Skip to content
Publicly Available Published by De Gruyter April 1, 2012

Translational animal models using veterinary patients – An example of canine osteoarthritis (OA)

  • Outi Vainio

Abstract

Background and purpose

The use of laboratory animals in pain research has powerfully contributed to our detailed understanding of the physiological mechanisms of pain. Animal models also represent an essential tool to screen and select novel drug molecules with potentially analgesic properties. Despite of the inevitable input of laboratory animal trials, recent studies have shown that animal pain models have repeatedly failed to predict clinical analgesic efficacy and adverse side effects of potential drug molecules in human pain patients. This paper provides a review of the laboratory animal models of OA, which have been developed to test efficacy of novel analgesics. The paper also presents spontaneous OA in canine veterinary patients, and methods to observe chronic pain in nonverbal dogs.

Methods

PubMed data base was searched as a reference list to locate most relevant articles. A number of 118 articles including 4 reviews were located. Web pages of 4 establishments and 2 private organizations were also accessed.

Results

The clinical expression and pathogenesis of naturally occurring OA in dogs is considered an analogous disease that occurs in humans, including pain and lameness. OA may occur in any joint in dogs as well as in humans. Primary idiopathic OA in dogs is rare, but certain breeds may be predisposed to it. For the most part, canine OA is considered secondary to acquired or congenital musculoskeletal disorders. Concomitant factors, such as aging and obesity, likely accelerate progression. However, mechanical factors appear to predominate in the etiopathogenesis of canine spontaneous OA. Both subjective (validated questionnaire) and objective (gait analysis) tools are available to measure OA related pain in dogs. Information on the prevalence of canine OA is limited, but rough surveys suggest that 11 million dogs in the United States and 5 million in Europe could suffer from OA. Ethical considerations concerning the use of privately owned dogs can be resolved by a careful experimental design.

Conclusion

Canine spontaneous OA could serve as a translational animal model that would more closely mimick clinical OA related pain conditions in humans. Privately owned dogs would make a solution to fix the gap between animal pain models and clinical trials when testing potential analgesic drug molecules. Close interdisciplinary cooperation would guarantee that both scientific and ethical intentions would be achieved.

Implications

The predictability of translational pain research would improve by using privately owned dogs as chronic pain models when testing novel analgesics.

1 Introduction

The use of laboratory animals in pain research has powerfully contributed to our detailed understanding of the physiological mechanisms of pain [1], including the initiation, maintenance and termination of a painful signal. In addition, animal models represent an essential tool to screen and select novel drug molecules with potentially analgesic properties.

2 Experiments used to test analgesics in laboratory animals

Testing the potency of novel analgesic drug molecules in laboratory animals has involved both acute and chronic painful stimuli. In a classical acute pain assay, a noxious stimulus is applied to an extreme body part where a simple withdrawal response can be detected and scored [2]. Both the latency to the response and the response threshold are recorded in these trials. Distinguishing between unconscious spinal withdrawal reflexes [2] and conscious motor responses that involve supraspinal affective components [3] is vital when recording responses or their latencies. In inflammatory pain experiments, the noxious stimulus is induced by an algogenic chemical, such as formalin [4], carrageenan [5] and Freund’s adjuvant [5], which are usually administered either subcutaneously or intraperitoneally [2]. Mediator-induced pain develops slowly but endures longer. Instead of an avoidance response, the affected animal expresses painful sensations by engaging in specific pain-related behavior which is associated with both the location of the discomfort and the species in question. Neuropathic pain models are frequently provoked by surgical constriction of a peripheral nerve lesion. Pain behaviors in neuropathic assays vary, the ultimate being self-mutilation [6,7].

When it became evident that simple experimental models were not analogous enough to complex clinical pain syndromes, researchers introduced complete disease models, such as cancer pain [8], post-operative pain [9,10,11], and surgically induced OA pain [12,13].

3 Challenges of translational pain research in laboratory animals

Despite of the inevitable input of animal trials in pain research, recent studies have shown that animal pain models have repeatedly failed to predict clinical analgesic efficacy and adverse side effects of potential drug molecules in human pain patients [1,14]. Several comprehensive reviews have covered the less satisfactory face validity of animal pain models [15,16,17,18,19,20], referring to the fact that the resemblance of symptoms of the pain tests are irrelevant to the clinical condition. Developing animal models that more closely mimick clinical pain conditions has become important in translational pain research [14,16].

