Grimace scale

Last updated
A drawing by Konrad Lorenz showing facial expressions of a dog Lorenz emotions.png
A drawing by Konrad Lorenz showing facial expressions of a dog

The grimace scale (GS), sometimes called the grimace score, is a method of assessing the occurrence or severity of pain experienced by non-human animals according to objective and blinded scoring of facial expressions, as is done routinely for the measurement of pain in non-verbal humans. Observers score the presence or prominence of "facial action units" (FAU), e.g. Orbital Tightening, Nose Bulge, Ear Position and Whisker Change. These are scored by observing the animal directly in real-time, or post hoc from photographs or screen-grabs from videos. The facial expression of the animals is sometimes referred to as the pain face.

Contents

The GS method of pain assessment is highly applicable to laboratory rodents as these are usually prey species which tend to inhibit the expression of pain to prevent appearing vulnerable to predators. For this reason, behavioural changes in these species are mainly observed with acute pain (hours) but are less pronounced in longer-lasting pain (days). [1]

For mice at least, the GS has been shown to be a highly accurate, repeatable and reliable means of assessing pain requiring only a short period of training for the observer. [2] [3] Across species, GS are proven to have high accuracy and reliability, and are considered useful for indicating both procedural and postoperative pain, and for assessing the efficacy of analgesics. [4] [5]

The overall accuracy of GS is reported as 97% for mice, 84% for rabbits, 82% for rats and 73.3% for horses.[ citation needed ]

History

Facial expressions have long been considered as indicators of emotion in both human and non-human animals. The biologist Charles Darwin considered that non-human animals exhibit similar facial expressions to emotional states as do humans. [6] The assessment of changes in human anatomy during facial expressions were successfully translated from humans to non-human primates, such as the chimpanzee (ChimpFACS) [7] and rhesus macaque (MaqFACS), [8] but were not originally applied to assess pain in these species. In 2010, a team of researchers successfully developed [9] the first method to assess pain using changes in facial expression in any non-human animal species. Broadly speaking, GS quantify spontaneous pain according to objective and blinded scoring of facial expressions, as is done routinely for the measurement of pain in non-verbal humans. Observers score the presence and extent of "facial action units" (FAU), e.g. Orbital Tightening, Nose Bulge, Ear Position and Whisker Change. These are scored in real-time by observing the animal directly, or, post hoc from photographs or screen-grabs from videos.

This method of pain assessment is highly applicable to prey animals which tend to inhibit the overt expression of pain to prevent appearing vulnerable to predators. For this reason, behavioural changes in these species are mainly observed with acute pain (hours) but are less pronounced in longer-lasting pain (days). [1]

GS offer advantages over other methods of pain assessment. For example, the analgesic morphine reduces pain but can affect other aspects of behaviour in pain-free animals, for example, excitement, increased activity or sedation, which can hamper traditional behavioural assessment of its action on pain. Morphine not only reduces the frequency of "pain faces" but has no effect on GS in baseline, pain-free mice. [10]

In mice

The GS for mice usually consists of five FAU, i.e. Orbital Tightening, Nose Bulge, Cheek Bulge, Ear position and Whisker Change. These are scored on a 0-2 scale where 0=the criterion is absent, 1=moderately present and 2=obviously present. In mice, the GS offers a means of assessing post-operative pain that is as effective as manual behavioural-based scoring, without the limitations of such approaches.

Facial grimacing by mice after undergoing laparotomy surgery indicates postoperative pain lasts for 36 to 48 h (and at relatively high levels for 8 to 12 h) with relative exacerbation during the early dark (active) photo-phase. Furthermore, the grimacing indicates that buprenorphine is fully efficacious at recommended doses against early postoperative pain, but carprofen and ketoprofen are efficacious only at doses much higher than currently recommended: acetaminophen is not efficacious. [11]

A study in 2014 examined postoperative pain in mice following surgical induction of myocardial infarction. The effectiveness of the GS at identifying pain was compared with a traditional welfare scoring system based on behavioural, clinical and procedure-specific criteria. It was reported that post hoc GS (but not real-time GS) indicated a significant proportion of the mice were in low-level pain at 24 h which were not identified as such by traditional assessment methods. Importantly, those mice identified as experiencing low-level pain responded to analgesic treatment, indicating the traditional methods of welfare assessment were insensitive in this aspect of pain recognition. [1]

Mice with induced sickle cell disease and their controls exhibited a "pain face" when tested on a cold plate, but sickle mice showed increased intensity compared to controls; this was confirmed using Von Frey filaments a traditional method of pain assessment. [12] GS have also been used to assess pain and methods of its alleviation in pancreatitis. [13] GS have also been used to test the degree of pain caused as a side-effect of therapeutic drugs and methods of mitigating the pain. [14]

