-maze are affected by genetic factors and retinal degeneration. Behav Neurosci 115(2):468-476. Crabbe, J. C., D. Wahlsten, and B. C. Dudek. 1999. Genetics of mouse behavior: Interactions with laboratory environment. Science 284(5420):1670-1672. Crabbe, J. C., P. Metten, I. Ponomarev, C. A. Prescott, and D. Wahlsten. 2006. Effects of g ­ enetic and procedural variation on measurement of alcohol sensitivity in mouse inbred strains. Behav Genet 36(4):536-552. Guillot, P. V., P. L. Roubertoux, and W. E. Crusio. 1994. Hippocampal mossy fiber distributions and intermale aggression in seven inbred mouse strains. Brain Res 660(1):167-169. Kliethermes, C. L. and J. C. Crabbe. 2006. Genetic independence of mouse measures of some aspects of novelty seeking. Proc Natl Acad Sci USA 103(13):5018-5023. Liu, X. and H. K. Gershenfeld. 2001. Genetic differences in the tail-suspension test and its relationship to imipramine response among 11 inbred strains of mice. Biol Psychiat 49(7):575-581. Logue, S. F., E. H. Owen, D. L. Rasmussen, and J. M. Wehner. 1997. Assessment of locomotor activity, acoustic and tactile startle, and prepulse inhibition of startle in inbred mouse strains and F1 hybrids: Implications of genetic background for single gene and qualitative trait loci analyses. Neuroscience 80(4):1075-1086. Lucki, I., A. Dalvi, and A. J. Mayorga. 2001. Sensitivity to the effects of pharmacologically selec- tive antidepressants in different strains of mice. Psychopharmacology 155(3):315-322. McFadyen, M. P., G. Kusek, V. J. Bolivar, and L. Flaherty. 2003. Differences among eight inbred strains of mice in motor ability and motor learning on a rotorod. Genes Brain Behav 2(4):214-219. Mogil, J. S., S. G. Wilson, K. Bon, S. E. Lee, K. Chung, P. Raber, J. O. Pieper, H. S. Hain, J. K. Belknap, L. Hubert, G. I. Elmer, J. M. Chung, and M. Devor. 1999. Heritability of nociception I: Responses of 11 inbred mouse strains on 12 measures of nociception. Pain 80(1-2):67-82. Moy, S. S., J. J. Nadler, A. Perez, R. P. Barbaro, J. M. Johns, T. R. Magnuson, J. Piven, and J. N. Crawley. 2004. Sociability and preference for social novelty in five inbred strains: An approach to assess autistic-like behavior in mice. Genes Brain Behav 3(5):287-302. Owen, E. H., S. F. Logue, D. L. Rasmussen, and J. M. Wehner. 1997. Assessment of learning by the Morris water task and fear conditioning in inbred mouse strains and F1 hybrids: Implications of genetic background for single gene mutations and quantitative trait loci analyses. Neuroscience 80(4):1087-1099. Rustay, N. R., D. Wahlsten, and J. C. Crabbe. 2003. Influence of task parameters on rotarod performance and sensitivity to ethanol in mice. Behav Brain Res 141(2):237-249. Trullas, R. and P. Skolnick. 1993. Differences in fear motivated behaviors among inbred mouse strains. Psychopharmacology 111(3):323-331. van Bogaert, M. J., L. Groenink, R. S. Oosting, K. G. Westphal, J. van der Gugten, and B. Olivier. 2006. Mouse strain differences in autonomic responses to stress. Genes Brain Behav 5(2):139-149.

54 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Wahlsten, D., P. Metten, and J. C. Crabbe. 2003a. A rating scale for wildness and ease of handling laboratory mice: Results for 21 inbred strains tested in two laboratories. Genes Brain Behav 2(2):71-79. Wahlsten, D., P. Metten, and J. C. Crabbe. 2003b. Survey of 21 inbred mouse strains in two laboratories reveals that BTBR T/+ tf/tf has severely reduced hippocampal commissure and absent corpus callosum. Brain Res 971(1):47-54. Wahlsten, D., S. F. Cooper, and J. C. Crabbe. 2005. Different rankings of inbred mouse strains on the Morris maze and a refined 4-arm water escape task. Behav Brain Res 165(1):36-51. Willott, J. F., L. Tanner, J. O’Steen, K. R. Johnson, M. A. Bogue, and L. Gagnon. 2003. Acoustic startle and prepulse inhibition in 40 inbred strains of mice. Behav Neurosci 117(4):716-727. Wong, A. A. and R. E. Brown. 2005. Visual detection, pattern discrimination and visual acuity in 14 inbred strains. Genes Brain Behav 5(5):389-403. Page 28  For instance, inbred strains differ in performance on anxiety, depression, and fear learning assays (Balogh and Wehner 2003). Bolivar, V. J., O. Pooler, and L. Flaherty. 2001. Inbred strain variation in contextual and cued fear conditioning behavior. Mamm Genome 12(8):651-656. Bouwknecht, J. A. and R. Paylor. 2002. Behavioral and physiological mouse assays for anxiety: A survey in nine mouse strains. Behav Brain Res 136(2):489-501. Cook, M. N., R. W. Williams, and L. Flaherty. 2001. Anxiety-related behaviors in the elevated zero-maze are affected by genetic factors and retinal degeneration. Behav Neurosci 115(2):468-476. Trullas, R. and P. Skolnick. 1993. Differences in fear motivated behaviors among inbred mouse strains. Psychopharmacology 111(3):323-331. Page 32  Environments that elicit or enhance stereotypies not as part of defined patho­physiology or disease models are typically suboptimal ­(Berkson and Mason 1964). Garner, J. P. and G. Mason. 