4 OA models in laboratory animals

The term OA does not describe a single elusive disease, but rather it refers to a number of related and overlapping disorders where joint affliction and disability are prominent symptoms. In 1995, a consensus workshop defined OA diseases as consequences of both mechanical and biologic events that destabilize the normal coupling of degradation and synthesis of articular cartilage chondrocytes, the extracellular matrix, and subchondral bone [21]. The etiology of OA is largely unknown, but is most likely multifactorial [22]. In patients, OA diseases result in a common joint pathology [12] characterized by degeneration of the articular cartilage [23]; other affected articular tissues may also include subchondral bone, synovial fluid, synovial membranes, and surrounding periarticular soft tissues [22,24]. Because arthritis or OA is the most frequent musculoskeletal disorder [25,26], a variety of either induced or naturally occurring animal models of OA [27] have been developed to test the therapeutic potency of drugs [28].

Induced models in mice [29], rats [12,13,30], guinea pigs [31], rabbits [28,32], cats [33], dogs [34,35,36], sheep [37], and goats [23] focus mainly on the knee joint. Impaired functioning of the knee joint has been induced by surgical intervention [12,33,34,37], by chemically injecting an irritant into the joint [12,38], by blunt trauma [32], or by mechanical load [29,31,39], thus leading to an adaptive response which modifies the articular cartilage structure and contributes to OA. Because the knee joint is the most common target joint affected by OA in humans [22] this may have favored the preference of knee joint models. In addition, the knee joint is of considerable size and is easy to access, thus rendering it suitable for a model. Other joints have occasionally been added. Simmons et al. [40] introduced an OA model for the equine metacarpopha-langeal joint, and genetically modified mouse models of OA are also currently available [41,42].

All mammals can develop degenerative joint diseases [43]. Naturally occurring OA has been described in mice [44,45,46], broiler chickens [47], guinea pigs [48,49,50], Syrian hamsters [51], non-human primates [52,53], aging cats [54,55] and dogs [55,56]. Salo et al. [30] found that chemically induced selective joint denervation in 2-month-old rats initiated a normal-like developmental process resulting in OA in the knee joint. The authors proposed that the loss of neurons with age may have contributed to a spontaneous developmental process, and therefore classified their OA model as normally occurring.

On the basis of insufficient data about the pathogenesis of OA pain, developing experimental models has been difficult [57]. The relevance of most animal models of OA is for the most part based on histopathological similarities to human disease [28]. This approach ignores differences in the speed of the degenerative process between an induced model and a naturally occurring disease. Most surgically induced models of OA have rapid and strict cartilage degeneration after manipulation. Whether these models can explain the pain mechanism of slowly developing clinical OA remains uncertain [57]. A swift development period also implies that fewer opportunities for experimental therapeutic assays are available. Spontaneous models, such as those seen in certain laboratory species and in privately owned dogs and cats, should offer a better opportunity to study the slowly progressive process that is characteristic to human OA [28,58], including the track records of drug-induced modification of disease progression. When Mao [16] recalled animal models mimicking clinical pain conditions, the author pushed for dialogues between researchers and clinical practitioners. For unspecified reasons, veterinarians with their animal patients were excluded from the call for discourse.

5 Canine spontaneous OA

Both the clinical expression and pathogenesis of naturally occurring canine hip dysplasia and OA are considered analogous diseases that occur in humans. Progressive and degenerative canine OA causes notable signs of pain, such as lameness and physical disability [56,59], rendering affected dogs reluctant to perform normal activities [60]. Pain and lameness may be acute or chronic [60,61], and OA may occur in any joint in dogs [55] as well as in humans [22].

Primary idiopathic OA in dogs is rare [61,62], but certain breeds maybe predisposed to it [60]. For the most part, canine OA is considered secondary to acquired or congenital musculoskeletal disorders [62]. Concomitant factors, such as aging [63] and obesity [64], likely accelerate progression. However, mechanical factors appear to predominate in the etiopathogenesis of canine spontaneous OA [58,63].