The mouse GS has been shown to be a highly accurate, repeatable and reliable means of assessing pain, requiring only a short period of training for the observer. [2] Assessment approaches that train deep neural networks to detect pain and no-pain images of mice may further speed up MGS scoring, with an accuracy of 94%. [15]

Sex and strain effects

It has been noted that DBA/2 strain mice, but not CBA strain mice, show an increase in GS score following only isoflurane anaesthesia, which should be taken into account when using the GS to assess pain. Administration of a common analgesic, buprenorphine, had no effect on the GS of either strain. [16]

There are interactions between the sex and strain of mice in their GS and also the method that is used to collect the data (i.e. real-time or post hoc), which indicates scorers need to consider these factors. [2]

Effects of non-painful procedures

It is important to establish whether methods of pain assessment in laboratory animals are influenced by other factors, especially those which are a normal part of routine procedures or husbandry. There is no difference in GS scores between mice handled using a tube compared with mice picked up by the tail, indicating these handling techniques are not confounding factors in GS assessment. [17] A similar study reported there was no difference between GS scores at baseline and immediately post ear notching (a method frequently used to identify laboratory mice), potentially indicating that the pain associated with ear notching is either too acute to assess using the GS tool or the practice is not painful. [18]

In rats

Closeup of the face of an agouti Russian blue rat Rat agouti russian blue.jpg
Closeup of the face of an agouti Russian blue rat

There are differences between the "pain face" of mice and rats. In mice, the nose and cheek at baseline have a smooth appearance, but in the presence of pain, change to distinct bulges in both the nose and cheek regions. By contrast, in rats at baseline, the nose and cheek regions show distinct bulging, and with pain, the bridge of the nose flattens and elongates, causing the whisker pads to flatten. As a consequence of these differences, the GS for rats sometimes use four FAU, i.e. Orbital Tightening, Nose/Cheek Flattening, Ear Changes and Whisker Changes. Nose/Cheek Flattening appears to show the highest correlation with the presence of pain in the rat. [3] [19]

GS for rats has been used to assess pain due to surgery, orthodontic tooth movement, osteoarthritis, acute chemotherapy-induced mucositis, and the efficacy of analgesics for these procedures and other painful conditions. [19] [20] [21] [22] [23] [24] [25] Furthermore, GS have been used to examine the effects of postoperative analgesia on the reduction of post-operative cognitive dysfunction in aged rats. [26]

As with mice, studies have examined the extent of agreement in assessing pain between rat GS and the use of von Frey filaments. Good agreement has been found between these [27] in relation to three models of pain (intraplantar carrageenan, intraplantar complete Freund's adjuvant and plantar incision). The GS score significantly increased in all pain models and the peak GS score also coincided with the development of paw hypersensitivity, although hypersensitivity persisted after GS scores returned to baseline. [28]

For rats, software (Rodent Face Finder) has been developed which successfully automates the most labour-intensive step in the process of quantifying the GS, i.e. frame-grabbing individual face-containing frames from digital video, which is hindered by animals not looking directly at the camera or poor images due to motion blurring. [29]

In rabbits

A GS for rabbits using four FAU, i.e. Orbital Tightening, Cheek Flattening, Nose Shape, Whisker Position (Ear Position is excluded from the analysis) has been developed (for exemplar images, see here ) and used to assess the effectiveness of an analgesic cream for rabbits having undergone ear-tattooing. [30] Similarly, a GS has been used to evaluate wellness in the post-procedural monitoring of rabbits. [31]

In horses

Based on the identification of FAU in rodents and rabbits, a GS for horses has been developed from post-operative (castration) individuals. This is based on six FAU, i.e. Stiffly Backwards Ears, Orbital Tightening, Tension Above the Eye Area, Prominent Strained Chewing Muscles, Mouth Strained and Pronounced Chin, Strained Nostrils and Flattening of the Profile (for exemplar images, see here.) [32] The HGS has thereafter been used to evaluate pain behavior in the laminitic horse, where it was concluded that the grimace scale can be used to assess the degree of pain also here, when compared to the Obel scale. [33]

A related study [34] describes the equine "pain face" after pain induction by a tourniquet on the antebrachium or topical capsaicin. The pain face here involves similar facial expressions described for the HGS; low and/or asymmetrical ears, an angled appearance of the eyes, a withdrawn and/or tense stare, medio-laterally dilated nostrils and tension of the lips, chin and certain mimetic muscles and can potentially be incorporated to improve existing pain evaluation tools. From the described pain face, The Equine Pain Scale has been developed. [35] Another pain scale has been described (EQUUS-FAP) which also has proven to assess acute pain in horses in a significant way. [36]