2002. Evidence for a relationship between cage stereotypies and behavioural disinhibition in laboratory rodents. Behav Brain Res 136(1):83-92. Garner, J. P., C. L. Meehan, and J. A. Mench. 2003. Stereotypies in caged parrots, schizophrenia and autism: Evidence for a common mechanism. Behav Brain Res 145(1-2):125-134. Mason, G. 1991. Stereotypies: A critical review. Anim Behav 41(6):1015-1037. Mason, G. J. and N. R. Latham. 2004. Can’t stop, won’t stop: Is stereotypy a reliable animal welfare indicator? Anim Welf 13(Suppl):557-569. Ridley, R. M. and H. F. Baker. 1982. Stereotypy in monkeys and humans. Psychol Med 12(1):61-72. Würbel, H. 2001. Ideal homes? Housing effects on rodent brain and behaviour. Trends N ­ eurosci 24(4):207-211. Page 32  Classic whole-body stereotypies include circling, pacing (dogs, primates), wall b ­ ouncing (dogs), and somersaulting and bar chewing (rodents), whereas self- or other-animal- directed stereotypies often involve the limbs and/or face and include such patterns as digit sucking, paw licking, and overgrooming (Bayne and Novak 1998). Callard, M. D., S. N. Bursten, and E. O. Price. 2000. Repetitive backflipping behaviour in captive roof rats (Rattus rattus) and the effects of cage enrichment. Anim Welf 9(2):139-152.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 55 Garner, J. P. 2005. Stereotypies and other abnormal repetitive behaviors: Potential impact on validity, reliability, and replicability of scientific outcomes. ILAR J 46(2):106-117. Hubrecht, R. C., J. A. Serpell, and T. B. Poole. 1992. Correlates of pen size and housing condi- tions on the behaviour of kennelled dogs. Appl Anim Behav Sci 34:365-383. Luescher, U. A., D. B. McKeown, and J. Halip. 1991. Stereotypic or obsessive-compulsive disorders in dogs and cats. Vet Clin North Am-Small 21(2):401-413. Morton, D. B., M. Jennings, G. R. Batchelor, D. Bell, L. Birke, K. Davies, J. R. Eveleigh, D. Gunn, M. Heath, B. Howard, P. Koder, J. Phillips, T. Poole, A. W. Sainsbury, G. Sales, D. J. A. Smith, M. Stauffacher, and R. J. Turner. 1993. Refinements in rabbit husbandry. Lab Anim 27(4):301-329. Waiblinger, E. and B. Konig. 2004. Refinement of gerbil housing and husbandry in the labora- tory. Anim Welf 13(Suppl):S229-S235. Page 34  Such protocols are widely used in gerontology research where diet has been shown to slow aging, extend lifespan, and reduce the incidence of age-related diseases in rodents (Goto et al. 2002). Goto, S., R. Takahashi, Z. Radak, and R. Sharma. 2007. Beneficial biochemical outcomes of late-onset dietary restriction in rodents. Ann N Y Acad Sci 1100:431-441. Jamieson, H. A., S. N. Hilmer, V. C. Cogger, A. Warren, R. Cheluvappa, D. R. Abernethy, A. V. Everitt, R. Fraser, R. de Cabo, and D. G. Le Couteur. 2007. Caloric restriction reduces age-related pseudocapillarization of the hepatic sinusoid. Exp Gerontol 42(4):374-378. Hyun, D. H., S. S. Emerson, D. G. Jo, M. P. Mattson, and R. de Cabo. 2006. Calorie restriction up-regulates the plasma membrane redox system in brain cells and suppresses oxidative stress during aging. Proc Natl Acad Sci USA 103(52):19908-19912. Seymour, E. M., R. V. Parikh, A. A. Singer, and S. F. Bolling. 2006. Moderate calorie restric- tion improves cardiac remodeling and diastolic dysfunction in the Dahl-SS rat. J Mol Cell Cardiol 41(4):661-668. Page 36  Types of behavior commonly explored to investigate the presence of stress include open-field activity, movements in an elevated plus maze, changes in innate behaviors (e.g., movement, grooming, feeding, sexual behavior), defensive behaviors (to external threats), and avoidance/escape (Beck and Luine 2002). Blanchard, R. J., C. R. McKittrick, and D. C. Blanchard. 2001. Animal models of social stress: Effects on behavior and brain neurochemical systems. Physiol Behav 73(3):261-271. Ely, D. R., V. Dapper, J. Marasca, J. B. Correa, G. D. Gamaro, M. H. Xavier, M. B. Michalowski, D. Catelli, R. Rosat, M. B. Ferreira, and C. Dalmaz. 1997. Effect of restraint stress on feeding behavior of rats. Physiol Behav 61(3):395-398. Harkin, A., T. J. Connor, J. M. O’Donnell, and J. P. Kelly. 2002. Physiological and behavioral responses to stress: What does a rat find stressful? Lab Anim 31(4):42-50. Mangiavacchi, S., F. Masi, S. Scheggi, B. Leggio, M. G. De Montis, and C. Gambarana. 2001. Long-term behavioral and neurochemical effects of chronic stress exposure in rats. J Neurochem 79(6):1113-1121. Pare, W. P. and S. M. Tejani-Butt. 1996. Effect of stress on the behavior and 5-HT system in Sprague-Dawley and Wistar Kyoto rat strains. Integr Physiol Beh Sci 31(2):112-121. Perrot-Sinal, T. S., A. Gregus, D. Boudreau, and L. E. Kalynchuk. 2004. Sex and repeated restraint stress interact to affect cat odor-induced defensive behavior in adult rats. Brain Res 1027:161-172.