A diagnosis of OA is based on clinical signs, physical examinations, radiographs, and synovial fluid analyses [61]. The approach to treating canine OA will vary based on its severity and location [55]. Management of OA involves a lifelong multimodal approach which aims primarily to alleviate pain and secondly to improve mobility [59], including activity control, weight management, physical therapy, nutritional support, and nutraceuticals [55,61]. Non-steroidal anti-inflammatory drugs (NSAIDs) are the dominant medical intervention for canine OA-related symptoms [59], and the scientific literature provides strong supportive evidence for the use of certain NSAIDs [65]. In the United States, NSAIDs have the largest number of reported adverse effects in companion animals [66]. In Sweden, canine specific NSAIDs came on market in the late 1990s. Thereafter statistical survival rates of Swedish aging canines have increased [67].

Humans possess genetic risk factors that influence their risk or hip OA, and which may also affect their outcomes of OA [68], see also the review by Valdes and Spector [69]. In medium-and large-size dog breeds, recent evidence has shown that hip and elbow dysplasia-related OA is associated with polygenetically inherited developmental abnormalities [70,71,72]. In a current paper [73], four hip dysplasia-associated and two OA-associated single-nucleotide polymorphisms (SNPs) and nearby candidate genes were identified in dogs. The SNPs identified included those near known genes reportedly associated with, or expressed in, OA in humans [74,75]. In addition to pre-clinical drug testing, a canine model could provide an opportunity to identify more potential genes underlying natural OA in humans.

6 Translational canine model of spontaneous OA

In clinical trials, most measures of treatment efficacy involve patient-reported outcomes [76]. In non-verbal dogs, this is unattainable, so recognition of treatment response is a challenge. In veterinary medicine, the owner of the animal or a veterinarian or both detect and report the management response. Measures of OA-related outcome in dogs have been validated in blinded, randomized, and placebo-controled trials [77,78]. The Helsinki chronic pain index developed by Hielm-Bjorkman et al. [79] was certified using veterinarians and owners to assess signs of pain in dogs. The process resulted in the potential use of 11 multi-factorial behavior- and locomotion-related questions in the assessing chronic pain in dogs [80]. A further study [81] concluded that pain-naive owners did not perceive signs of chronic pain in their dogs, but the authors suggested that the owners could learn the detection of signs of pain through training. That said, the dog owners could nevertheless detect pain diminish and return after starting and discontinuing NSAIDs, respectively. The canine brief pain inventory [77] used the subjective assessment of efficacy of treatments administered by a trained owner who completed a questionnaire both before and after the treatment period. The questionnaire was tested in a double-blind, randomized, placebo-controlled trial and was able to detect improvements in pain scores in dogs with OA treated with NSAID or placebo. A client-based clinical metrology instrument was initially validated for the evaluation of canine OA [78]. Validation was based on a prospective cohort study where the dog owners completed a questionnaire before a gait analysis on a force platform, which served as an external standard measure. The authors concluded that the instrument was worthy of continued investigation.

Gait analysis, measurement of ground reaction forces of each leg using a force plate, provides a quantitative description of quadrupedal gait [82]. Gait analysis provides a non-invasive assessment of lameness beyond subjective evaluation [83]. When necessary, an intra-articular anesthetic injection could serve to decouple gait mechanics from pain originating in other organs, such as the muscles or skeleton [84]. Several studies have established the use of force platform in gait analysis in healthy dogs of various breeds [83,85,86,87,88,89] or of a certain breed [90], including comparative gait analysis of two breeds [91,92]. The trotting gait was more sensitive than the walking gait for differentiation of dogs with low-grade hind limb lameness [93].

Gait analysis successfully served to detect acute pain in dogs; pain was provoked by synovitis which was induced by intra-articularly injected sodium urate crystals [94], or surgical intervention [82]. Effect of chronic pain on gait was distinguished in dogs with surgically induced hindlimb lameness [95]. Gait analysis was also used for detecting the effect of exercise in dogs with naturally occurring hindlimb OA [96], as well as for evaluating effect of surgical technique prior to and after reconstructive surgery on experimental dogs [97], and on veterinary orthopedic patients [98,99]. The method was also used to confirm the postoperative efficacy of NSAIDs in privately owned dogs which underwent a reconstructive cruciate surgery [100]. A three-dimensional kinematic canine hind limb model was recently created by adapting techniques and algorithms developed for humans [101].