To map and explain the different facial expressions seen in the equine face during acute pain, an equine facial action coding system (EquiFACS) has been developed. Seventeen FAU have been identified and the involved anatomical structures behind each facial expression are explained and compared to facial expressions seen in other species. [37]

In cats

A preliminary study based on landmarks and distances between ears and in the muzzle demonstrated that observers shown facial images from painful and pain-free cats had difficulty in identifying pain-free from painful cats, with only 13% of observers being able to discriminate more than 80% of painful cats. Accuracy (based on a dichotomous judgement - pain or no pain) ranged from 18 to 94%. [38]

A complete GS (Feline Grimace Scale - FGS) for cats was published in 2019 to detect naturally-occurring acute pain. Five FAU were identified: ear position, orbital tightening, muzzle tension, whiskers change and head position. Each FAU receives a score from 0 to 2 and a total pain score is calculated as the sum of the FAU's scores divided by the total possible score excluding those AU marked as "not possible to score" (i.e. 4/10 = 0.4 or 4/8 = 0.5).  A training manual is available as "Supplementary information" within the original article. [39]

The FGS has been thoroughly validated and reported high discriminative ability, good overall inter-rater reliability, excellent intra-rater reliability, and excellent internal consistency. The FGS scores were higher in painful than in control cats; a very strong correlation with another validated instrument for pain assessment in cats was observed and the FGS detected response to analgesic treatment (scores after analgesia were lower than before). Additionally, an analgesic threshold was determined (total pain score >0.39 out of 1.0). The FGS is an easy and quick-to-use tool for acute pain assessment in cats. [39]

The clinical applicability of the FGS in cats undergoing ovariohysterectomy has been explored by comparing the scores assigned in real-time by an experienced observer with those scores assigned to still images, and good agreement has been reported. [40] The FGS is also a reliable tool for pain assessment in cats undergoing dental extractions and the caregiver's presence did not affect FGS scores. [41]

In sheep

A GS for sheep has been developed to detect pain caused by naturally occurring diseases such as footrot and mastitis. [42] A GS has been used to assess pain due to the routine husbandry procedure of tail-docking in lambs. There was high reliability between and within the observers, and high accuracy. Restraint of the lambs during the tail-docking caused changes in facial expression, which needs to be taken into account in use of the GS. [43]

In ferrets

Facial musculature of ferrets and compared lateral photographs of ferret faces were studied before and after intraperitoneal telemetry probe implantation. The FAU orbital tightening, nose bulging, cheek bulging, ear changes and whisker retraction were identified as potential indicators of pain in ferrets. All AU-scores assigned to the photographs taken five hours after surgery were significantly higher compared to their time-matched baseline scores. Further analysis using weights that were obtained using a Linear Discriminant Analysis revealed that scoring orbital tightening alone was sufficient to make this distinction with high sensitivity, specificity and accuracy. [44]

See also

Related Research Articles

A facial expression is one or more motions or positions of the muscles beneath the skin of the face. According to one set of controversial theories, these movements convey the emotional state of an individual to observers. Facial expressions are a form of nonverbal communication. They are a primary means of conveying social information between humans, but they also occur in most other mammals and some other animal species.

Lactucarium is the milky fluid secreted by several species of lettuce, especially Lactuca virosa, usually from the base of the stems. It is known as lettuce opium because of its sedative and analgesic properties. It has also been reported to promote a mild sensation of euphoria. Because it is a latex, lactucarium physically resembles opium, in that it is excreted as a white fluid and can be reduced to a thick smokable solid.

<span class="mw-page-title-main">Methylsulfonylmethane</span> Chemical compound

Dimethyl sulfone (DMSO2) is an organosulfur compound with the formula (CH3)2SO2. It is also known by several other names including methyl sulfone and (especially in alternative medicine) methylsulfonylmethane (MSM). This colorless solid features the sulfonyl functional group and is the simplest of the sulfones. It is relatively inert chemically and is able to resist decomposition at elevated temperatures. It occurs naturally in some primitive plants, is present in small amounts in many foods and beverages, and is marketed (under the MSM name) as a dietary supplement. It is sometimes used as a cutting agent for illicitly manufactured methamphetamine. It is also commonly found in the atmosphere above marine areas, where it is used as a carbon source by the airborne bacteria Afipia. Oxidation of dimethyl sulfoxide produces the sulfone, both under laboratory conditions and metabolically.

The dot-probe paradigm is a test used by cognitive psychologists to assess selective attention.