56 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Retana-Marquez, S., E. D. Salazar, and J. Velazquez-Moctezuma. 1996. Effect of acute and chronic stress on masculine sexual behavior in the rat. Psychoneuroendocrinology 21(1):39-50. Rittenhouse, P. A., C. Lopez-Rubalcava, G. D. Stanwood, and I. Lucki. 2002. Amplified b ­ ehavioral and endocrine responses to forced swim stress in the Wistar-Kyoto rat. Psycho­ neuroendocrinology 27(3):303-318. Page 37  Glucocorticoids are typically measured in blood serum or plasma but can also be quantified in saliva, urine, feces, and hair (Abelson et al. 2005). Anderson, S. M., G. A. Saviolakis, R. A. Bauman, K. Y. Chu, S. Ghosh, and G. J. Kant. 1996. Effects of chronic stress on food acquisition, plasma hormones, and the estrous cycle of female rats. Physiol Behav 60:325-329. Atkinson, H. C., S. A. Wood, Y. M. Kershaw, E. Bate, and S. L. Lightman. 2006. Diurnal varia- tion in the responsiveness of the hypothalamic-pituitary-adrenal axis of the male rat to noise stress. J Neuroendocrinol 18(7):526-533. Belz, E. E., J. S. Kennell, R. K. Czambel, R. T. Rubin, and M. E. Rhodes. 2003. Environmental enrichment lowers stress-responsive hormones in singly housed male and female rats. Pharmacol Biochem Behav 76(3-4):481-486. Blanchard, R. J., C. R. McKittrick, and D. C. Blanchard. 2001. Animal models of social stress: Effects on behavior and brain neurochemical systems. Physiol Behav 73(3):261-271. Brown, K. J. and N. E. Grunberg. 1995. Effects of housing on male and female rats: Crowding stresses males but calms females. Physiol Behav 58(6):1085-1089. Cavigelli, S. A., S. L. Monfort, T. K. Whitney, Y. S. Mechref, M. Novotny, and M. K. ­McClintock. 2005. Frequent serial fecal corticoid measures from rats reflect circadian and ovarian cortico­sterone rhythms. J Endocrinol 184(1):153-163. Culman, J., I. J. Kopin, and J. M. Saavedra. 1991. Regulation of corticotropin-releasing hor- mone and pituitary-adrenocortical response during acute and repeated stress in the rat. Endocr Reg 25(3):151-158. Davenport, M. D, S. T. Tiefenbacher, C. K. Lutz, M. A. Novak, and J. S. Meyer. 2006. Analysis of endogenous cortisol concentrations in the hair of rhesus macaques. Gen Comp Endocr 147(3):255-261. De Boer, S. F., J. L. Slangen, and G. J. van der Gugten. 1988. Adaptation of plasma catechol- amine and corticosterone responses to short-term repeated noise stress in rats. Physiol Behav 44(2):273-280. De Boer, S. F., S. J. Koopmans, J. L. Slangen, and G. J. van der Gugten. 1990. Plasma c ­ atecholamine, corticosterone and glucose responses to repeated stress in rats: Effect of interstressor interval length. Physiol Behav 47(6):1117-1124. Dhabhar, F. S., B. S. McEwen, and R. L. Spencer. 1993. Stress response, adrenal steroid recep- tor levels and corticosteroid-binding globulin levels—a comparison between Sprague- Dawley, Fischer 344 and Lewis rats. Brain Res 616(1-2):89-98. Donnerer, J. and F. Lembeck. 1990. Different control of the adrenocorticotropin-corticosterone response and of prolactin secretion during cold stress, anesthesia, surgery, and nicotine injection in the rat: Involvement of capsaicin-sensitive sensory neurons. Endocrinology 126(2):921-926. Faraday, M. M., K. H. Blakeman, and N. E. Grunberg. 2005. Strain and sex alter effects of stress and nicotine on feeding, body weight, and HPA axis hormones. Pharmacol Biochem Be 80(4):577-589. Fediuc, S., J. E. Campbell, and M. C. Riddell. 2006. Effect of voluntary wheel running on circadian corticosterone release and on HPA axis responsiveness to restraint stress in Sprague-Daley rats. J Appl Physiol 100(6):1867-1875.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 57 Guo, A. L., F. Petraglia, M. Criscuolo, G. Ficarra, R. E. Nappi, M. Palumbo, A. Valentini, and A. R. Genazzani. 