Information on the prevalence of canine OA is limited [63], but a rough survey carried out in the Unites States suggested that approximately 20% of the canine population over 1 year of age suffer from OA [56]. Quessy [102] found that 30% of all dogs suffered from OA but the source of the information was not specified. In certain canine breeds, the prevalence of hip dysplasia ranged from 41% to 73% [103]. In 2007, the American Veterinary Medical Association announced on its home page [104] that there are 72 million dogs in the United States. If one-fifth of them are younger than 1 year, then 11 million dogs in the United States could suffer from OA. This number includes predisposition to OA in any joint. The Federation Cynologique Internationale claims on its web page [105] that in 2009, approximately 35 million dogs lived in Europe. If the prevalence of OA in Europe is the same as that in the United States, then more than 5 million European dogs would be affected by OA.

7 Ethical considerations

The use of laboratory animals in biomedical research has been questioned. Sensitive societies have enlisted public opinion in support of legislation for the protection of animals in biomedical research. In 2010, the European Union adopted a new directive [106] to update the 1986 directive on the protection of animals used for scientific purposes. In the United States, the Health Research Extension Act of 1985 [107] provides guidelines for animal use. The aim to reduce the number of animals used in science is incorporated in both laws. The acceptance of privately owned animals as translational models would support this aim.

Pet dogs as scientific objects will per se foreground ethical inquiries. It is unacceptable to cause detriment or to risk the welfare of privately owned animals. However, potential analgesics intended for human use could be tested on pet dogs in the same way as novel veterinary medicines. The European Medicines Agency has assigned rules governing medicinal products in the European Union, including veterinary products [108]; the United States Food and Drug Administration has similar regulations [109] for the development of veterinary products.

As there is a need to fill the gap between basic science and clinical implications [110,111,112], veterinary patients suffering from naturally occurring OA could provide the missing linkage. In an optimal case, the potential test medicine would be appropriate for human and animal consumption.

Scientific proof of the efficacy of an experimental medicine in canines may request inclusion of a placebo group. This demand could prove problematic but a careful experimental design could include a placebo group if a rescue analgesic could be offered in the event signs of unacceptable pain were observed in the dogs involved. There is no published information on the attitudes of animal owners toward medical trials performed on their pets, but such owners are aware that debilitating OA may lead to untimely euthanasia of their pet [113,114]. Anecdotally, decent and caring owners are known to be favorable to bringing their pets to clinical trials, which may promise the alleviation of pain for their four-legged family member.

Pain research carried out on animals mostly lacks discussion about the affective modality of pain. The description of pain by the International Association for the Study of Pain (IASP) Subcommittee on Taxonomy [115] relates to pain in humans. The IASP special interest group for the study of pain in animals is designated to pain in non-human species [116]. In veterinary medicine pain is not precisely defined. The American College of Veterinary Anesthesiologists has aligned itself with a declaration that an individual animal may or may not experience pain in response to nociception [117]; and further, that it is difficult to compare the experience of pain in animals to that in people. Molony and Kent [118] used the following working definition: animal pain is an aversive sensory and emotional experience representing an awareness by the animal of damage or threat to the integrity of its tissues.

The research community does not share exclusive scientific evidence of the affective modality of non-human pain. However, there is still less evidence of its absence, at least in mammals. In animals, affective dimension of pain sense most probably differs from that of humans. Although different, some kind of affective component of non-human pain would add a new element to the late-phase preclinical testing of medical candidates. Poole et al. [84] suggested that the emotional component of pain should be considered in pain assessment in preclinical laboratory animal models in the treatment studies of OA. The authors remind that the affective modality of OA pain in laboratory animals has no standardized clinical tests but they indicated that such a test should be developed using behavioral output. For pet dogs suffering from naturally occurring OA, validated methods of pain recognition do exist, including subjective (dog owner, veterinarian) [77,78,79,80,81] and objective (vertical force analysis) measures [82,83] but the methods do not discriminate the emotional component of pain. In clinical trials carried out on veterinary patients, both sensory-discriminative and affective-motivational modalities of pain could be distinguished in a careful experimental design where behavioral methods would be combined to neurophysiological tools, such as electroencephalographic recording [119]. Dogs have demonstrated special social and cognitive skills [120,121] that have developed through the domestication process. The close coevolution of dogs with humans has improved the interspecies communicative abilities of dogs [122,123,124], which would facilitate the objective detection of affective modality of pain in this special non-human species.