<span class="mw-page-title-main">Emotion in animals</span> Research into similarities between non-human and human emotions

Emotion is defined as any mental experience with high intensity and high hedonic content. The existence and nature of emotions in non-human animals are believed to be correlated with those of humans and to have evolved from the same mechanisms. Charles Darwin was one of the first scientists to write about the subject, and his observational approach has since developed into a more robust, hypothesis-driven, scientific approach. Cognitive bias tests and learned helplessness models have shown feelings of optimism and pessimism in a wide range of species, including rats, dogs, cats, rhesus macaques, sheep, chicks, starlings, pigs, and honeybees. Jaak Panksepp played a large role in the study of animal emotion, basing his research on the neurological aspect. Mentioning seven core emotional feelings reflected through a variety of neuro-dynamic limbic emotional action systems, including seeking, fear, rage, lust, care, panic and play. Through brain stimulation and pharmacological challenges, such emotional responses can be effectively monitored.

Opioid-induced hyperalgesia (OIH) or opioid-induced abnormal pain sensitivity, also called paradoxical hyperalgesia, is an uncommon condition of generalized pain caused by the long-term use of high dosages of opioids such as morphine, oxycodone, and methadone. OIH is not necessarily confined to the original affected site. This means that if the person was originally taking opioids due to lower back pain, when OIH appears, the person may experience pain in the entire body, instead of just in the lower back. Over time, individuals taking opioids can also develop an increasing sensitivity to noxious stimuli, even evolving a painful response to previously non-noxious stimuli (allodynia). This means that if the person originally felt pain from twisting or from sitting too long, the person might now additionally experience pain from a light touch or from raindrops falling on the skin.

<span class="mw-page-title-main">RB-101</span> Chemical compound

RB-101 is a drug that acts as an enkephalinase inhibitor, which is used in scientific research.

<span class="mw-page-title-main">Neramexane</span> Chemical compound

Neramexane is a drug related to memantine, which acts as an NMDA antagonist and has neuroprotective effects. It is being developed for various possible applications, including treatment of tinnitus, Alzheimer's disease, drug addiction and as an analgesic. Animal studies have also suggested antidepressant and nootropic actions, so there are a wide range of potential applications this drug may be used for. It also acts as a nicotinic acetylcholine receptor antagonist.

<span class="mw-page-title-main">Ro64-6198</span> Chemical compound

Ro64-6198 is a opioid drug used in scientific research. It acts as a potent and selective agonist for the nociceptin receptor, also known as the ORL-1 receptor, with over 100x selectivity over the other opioid receptors. It produces anxiolytic effects in animal studies equivalent to those of benzodiazepine drugs, but has no anticonvulsant effects and does not produce any overt effects on behaviour. However it does impair short-term memory, and counteracts stress-induced anorexia. It also has antitussive effects, and reduces the rewarding and analgesic effects of morphine, although it did not prevent the development of dependence. It has been shown to reduce alcohol self-administration in animals and suppressed relapses in animal models of alcoholism, and ORL-1 agonists may have application in the treatment of alcoholism.

<span class="mw-page-title-main">Pain in fish</span> Overview about the pain in fish

Fish fulfill several criteria proposed as indicating that non-human animals experience pain. These fulfilled criteria include a suitable nervous system and sensory receptors, opioid receptors and reduced responses to noxious stimuli when given analgesics and local anaesthetics, physiological changes to noxious stimuli, displaying protective motor reactions, exhibiting avoidance learning and making trade-offs between noxious stimulus avoidance and other motivational requirements.

<span class="mw-page-title-main">Pain in animals</span> Overview about pain in animals

Pain negatively affects the health and welfare of animals. "Pain" is defined by the International Association for the Study of Pain as "an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage." Only the animal experiencing the pain can know the pain's quality and intensity, and the degree of suffering. It is harder, if even possible, for an observer to know whether an emotional experience has occurred, especially if the sufferer cannot communicate. Therefore, this concept is often excluded in definitions of pain in animals, such as that provided by Zimmerman: "an aversive sensory experience caused by actual or potential injury that elicits protective motor and vegetative reactions, results in learned avoidance and may modify species-specific behaviour, including social behaviour." Nonhuman animals cannot report their feelings to language-using humans in the same manner as human communication, but observation of their behaviour provides a reasonable indication as to the extent of their pain. Just as with doctors and medics who sometimes share no common language with their patients, the indicators of pain can still be understood.

<span class="mw-page-title-main">Pain in crustaceans</span> Ethical debate

Pain in crustaceans is a scientific debate which questions whether they experience pain or not. Pain is a complex mental state, with a distinct perceptual quality but also associated with suffering, which is an emotional state. Because of this complexity, the presence of pain in an animal, or another human for that matter, cannot be determined unambiguously using observational methods, but the conclusion that animals experience pain is often inferred on the basis of likely presence of phenomenal consciousness which is deduced from comparative brain physiology as well as physical and behavioural reactions.