1994. Acute stress- or lipopolysaccharide-induced corticosterone secretion in female rats is independent of the oestrous cycle. Eur J Endocrinol 131(5):535-539. Hall, F. S., J. M. Sundstrom, J. Lerner, and A. Pert. 2001. Enhanced corticosterone release after a modified forced swim test in Fawn hooded rats is independent of rearing experience. Pharmacol Biochem Be 69(3-4):629-634. Harper, J. M. and S. N. Austad. 2000. Fecal glucocorticoids: A noninvasive method of measur- ing adrenal activity in wild and captive rodents. Physiol Biochem Zool 73(1):12-22. Ishikawa, M., S. Ohdo, H. Watanabe, C. Hara, and N. Ogawa. 1995. Alteration in circadian rhythm of plasma corticosterone in rats following sociopsychological stress induced by communication box. Physiol Behav 57(1):41-47. Kirschbaum, C. and D. H. Hellhammer. 1994. Salivary cortisol in psychoneuro­endocrine research: Recent developments and applications. Psychoneuroendocrinology 19(4):313-333. Klosterman, L. L, J. T. Murai, and P. K. Siiteri. 1986. Cortisol levels, binding, and prop- erties of corticosteroid-binding globulin in the serum of primates. Endocrinology 118(1):424-434. Ling, S. and F. Jamali. 2003. Effect of cannulation surgery and restraint stress on the plasma corticosterone concentration in the rat: Application of an improved corticosterone HPLC assay. J Pharm Pharm Sci 6(2):246-251. Livezey, G. T., J. M. Miller, and W. H. Vogel. 1985. Plasma norepinephrine, epinephrine and corticosterone stress responses to restraint in individual male and female rats, and their correlations. Neurosci Lett 62(1):51-56. Lynch, J. W., M. Z. Khan, J. Altmann, M. N. Njahira, and N. Rubenstein. 2003. Concentrations of four fecal steroids in wild baboons: Short-term storage conditions and consequences for data interpretation. Gen Comp Endocr 132(2):264-271. Marti, O., A. Gavalda, T. Jolin, and A. Armario. 1993. Effect of regularity of exposure to chronic immobilization stress on the circadian pattern of pituitary adrenal hormones, growth hormone, and thyroid stimulating hormone in the adult male rat. Psychoneuro- endocrinology 18(1):67-77. Moncek, F., R. Duncko, B. B. Johansson, and D. Jezova. 2004. Effect of environmental enrich- ment on stress related systems in rats. J Neuroendocrinol 16(5):423-431. Orr, T. E., J. L. Meyerhoff, E. H. Mougey, and B. N. Bunnell. 1990. Hyperresponsiveness of the rat neuroendocrine system due to repeated exposure to stress. Psychoneuroendocrinology 15(5-6):317-328. Rivier, C. and W. Vale. 1987. Diminished responsiveness of the hypothalamic-pituitary-­adrenal axis of the rat during exposure to prolonged stress: A pituitary-mediated mechanism. Endocrinology 121(4):1320-1328. Royo, F., N. Bjork, H. E. Carlsson, S. Mayo, and J. Hau. 2004. Impact of chronic catheteriza- tion and automated blood sampling (Accusampler) on serum corticosterone and fecal immuno­reactive corticosterone metabolites and immunoglobulin A in male rats. J Endo- crinol 180(1):145-153. Sakellaris, P. C. and J. Vernikos-Danellis. 1975. Increased rate of response of the pituitary- a ­ drenal system in rats adapted to chronic stress. Endocrinology 97(3):597-602. Sapolsky, R., M., L. C. Krey, and B. S. McEwen. 1983. The adrenocortical stress-response in the aged male rat: impairment of recovery from stress. Exp Gerontol 18(1):55-64. Tannenbaum, B. M., W. Rowe, S. Sharma, J. Diorio, A. Steverman, M. Walker, and M. J. Meaney. 1997a. Dynamic variations in plasma corticosteroid-binding globulin and basal HPA activity following acute stress in adult rats. J Neuroendocrinol 9(3):163-168. Tannenbaum, B. M., D. N. Brindley, G. S. Tannenbaum, M. F. Dallman, M. D. McArthur, and M. J. Meaney. 1997b. High-fat feeding alters both basal and stress-induced hypothalamic- pituitary-adrenal activity in the rat. Am J Physiol 273(6 Pt 1):E1168-E1177.