8 Conclusions

When improving the predictability of translational pain research, canine veterinary patients offer benefits over other models. Firstly, trials in animals with naturally slowly progressive and degenerative OA would increase the predictability of the model, and could therefore improve the predictive veracity for drug candidate selection. Secondly, in addition to their special coevolutionary character, dogs share both their living environment and way of life with humans, which make them the closest animal model to humans. Thirdly, there are more than 5 million OA-defected dogs in Europe and 11 million in the United States, thus ensuring that the number of pet dogs available is sufficient to perform controlled blinded randomized clinical trials within a reasonable time schedule. Fourthly, the inclusion of privately owned dogs in analgesic drug development programs would be a win-win situation for both parties.

It is difficult to find any serious objection to why we should not expand animal trials to include privately owned animals. Can we afford to refuse the use of pet dogs as a model in translational pain research?


DOI of refers to article: 10.1016/j.sjpain.2012.01.001.



Tel.: +358 9 19157316; fax: +358 9 19157300.

References

[1] Langley CK, Aziz Q, Bountra C, Gordon N, Hawkins P, Jones A, Langley G, Nurmikko T, Tracey I. Volunteer studies in pain research – opportunities and challenges to replace animal experiments. The report and recommendations of a focus on alternatives workshop. NeuroImage 2008;42:467–73.Search in Google Scholar

[2] Le Bars D, Gozariu M, Cadden SW. Animal models of nociception. Pharmacol Rev 2001;53:597–652.Search in Google Scholar

[3] Braz JM, Nassar MA, Wood JN, Basbaum AI. Parallel “pain” pathways arise from subpopulations of primary afferent nociceptor. Neuron 2005;47:787–93.Search in Google Scholar

[4] Dubuisson D, Dennis SG. The formalin test: a quantitative study of the analgesic effects of morphine, meperidine, and brain stem stimulation in rats and cats. Pain 1997;4:161–74.Search in Google Scholar

[5] Iadarola MJ, Brady LS, Draisci G, Dubner R. Enhancement of dynorphin gene expression in spinal cord following experimental inflammation: stimulus specificity, behavioural parameters and opioid receptor binding. Pain 1988;35:313–26.Search in Google Scholar

[6] Wall PD, Devor M, Inbal R, Scadding JW, Schonfeld D, Selzer Z, Tomkiewicz MM. Autotomy following peripheral nerve lesions: experimental anaesthesia dolorosa. Pain 1979;7:103–13.Search in Google Scholar

[7] Kauppila T. Correlation between autotomy-behaviour and current theories of neuropathic pain. Neurosci Biobehav Rev 1998;23:111–29.Search in Google Scholar

[8] Pacharinsak C, Beitz A. Animal models of cancer pain. Comp Med 2008;58:220–33.Search in Google Scholar

[9] Brennan TJ, Vandermeulen EP, Gebhart GF. Characterization of a rat model of incisional pain. Pain 1996;64:493–501.Search in Google Scholar

[10] Gonzalez MI, Field MJ, Bramwell S, McCleary S, Singh L. Ovarhysterectomy in the rat: a model of surgical pain for evaluation of pre-emptive analgesia? Pain 2000;88:79–88.Search in Google Scholar

[11] Martin TJ, Buechler NL, Kahn W, Crews JC, Eisenach JC. Effects of laparotomy on spontaneous exploratory activity and conditioned operant responding in the rat – a model for postoperative pain. Anesthesiology 2004;101:191–203.Search in Google Scholar

[12] Fernihough J, Gentry C, Malcangio M, Fox A, Rediske J, Pellas T, Kidd B, Bevan S, Winter J. Pain related behaviour in two models of osteoarthritis in the rat knee. Pain 2004;112:83–93.Search in Google Scholar

[13] Bove SE, Laemont KD, Brooker RM, Osborn MN, Sanchez BM, Guzman RE, Hook KE, Juneau PL, Connor JR, Kilgore KS. Surgically induced osteoarthritis in the rat results in the development of both osteoarthritis-like joint pain and secondary hyperalgesia. Osteoarthr Cartilage 2006;14:1041–8.Search in Google Scholar

[14] Kola I, Landis J. Can pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov 2004;3:711–5.Search in Google Scholar

[15] Vierck CJ, Hansson PT, Yezierski RP. Clinical and pre-clinical pain assessing: are we measuring the same thing? Pain 2008;135:7–10.Search in Google Scholar