Palmitoylethanolamide (PEA) is an endogenous fatty acid amide, and lipid modulator PEA has been studied in in vitro and in vivo systems using exogenously added or dosed compound; there is evidence that it binds to a nuclear receptor, through which it exerts a variety of biological effects, some related to chronic inflammation and pain.

Pain is often regarded as the fifth vital sign in regard to healthcare because it is accepted now in healthcare that pain, like other vital signs, is an objective sensation rather than subjective. As a result nurses are trained and expected to assess pain.

<span class="mw-page-title-main">Tail flick test</span> Pain response test

The tail flick test is a test of the pain response in animals, similar to the hot plate test. It is used in basic pain research and to measure the effectiveness of analgesics, by observing the reaction to heat. It was first described by D'Amour and Smith in 1941.

Nutriepigenomics is the study of food nutrients and their effects on human health through epigenetic modifications. There is now considerable evidence that nutritional imbalances during gestation and lactation are linked to non-communicable diseases, such as obesity, cardiovascular disease, diabetes, hypertension, and cancer. If metabolic disturbances occur during critical time windows of development, the resulting epigenetic alterations can lead to permanent changes in tissue and organ structure or function and predispose individuals to disease.

Animal welfare science is the scientific study of the welfare of animals as pets, in zoos, laboratories, on farms and in the wild. Although animal welfare has been of great concern for many thousands of years in religion and culture, the investigation of animal welfare using rigorous scientific methods is a relatively recent development. The world's first Professor of Animal Welfare Science, Donald Broom, was appointed by Cambridge University (UK) in 1986.

<span class="mw-page-title-main">Pain in amphibians</span> Ethical issue

Pain is an aversive sensation and feeling associated with actual, or potential, tissue damage. It is widely accepted by a broad spectrum of scientists and philosophers that non-human animals can perceive pain, including pain in amphibians.

<span class="mw-page-title-main">Pain in cephalopods</span> Contentious issue

Pain in cephalopods is a contentious issue. Pain is a complex mental state, with a distinct perceptual quality but also associated with suffering, which is an emotional state. Because of this complexity, the presence of pain in non-human animals, or another human for that matter, cannot be determined unambiguously using observational methods, but the conclusion that animals experience pain is often inferred on the basis of likely presence of phenomenal consciousness which is deduced from comparative brain physiology as well as physical and behavioural reactions.

Jeffrey S. Mogil, FCAHS, FRSC is a Canadian neuroscientist and the E.P. Taylor Professor of Pain Studies and Canada Research Chair in the Genetics of Pain at McGill University. He is known for his work in the genetics of pain, for being among the first scientists to demonstrate sex differences in pain perception, and for identifying previously unknown factors and confounds that affect the integrity of contemporary pain research. He has an h-index of 90.