58 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Tuli, J., J. A. Smith, and D. B. Morton. 1995a. Corticosterone, adrenal and spleen weight in mice after tail bleeding, and its effect on nearby animals. Lab Anim 29(1):90-95. Tuli, J., J. A. Smith, and D. B. Morton. 1995b. Effects of acute and chronic restraint on the adrenal gland weight and serum corticosterone concentration of mice and their faecal output of oocysts after infection with Eimeria apionoides. Res Vet Sci 59(1):82-86. Tuli, J., J. A. Smith, and D. B. Morton. 1995c. Stress measurements in mice after transportation. Lab Anim 29(2):132-138. Vachon, P. and J. P. Moreau. 2001. Serum corticosterone and blood glucose in rats after two jugular vein blood sampling methods: Comparison of the stress response. Contemp Top Lab Anim 40(5):22-24. Vahl, T. P., Y. M. Ulrich-Lai, M. M. Ostrander, C. M. Dolgas, E. E. Elfers, R. J. Seeley, D. A. D’Alessio, and J. P. Herman. 2005. Comparative analysis of ACTH and corticosterone sampling methods in rats. Am J Physiol-Endoc M 289:E823-E828. Viau, V. and M. J. Meaney. 1991. Variations in the hypothalamic-pituitary-adrenal response to stress during the estrous cycle in the rat. Endocrinology 129(5):2503-2511. Viveros, M. P., R. Hernandez, I. Martinez, and P. Gonzalez. 1988. Effects of social isolation and crowding upon adrenocortical reactivity and behavior in the rat. Rev Esp Fisiol 44(3):315-321. Weinstock, M., M. Razin, D. Schorer-Apelbaum, D. Men, and R. McCarty. 1998. Gender dif- ferences in sympathoadrenal activity in rats at rest and in response to footshock stress. Int J Dev Neurosci 16(3-4):289-295. Windle, R. J., S. A. Wood, N. Shanks, P. Perks, G. L. Conde, A. P. da Costa, C. D. Ingram, and S. L. Lightman. 1997. Endocrine and behavioural responses to noise stress: Comparison of virgin and lactating female rats during non-disrupted maternal activity. J Neuroendo- crinol 9(6):407-414. Windle, R. J., S. A. Wood, S. L. Lightman, and C. D. Ingram. 1998a. The pulsatile character- istics of hypothalamo-pituitary-adrenal activity in female Lewis and Fischer 344 rats and its relationship to differential stress responses. Endocrinology 139(10):4044-4052. Page 38  While some (prolactin, α-MSH, oxytocin) increase during stress, others decrease (growth hormone, luteinizing hormone, prolactin), depending on the animal species and the physiological state in which stress occurs (Armario et al. 1984). Bingaman, E. W., L. D. Van de Kar, J. M. Yracheta, Q. Li, and T. S. Gray. 1995. Castration attenuates prolactin response but potentiates ACTH response to conditioned stress in the rat. Am J Physiol 269(4 Pt 2):R856-R863. Brown, G. M., D. S. Schalch, and S. Reichlin. 1971. Patterns of growth hormone and cortisol responses to psychological stress in the squirrel monkey. Endocrinology 88(4):956-963. Brown, G. M., J. Seggie, and J. Feldmann. 1977. Effect of psychosocial stimuli and limbic l ­esions on prolactin at rest and following stress. Clin Endocrinol 6(Suppl):29S-41S. Cates, P. S., M. L. Forsling, and K. T. O’byrne. 1999. Stress-induced suppression of pulsatile luteinising hormone release in the female rat: Role of vasopressin. J Neuroendocrinol 11(9):677-683. Collu, R., J. C. Jequier, J. Letarte, G. Leboeuf, and J. R. Ducharme. 1973. Effect of stress and hypothalamic deafferentation on the secretion of growth hormone in the rat. Neuro­ endocrinology 11(3):183-190. Cronin, M. T., B. J. Siegel, and G. P. Moberg. 1981. Effect of behavioral stress on plasma levels of growth hormone in sheep. Physiol Behav 26(5):887-890. Day, T. A., M. J. West, and J. O. Willoughby. 1983. Stress suppression of growth hormone s ­ ecretion in the rat: Effects of disruption of inhibitory noradrenergic afferents to the m ­ edian eminence. Aust J Biol Sci 36(5-6):525-530.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 59 Dunn, J. D., W. J. Schindler, M. D. Hutchins, L. E. Scheving, and C. Turpen. 1973. Daily variation in rat growth hormone concentration and the effect of stress on periodicity. Neuroendocrinology 13(1):69-78. Furudate, S., H. Ashihara, and T. Nakano. 1989. Prolactin secretion and its response to stress during the estrous cycle of the rats. Jikken Dobutsu 38(4):313-318. Gala, R. R. 1990. The physiology and mechanisms of the stress-induced changes in prolactin secretion in the rat. Life Sci 46(20):1407-1420. Gala, R. R. and D. J. Haisenleder. 1986. Restraint stress decreases afternoon plasma prolactin levels in female rats: Influence of neural antagonists and agonists on restraint-induced changes in plasma prolactin and corticosterone. Neuroendocrinology 43(2):115-123. Hashimoto, K., K. Murakami, T. Takao, S. Makino, M. Sugawara, and Z. Ota. 1989. Effect of acute ether or restraint stress on plasma corticotropin-releasing hormone, vasopressin and oxytocin levels in the rat. Acta Med Okayama 43(3):161-167. Husain, M. K., W. M. Manger, T. W. Rock, R. J. Weiss, and A. G. Frantz. 1979. Vasopressin release due to manual restraint in the rat: Role of body compression and comparison with other stressful stimuli. Endocrinology 104(3):641-644. Jahn, G. A. and R. P. Deis. 1986. Stress-induced prolactin release in female, male and andro- genized rats: Influence of progesterone treatment. J Endocrinol 110(3):423-428. Kant, G. J., R. H. Lenox, B. N. Bunnell, E. H. Mougey, L. L. Pennington, and J. L. Meyerhoff. 1983. Comparison of stress response in male and female rats: Pituitary cyclic AMP and plasma prolactin, growth hormone and corticosterone. Psychoneuroendocrinology 8(4):421-428. Khorram, O., J. C. Bedran de Castro, and S. M. McCann. 1985. Stress-induced secretion of alpha-melanocyte-stimulating hormone and its physiological role in modulating the secretion of prolactin and luteinizing hormone in the female rat. Endocrinology 117(6):2483-2489. Krulich, L., E. Hefco, P. Illner, and C. B. Read. 1974. The effects of acute stress on the secretion of LH, FSH, prolactin and GH in the normal male rat, with comments on their statistical evaluation. Neuroendocrinology 16(5-6):293-311. Lang, R. E., J. W. Heil, D. Ganten, K. Hermann, T. Unger, and W. Rascher. 1983. Oxytocin unlike vasopressin is a stress hormone in the rat. Neuroendocrinology 37(4):314-316. Mattheij, J. A. and T. A. van Pijkeren. 1977. Plasma prolactin in undisturbed cannulated male rats; effects of perphenazine, frequent sampling, stress and castration plus oestrone treat- ment. Acta Endocrinol 84(1):51-61. Morehead, M. H. and R. R. Gala. 1987. Restraint stress depresses prolactin surges in pseudo­ pregnant rats and adrenalectomy does not alter the response. Life Sci 41(12):1491-1498. Morehead, M. H., K. J. Lookingland, and R. R. Gala. 1990. Stress-induced suppression of the prolactin afternoon surge in ovariectomized, estrogen-treated rats and the nocturnal surge in pseudopregnant rats are accompanied by an increase in median eminence dihydroxy- phenylacetic acid concentrations. Neuroendocrinology 51(2):208-212. Muir, J. L. and H. P. Pfister. 1986. Corticosterone and prolactin responses to predictable and unpredictable novelty stress in rats. Physiol Behav 37(2):285-288. Neill, J. D. 1970. Effect of “stress” on serum prolactin and luteinizing hormone levels during the estrous cycle of the rat. Endocrinology 87(6):1192-1197. Reynaert, R., S. Marcus, M. De Paepe, and G. Peeters. 1976. Influences of stress, age and sex on serum growth hormone and free fatty acid levels in cattle. Horm Metab Res 8(2):109-114. Riegle, G. D. and J. Meites. 1976a. Effects of aging on LH and prolactin after LHRL L-dopa, methyl-dopa, and stress in male rat. P Soc Exp Biol Med 151(3):507-511. Riegle, G. D. and J. Meites. 1976b. The effect of stress on serum prolactin in the female rat. P Soc Exp Biol Med 152(3):441-448.

60 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Seggie, J. A. and G. M. Brown. 1975. Stress response patterns of plasma corticosterone, p ­ rolactin, and growth hormone in the rat, following handling or exposure to novel envi- ronment. Can J Physiol Pharmacol 53(4):629-637. Siegel, R. A., N. Conforti, and I. Chowers. 1980. Neural pathways mediating the prolactin secretory response to acute neurogenic stress in the male rat. Brain Res 198(1):43-53. Simms, D. D., L. V. Swanson, and R. Bogart. 1978. Effect of collection stress on serum growth hormone levels in pygmy goats. J Anim Sci 46(2):458-462. Yelvington, D. B., G. K. Weiss, and A. Ratner. 1984. Effect of corticosterone on the prolactin response to psychological and physical stress in rats. Life Sci 35(16):1705-1711. Page 39  Moreover, their usefulness is subjected to the same limitations as discussed above, although chronic indwelling vascular catheters and automated blood collection systems may circumvent this limitation to some degree (Abelson et al. 2005). Atkinson, H. C., S. A. Wood, Y. M. Kershaw, E. Bate, and S. L. Lightman. 2006. Diurnal varia- tion in the responsiveness of the hypothalamic-pituitary-adrenal axis of the male rat to noise stress. J Neuroendocrinol 18(7):526-533. Lightman, S. L., R. J. Windle, M. D. Julian, M. S. Harbuz, N. Shanks, S. A. Wood, Y. M. K ­ ershaw, and C. D. Ingram. 2000. Significance of pulsatility in the HPA axis. Novar Fnd Symp 227:244-257. Rogers, W. R., J. H. Lucas, B. C. Mikiten, H. D. Smith, and J. L. Orr. 1995. Chronically in- dwelling venous cannula and automatic blood sampling system for use with nonhuman primates exposed to 60 Hz electric and magnetic fields. Bioelectromagnetics Suppl 3:103-110. Windle, R. J., S. A. Wood, N. Shanks, P. Perks, G. L. Conde, A. P. da Costa, C. D. Ingram, and S. L. Lightman. 1997. Endocrine and behavioural responses to noise stress: Comparison of virgin and lactating female rats during non-disrupted maternal activity. J Neuroendo- crinol 9(6):407-414. Windle, R. J., S. A. Wood, S. L. Lightman, and C. D. Ingram. 1998a. The pulsatile character- istics of hypothalamo-pituitary-adrenal activity in female Lewis and Fischer 344 rats and its relationship to differential stress responses. Endocrinology 139(10):4044-4052. Windle, R. J., S. A. Wood, N. Shanks, S. L. Lightman, and C. D. Ingram. 1998b. Ultradian rhythm of basal corticosterone release in the female rat: Dynamic interaction with the response to acute stress. Endocrinology 139(2):443-450. Page 39  Many different types of stressors cause the rapid activation of the sympathetic division of the autonomic nervous system (ANS) (Blanc et al. 1991). Blanc, J., M. L. Grichois, M. Vincent, and J. L. Elghozi. 1994. Spectral analysis of blood pres- sure and heart rate variability in response to stress from air-jet in the Lyon rat. J Auton Pharmacol 14(1):37-48. Inagaki, H., M. Kuwahara, and H. Tsubone. 2004. Effects of psychological stress on autonomic control of heart in rats. Exp Anim 53(4):373-378. McDougall, S. J., J. R. Paull, R. E. Widdop, and A. J. Lawrence. 2000. Restraint stress: Dif- ferential cardiovascular responses in Wistar-Kyoto and spontaneously hypertensive rats. Hypertension 35:126-129. Saunders, P. R., P. Miceli, B. A. Vallance, L. Wang, S. Pinto, G. Tougas, M. Kamath, and K. Jacobson. 2006. Noradrenergic and cholinergic neural pathways mediate stress-induced reactivation of colitis in the rat. Autonom Neurosci 124(1-2):56-68.