References

  1. 1 2 3 Faller, K.M., McAndrew, D.J., Schneider, J.E. and Lygate, C.A. (2015). "Refinement of analgesia following thoracotomy and experimental myocardial infarction using the Mouse Grimace Scale". Experimental Physiology. 100 (2): 164–172. doi:10.1113/expphysiol.2014.083139. PMC   4340041 . PMID   25480160.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  2. 1 2 3 Miller, A.L.; Leach, M.C. (2015). "The mouse grimace scale: a clinically useful tool?". PLOS ONE. 10 (9): e0136000. Bibcode:2015PLoSO..1036000M. doi: 10.1371/journal.pone.0136000 . PMC   4583436 . PMID   26406227.
  3. 1 2 Whittaker, A.L.; Howarth, G.S. (2014). "Use of spontaneous behaviour measures to assess pain in laboratory rats and mice: How are we progressing?". Applied Animal Behaviour Science. 151: 1–12. doi:10.1016/j.applanim.2013.11.001.
  4. Chambers, C.T.; Mogil, J.S. (2015). "Ontogeny and phylogeny of facial expression of pain" (PDF). Pain. 156 (5): 798–799. doi:10.1097/j.pain.0000000000000133. PMID   25887392. S2CID   2060896.
  5. van Rysewyk, S. (2016). "Nonverbal indicators of pain". Animal Sentience. 1 (3): 30.
  6. Darwin, Charles (1872). The Expression of the Emotions in Man and Animals by Charles Darwin. Cambridge Core (published 2013). doi:10.1017/cbo9781139833813. ISBN   9781139833813.
  7. Parr, L.A., Waller, B.M., Vick, S.J. and Bard, K.A. (2007). "Classifying chimpanzee facial expressions using muscle action". Emotion. 7 (1): 172–181. doi:10.1037/1528-3542.7.1.172. PMC   2826116 . PMID   17352572.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  8. Parr, L.A., Waller, B.M., Burrows, A.M., Gothard, K.M. and Vick, S.J. (2010). "Brief communication: MaqFACS: A muscle‐based facial movement coding system for the rhesus macaque". American Journal of Physical Anthropology. 143 (4): 625–630. doi:10.1002/ajpa.21401. PMC   2988871 . PMID   20872742.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  9. Langford, D.J., Bailey, A.L., Chanda, M.L., Clarke, S.E., Drummond, T.E., Echols, S., Glick, S., Ingrao, J., Klassen-Ross, T., Lacroix-Fralish, M.L., Matsumiya, L., Sorge, R.E., Sotocinal, S.G., Tabaka, J.M., Wong, D., van den Maagdenberg, A.M., Ferrari, M.D., Craig, K.D. and Mogil, J.S. (2010). "coding of facial expressions of pain in the laboratory mouse". Nature Methods. 7 (6): 447–449. doi:10.1038/nmeth.1455. PMID   20453868. S2CID   16703705.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  10. Flecknell, P.A. (2010). "Do mice have a pain face?" (PDF). Nature Methods. 7 (6): 437–438. doi:10.1038/nmeth0610-437. PMID   20508638. S2CID   2944510.
  11. Matsumiya, L.C., Sorge, R.E., Sotocinal, S.G., Tabaka, J.M., Wieskopf, J.S., Zaloum, A., ... & Mogil, J.S. (2012). "Using the Mouse Grimace Scale to reevaluate the efficacy of postoperative analgesics in laboratory mice". Journal of the American Association for Laboratory Animal Science. 51 (1): 42–9. PMC   3276965 . PMID   22330867.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  12. Mittal, A.M., Lamarre, Y.Y. and Gupta, K. (2014). "Observer based objective pain quantification in sickle mice using grimace scoring and body parameters" (PDF). Blood. 124 (21): 4907. doi:10.1182/blood.V124.21.4907.4907. Archived from the original (PDF) on 2016-08-15. Retrieved 2016-01-12.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  13. Jurik, A., Ressle, A., Schmid, R.M., Wotjak, C.T. and Thoeringer, C.K. (2014). "Supraspinal TRPV1 modulates the emotional expression of abdominal pain" (PDF). Pain. 155 (10): 2153–2160. doi:10.1016/j.pain.2014.08.012. PMID   25139591. S2CID   3196042.{{cite journal}}: CS1 maint: multiple names: authors list (link)[ permanent dead link ]
  14. Melemedjian, O.K., Khoutorsky, A., Sorge, R.E., Yan, J., Asiedu, M.N., Valdez, A., ... & Price, T.J. (2013). "mTORC1 inhibition induces pain via IRS-1-dependent feedback activation of ERK". Pain. 154 (7): 1080–1091. doi:10.1016/j.pain.2013.03.021. PMC   3742001 . PMID   23607966.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  15. Tuttle, Alexander H; Molinaro, Mark J; Jethwa, Jasmine F; Sotocinal, Susana G; Prieto, Juan C; Styner, Martin A; Mogil, Jeffrey S; Zylka, Mark J (January 2018). "A deep neural network to assess spontaneous pain from mouse facial expressions". Molecular Pain. 14: 174480691876365. doi:10.1177/1744806918763658. ISSN   1744-8069. PMC   5858615 . PMID   29546805.
  16. Miller, A., Kitson, G., Skalkoyannis, B. and Leach, M. (2015). "The effect of isoflurane anaesthesia and buprenorphine on the mouse grimace scale and behaviour in CBA and DBA/2 mice". Applied Animal Behaviour Science. 172 (58–62): 58–62. doi:10.1016/j.applanim.2015.08.038. PMC   4768077 . PMID   26937061.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  17. Miller, A.L.; Leach, M.C. (2015). "The effect of handling method on the mouse grimace scale in two strains of laboratory mice". Laboratory Animals. 50 (4): 305–307. doi:10.1177/0023677215622144. PMC   4976650 . PMID   26657061.
  18. Miller, A.L.; Leach, M.C. (2014). "Using the mouse grimace scale to assess pain associated with routine ear notching and the effect of analgesia in laboratory mice". Laboratory Animals. 49 (2): 117–120. doi:10.1177/0023677214559084. PMID   25378137. S2CID   10093354.
  19. 1 2 Sotocinal, S.G., Sorge, R E., Zaloum, A., Tuttle, A.H., Martin, L.J., Wieskopf, J.S., ... & McDougall, J.J. (2011). "The Rat Grimace Scale: a partially automated method for quantifying pain in the laboratory rat via facial expressions" (PDF). Molecular Pain. 7 (1): 1744–8069–7–55. doi: 10.1186/1744-8069-7-55 . PMC   3163602 . PMID   21801409.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  20. Chi, H., Kawano, T., Tamura, T., Iwata, H., Takahashi, Y., Eguchi, S., ... & Yokoyama, M. (2013). "Postoperative pain impairs subsequent performance on a spatial memory task via effects on N-methyl-D-aspartate receptor in aged rats". Life Sciences. 93 (25): 986–993. doi:10.1016/j.lfs.2013.10.028. PMID   24211778.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  21. Liao, L., Long, H., Zhang, L., Chen, H., Zhou, Y., Ye, N. and Lai, W. (2014). "Evaluation of pain in rats through facial expression following experimental tooth movement". European Journal of Oral Sciences. 122 (2): 121–124. doi:10.1111/eos.12110. PMID   24428464.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  22. Long, H., Liao, L., Gao, M., Ma, W., Zhou, Y., Jian, F., ... & Lai, W. (2015). "Periodontal CGRP contributes to orofacial pain following experimental tooth movement in rats". Neuropeptides. 52: 31–37. doi:10.1016/j.npep.2015.06.006. PMID   26164378. S2CID   804877.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  23. Davis, M.E. (2014). The Effect Of Sumatriptan On Clinically Relevant Behavioral Endpoints In A Recurrent Nitroglycerin Migraine Model In Rats (PDF) (Thesis). The University of Mississippi.
  24. Sperry, Megan M.; Yu, Ya-Hsin; Welch, Rachel L.; Granquist, Eric J.; Winkelstein, Beth A. (2018-09-17). "Grading facial expression is a sensitive means to detect grimace differences in orofacial pain in a rat model". Scientific Reports. 8 (1): 13894. Bibcode:2018NatSR...813894S. doi:10.1038/s41598-018-32297-2. ISSN   2045-2322. PMC   6141616 . PMID   30224708.
  25. Whittaker, A L; Leach, M C; Preston, F L; Lymn, K A; Howarth, G S (2015-07-10). "Effects of acute chemotherapy-induced mucositis on spontaneous behaviour and the grimace scale in laboratory rats". Laboratory Animals. 50 (2): 108–118. doi:10.1177/0023677215595554. ISSN   0023-6772. PMID   26162377. S2CID   8823849.
  26. Kawano, T., Takahashi, T., Iwata, H., Morikawa, A., Imori, S., Waki, S., ... & Yokoyama, M. (2014). "Effects of ketoprofen for prevention of postoperative cognitive dysfunction in aged rats". Journal of Anesthesia. 28 (6): 932–936. doi:10.1007/s00540-014-1821-y. PMID   24676769. S2CID   25524002.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  27. De Rantere, D. (2014). The Evaluation of the Rat Grimace Scale and Ultrasonic Vocalisations as Novel Pain Assessment Tools in Laboratory Rats (PDF) (Thesis). University of Calgary.
  28. De Rantere, D., Schuster, C.J., Reimer, J.N. and Pang, D.S.J. (2015). "The relationship between the Rat Grimace Scale and mechanical hypersensitivity testing in three experimental pain models". European Journal of Pain. 20 (3): 417–426. doi:10.1002/ejp.742. PMID   26132473. S2CID   205795719.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  29. Oliver, V., De Rantere, D., Ritchie, R., Chisholm, J., Hecker, K.G. and Pang, D.S. (2014). "Psychometric assessment of the Rat Grimace Scale and development of an analgesic intervention score". PLOS ONE. 9 (5): e97882. Bibcode:2014PLoSO...997882O. doi: 10.1371/journal.pone.0097882 . PMC   4024023 . PMID   24838111.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  30. Keating, S.C., Thomas, A.A., Flecknell, P.A. and Leach, M.C. (2012). "Evaluation of EMLA cream for preventing pain during tattooing of rabbits: changes in physiological, behavioural and facial expression responses". PLOS ONE. 7 (9): e44437. Bibcode:2012PLoSO...744437K. doi: 10.1371/journal.pone.0044437 . PMC   3436883 . PMID   22970216.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  31. Hampshire, V.; Robertson, S. (2015). "Using the facial grimace scale to evaluate rabbit wellness in post-procedural monitoring". Lab Animal. 44 (7): 259–260. doi: 10.1038/laban.806 . PMID   26091129.