RECOGNITION AND ASSESSMENT OF STRESS AND DISTRESS 61 Stiedl, O., M. Meyer, T. Kishimoto, M. G. Rosenfeld, and J. Spiess. 2003. Stress-mediated heart rate dynamics after deletion of the gene encoding corticotropin-releasing factor receptor 2. Eur J Neurosci 17(10):2231-2235. Tornatzky, W. and K. A. Miczek. 1994. Behavioral and autonomic responses to intermittent social stress: Differential protection by clonidine and metoprolol. Psychopharmacology 116(2):346-356. van den Buuse, M., G. Lambert, M. Fluttert, and N. Eikelis. 2001a. Cardiovascular and b ­ ehavioural responses to psychological stress in spontaneously hypertensive rats: Effect of treatment with DSP-4. Behav Brain Res 119(2):131-142. Wood, S. K., R. E. Verhoeven, A. Z. Savit, K. C. Rice, P. S. Fischbach, and J. H. Woods. 2006. Facilitation of cardiac vagal activity by CRF-R1 antagonists during swim stress in rats. Neuropsychopharmacology 31(12):2580-2590. Xie, Y. F., Q. Jiao, S. Guo, F. Z. Wang, J. M. Cao, and Z. G. Zhang. 2005. Role of parasym- pathetic overactivity in water immersion stress-induced gastric mucosal lesion in rat. J Appl Physiol 99(6):2416-2422. Zhang, W. and P. Thoren. 1998. Hyper-responsiveness of adrenal sympathetic nerve activity in spontaneously hypertensive rats to ganglionic blockade, mental stress and neuron­ glucopenia. Pflugers Arch 437(1):56-60. Page 39  Some stressors may also increase the activity of the parasympathetic division affecting both core body temperature and the gastrointestinal system (e.g., disturbed intestinal absorp- tion, gastric ulceration, colitis; Johnson et al. 2002). Ray, A., R. M. Sullivan, and P. G. Henke. 1987. Adrenergic modulation of gastric stress pathol- ogy in rats: A cholinergic link. J Auton Nerv Syst 20(3):265-268. Saunders, P. R., N. Hanssen, and M. H. Perdue. 1997. Cholinergic nerves mediate stress- induced intestinal transport abnormalities in Wistar-Kyoto rats. Am J Physiol 273: G486-G490. Saunders, P. R., P. Miceli, B. A. Vallance, L. Wang, S. Pinto, G. Tougas, M. Kamath, and K. Jacobson. 2006. Noradrenergic and cholinergic neural pathways mediate stress-induced reactivation of colitis in the rat. Autonom Neurosci 124(1-2):56-68. Xie, Y. F., Q. Jiao, S. Guo, F. Z. Wang, J. M. Cao, and Z. G. Zhang. 2005. Role of parasym- pathetic overactivity in water immersion stress-induced gastric mucosal lesion in rat. J Appl Physiol 99(6):2416-2422. Page 39  For example, telemetry in conscious, ­unrestrained animals is an effective method for the continuous monitoring of physiologic alterations in heart rate, respiration, blood pressure, ECG, and body temperature (Akutsu et al. 2002). Azar, T., J. Sharp, and D. Lawson. 2005. Stress-like cardiovascular responses to common procedures in male versus female spontaneously hypertensive rats. Contemp Top Lab Anim 44(3):25-30. Brockway, B. P., P. A. Mills, and S. H. Azar. 1991. A new method for continuous chronic measurement and recording of blood pressure, heart rate and activity in the rat via radio- telemetry. Clin Exp Hypertens A 13(5):885-895. Caplea, A., D. Seachrist, G. Dunphy, and D. Ely. 2000. SHR Y chromosome enhances the nocturnal blood pressure in socially interacting rats. Am J Physiol Heart Circ Physiol 279(1):H58-H66. Harkin, A., T. J. Connor, J. M. O’Donnell, and J. P. Kelly. 2002. Physiological and behavioral responses to stress: What does a rat find stressful? Lab Anim 31(4):42-50.

62 RECOGNITION AND ALLEVIATION OF DISTRESS IN LABORATORY ANIMALS Irvine, R. J., J. White, and R. Chan. 1997. The influence of restraint on blood pressure in the rat. J Pharmacol Toxicol 38(3):157-162. Kramer, K., J. A. Grimbergen, L. van der Gracht, D. J. van Iperen, R. J. Jonker, and A. Bast. 1995. The use of telemetry to record electrocardiogram and heart rate in freely swimming rats. Method Find Exp Clin 17(2):107-112. Lawson, D. M., M. Churchill, and P. C. Churchill. 2000. The effects of housing enrichment on cardiovascular parameters in spontaneously hypertensive rats. Contemp Top Lab Anim 39(1):9-13. Lemaire, V. and P. Mormede. 1995. Telemetered recording of blood pressure and heart rate in different strains of rats during chronic social stress. Physiol Behav 58(6):1181-1188. Nakashima, T., M. Akamatsu, A. Hatanaka, and T. Kiyohara. 2004. Attenuation of stress- induced elevations in plasma ACTH level and body temperature in rats by green odor. Physiol Behav 80(4):481-488. Rubini, R., A. Porta, G. Baselli, S. Cerutti, and M. Paro. 1993. Power spectrum analysis of cardiovascular variability monitored by telemetry in conscious unrestrained rats. J Auton Nerv Syst 45(3):181-190. Sato, K., F. Chatani, and S. Sato. 1995. Circadian and short-term variabilities in blood pres- sure and heart rate measured by telemetry in rabbits and rats. J Autonom Nerv Syst 54(3):235-246. Schierok, H., M. Markert, M. Pairet, and B. Guth. 2000. Continuous assessment of ­multiple vital physiological functions in conscious freely moving rats using telemetry and a p ­ lethysmography system. J Pharmacol Toxicol 43(3):211-217. Sharp, J., T. Zammit, T. Azar, and D. Lawson. 2002a. Does witnessing experimental procedures produce stress in male rats? Contemp Top Lab Anim 41(5):8-12. Sharp, J. L., T. Zammit, T. Azar, and D. M. Lawson. 2002b. Stress-like responses to com- mon procedures in male rats housed alone or with other rats. Contemp Top Lab Anim 41(4):8-14. Sharp, J. L., T. Zammit, and D. Lawson. 2002c. Stress-like responses to common procedures in rats: Effect of the estrous cycle. Contemp Top Lab Anim 41:15-22. Sharp, J., T. Zammit, T. Azar, and D. Lawson. 2003a. Are “by-stander” female Sprague-Dawley rats affected by experimental procedures? Contemp Top Lab Anim 42(1):19-27. Sharp, J., T. Zammit, T. Azar, and D. Lawson. 2003b. Stress-like responses to common procedures in individually and group-housed female rats. Contemp Top Lab Anim 42(1):9-18. Sharp, J., T. Azar, and D. Lawson. 2005a. Effects of a cage enrichment program on heart rate, blood pressure, and activity of male Sprague-Dawley and spontaneously hypertensive rats monitored by radiotelemetry. Contemp Top Lab Anim 44(2):32-40. Sharp, J., T. Azar, and D. Lawson. 2005b. Selective adaptation of male rats to repeated social encounters and experimental manipulations. Contemp Top Lab Anim 44(2):28-31. van den Buuse, M. 1994. Circadian rhythms of blood pressure, heart rate, and locomotor activity in spontaneously hypertensive rats as measured with radio-telemetry. Physiol Behav 55(4):783-787. van den Buuse. M., S. A. Van Acker, M. Fluttert, and E. R. De Kloet. 2001b. Blood pressure, heart rate, and behavioral responses to psychological “novelty” stress in freely moving rats. Psychophysiology 38(3):490-499. Wood, S. K., R. E. Verhoeven, A. Z. Savit, K. C. Rice, P. S. Fischbach, and J. H. Woods. 2006. Facilitation of cardiac vagal activity by CRF-R1 antagonists during swim stress in rats. Neuropsychopharmacology 31(12):2580-2590.

Next: 4 Avoiding, Minimizing, and Alleviating Distress »
Recognition and Alleviation of Distress in Laboratory Animals Get This Book
×
Buy Paperback | $52.00
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

Scientific advances in our understanding of animal physiology and behavior often require theories to be revised and standards of practice to be updated to improve laboratory animal welfare. This new book from the Institute for Laboratory Animal Research (ILAR) at the National Research Council, Recognition and Alleviation of Distress in Laboratory Animals, focuses on the stress and distress which is experienced by animals when used in laboratory research. This book aims to educate laboratory animal veterinarians; students, researchers, and investigators; animal care staff, as well as animal welfare officers on the current scientific and ethical issues associated with stress and distress in laboratory animals. It evaluates pertinent scientific literature to generate practical and pragmatic guidelines. Recognition and Alleviation of Distress in Laboratory Animals focuses specifically on the scientific understanding of the causes and the functions of stress and distress, the transformation of stress to distress, and the identification of principles for the recognition and alleviation of distress. This book discusses the role of humane endpoints in situations of distress and principles for the minimization of distress in laboratory animals. It also identifies areas in which further scientific investigation is needed to improve laboratory animal welfare in order to adhere to scientific and ethical principles that promote humane care and practice.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  6. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  7. ×

    View our suggested citation for this chapter.

    « Back Next »
  8. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!