  32. Dalla Costa, E., Minero, M., Lebelt, D., Stucke, D., Canali, E. and Leach, M.C. (2014). "Development of the Horse Grimace Scale (HGS) as a pain assessment tool in horses undergoing routine castration". PLOS ONE. 9 (3): e92281. Bibcode:2014PLoSO...992281D. doi: 10.1371/journal.pone.0092281 . PMC   3960217 . PMID   24647606.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  33. Dalla Costa, Emanuela; Stucke, Diana; Dai, Francesca; Minero, Michela; Leach, Matthew; Lebelt, Dirk (3 August 2016). "Using the Horse Grimace Scale (HGS) to Assess Pain Associated with Acute Laminitis in Horses (Equus caballus)". Animals. 6 (8): 47. doi: 10.3390/ani6080047 . PMC   4997272 . PMID   27527224.
  34. Gleerup, K.B., Forkman, B., Lindegaard, C. and Andersen, P.H. (2015). "An equine pain face". Veterinary Anaesthesia and Analgesia. 42 (1): 103–114. doi:10.1111/vaa.12212. PMC   4312484 . PMID   25082060.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  35. Gleerup, K. B.; Lindegaard, C. (January 2016). "Recognition and quantification of pain in horses: A tutorial review". Equine Veterinary Education. 28 (1): 47–57. doi: 10.1111/eve.12383 .
  36. van Loon, Johannes P.A.M.; Van Dierendonck, Machteld C. (February 2017). "Monitoring equine head-related pain with the Equine Utrecht University scale for facial assessment of pain (EQUUS-FAP)". The Veterinary Journal. 220: 88–90. doi:10.1016/j.tvjl.2017.01.006. PMID   28190503.
  37. Wathan, Jen; Burrows, Anne M.; Waller, Bridget M.; McComb, Karen; Hillmann, Edna (5 August 2015). "EquiFACS: The Equine Facial Action Coding System". PLOS ONE. 10 (8): e0131738. Bibcode:2015PLoSO..1031738W. doi: 10.1371/journal.pone.0131738 . PMC   4526551 . PMID   26244573.
  38. Holden, E.; Calvo, G.; Collins, M.; Bell, A.; Reid, J.; Scott, E. M.; Nolan, A. M. (2014). "Evaluation of facial expression in acute pain in cats". Journal of Small Animal Practice. 55 (12): 615–621. doi:10.1111/jsap.12283. ISSN   1748-5827. PMID   25354833.
  39. 1 2 Evangelista, Marina C.; Watanabe, Ryota; Leung, Vivian S. Y.; Monteiro, Beatriz P.; O’Toole, Elizabeth; Pang, Daniel S. J.; Steagall, Paulo V. (2019-12-13). "Facial expressions of pain in cats: the development and validation of a Feline Grimace Scale". Scientific Reports. 9 (1): 19128. Bibcode:2019NatSR...919128E. doi: 10.1038/s41598-019-55693-8 . ISSN   2045-2322. PMC   6911058 . PMID   31836868.
  40. Evangelista, Marina C.; Benito, Javier; Monteiro, Beatriz P.; Watanabe, Ryota; Doodnaught, Graeme M.; Pang, Daniel S. J.; Steagall, Paulo V. (2020-04-14). "Clinical applicability of the Feline Grimace Scale: real-time versus image scoring and the influence of sedation and surgery". PeerJ. 8: e8967. doi: 10.7717/peerj.8967 . ISSN   2167-8359. PMC   7164424 . PMID   32322445.
  41. Watanabe, Ryota; Doodnaught, Graeme M.; Evangelista, Marina C.; Monteiro, Beatriz P.; Ruel, Hélène L. M.; Steagall, Paulo V. (2020). "Inter-Rater Reliability of the Feline Grimace Scale in Cats Undergoing Dental Extractions". Frontiers in Veterinary Science. 7: 302. doi: 10.3389/fvets.2020.00302 . ISSN   2297-1769. PMC   7272704 . PMID   32548134.
  42. McLennan, K.M., Rebelo, C.J., Corke, M.J., Holmes, M.A., Leach, M.C. and Constantino-Casas, F. (2016). "Development of a facial expression scale using footrot and mastitis as models of pain in sheep". Applied Animal Behaviour Science. 176: 19–26. doi:10.1016/j.applanim.2016.01.007. hdl: 10034/600536 .{{cite journal}}: CS1 maint: multiple names: authors list (link)
  43. Guesgen, M.; Leach, M. (2014). "Assessing pain using the lamb grimace scale (LGS)" (PDF). RSPCA. Retrieved January 11, 2015.
  44. Reijgwart, Marsinah L.; Schoemaker, Nico J.; Pascuzzo, Riccardo; Leach, Matthew C.; Stodel, Melanie; de Nies, Loes; Hendriksen, Coenraad F. M.; van der Meer, Miriam; Vinke, Claudia M. (2017-11-13). Staffieri, Francesco (ed.). "The composition and initial evaluation of a grimace scale in ferrets after surgical implantation of a telemetry probe". PLOS ONE. 12 (11): e0187986. Bibcode:2017PLoSO..1287986R. doi: 10.1371/journal.pone.0187986 . ISSN   1932-6203. PMC   5683639 . PMID   29131858. CC-BY icon.svg Material was copied from this source, which is available under a Creative Commons Attribution 4.0 International License.