[16] Mao J. Translational pain research: achievements and challenges. J Pain 2009;10:1001–11.Search in Google Scholar

[17] Mogil JS. Animal models of pain: progress and challenges. Nat Rev Neurosci 2009;10:283–94.Search in Google Scholar

[18] Rice ASC, Cimino-Brown D, Eisenach JC, Kontinen VK, Lacroix-Fralish ML, Machin I and (on behalf of the Preclinical Pain Consortium), Mogil JS, Stöhr T. Animal models and the prediction of efficacy in clinical trials of analgesic drugs: a critical appraisal and call for uniform reporting standards. Pain 2009;139:243–7.Search in Google Scholar

[19] Lascelles BDX, Flecknell PA. Do animal models tell us about human pain? Pain Clin Updates 2010;18:1–6.Search in Google Scholar

[20] Mogil JS, Davis KD, Derbyshire SW. The necessity of animal models in pain research. Pain 2010;151:12–7.Search in Google Scholar

[21] Kuettner KE, Goldberg VM. Osteoarthritis disorders. Rosemont: American Academy of Orthopaedic Surgeons; 1995. p. xxii–ii.Search in Google Scholar

[22] Wieland HA, Michaelis M, Kirschbaum BJ, Rudolphi KA. Osteoarthritis – an untreatable disease? Nat Rev Drug Discov 2005;4:331–45.Search in Google Scholar

[23] Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum 2003;48:3464–74.Search in Google Scholar

[24] Creamer P, Hochberg MC. Osteoarthritis. Lancet 1997;350:503–9.Search in Google Scholar

[25] Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D. Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain 2006;10:287–333.Search in Google Scholar

[26] Pelletier JP, Martel-Pelletier J, Raynauld JP. Most recent developments in strategies to reduce the progression of structural changes in osteoarthritis: today and tomorrow. Arthritis Res Ther 2006;8:206.Search in Google Scholar

[27] Bendele A, McComb J, Gould T, McAbee T, Senello G, Chlipala E, Guy M. Animal models of arthritis: relevance to human disease. Toxicol Pathol 1999;27:134–42.Search in Google Scholar

[28] Bendele AM. Animal models of osteoarthritis. JMusculoskelet Neuronal Interact 2001;1:363–76.Search in Google Scholar

[29] Poulet B, Hamilton RW, Shefelbine A, Pitsillides AA. Characterizing a novel and adjustable noninvasive murine joint loading model. Arthritis Rheum 2011;63:137–47.Search in Google Scholar

[30] Salo PT, Hogervorst T, Seerattan RA, Rucker D, Bray RC. Selective joint denervation promotes knee osteoarthritis in the aging rat. J Orthop Res 2002;20:1256–64.Search in Google Scholar

[31] Wei L, Hjerpe A, Brismar BH, Svensson O. Effect of load on articular cartilage matrix and the development of guinea-pig osteoarthritis. Osteoarthr Cartilage 2001;9:447–53.Search in Google Scholar

[32] Isaac DI, Meyer EG, Kopke KS, Haut RC. Chronic changes in the rabbit tibial plateau following blunt trauma to the tibiofemoral joint. J Biomech 2010;43:1682–8.Search in Google Scholar

[33] Boyd SK, Muller R, Leonard T, Herzog W. Long-term periarticular bone adaptation in a feline knee injury model for post-traumatic experimental osteoarthritis. Osteoarthr Cartilage 2005;13:235–42.Search in Google Scholar

[34] Pond MJ, Nuki G. Experimentally-induced osteoarthritis in the dog. Ann Rheum Dis 1973;32:387–8.Search in Google Scholar

[35] Brandt KD, Thonar EJ-MA. Lack of association between serum keratin sulfate concentrations and cartilage changes of osteoarthritis after transaction of the anterior cruciate ligament in the dog. Arthritis Rheum 1989;32: 647–51.Search in Google Scholar

[36] Mastbergen SC, Marijnissen AC, Vianen ME, van Roermund PM, Bijlsma JW, Lafeber FP. The canine ‘groove’ model of osteoarthritis is more than simply the expression of surgically applied damage. Osteoarthr Cartilage 2006;14:39–46.Search in Google Scholar

[37] Appleyard RC, Ghosh P, Swain MV. Biomedical, histological and immunohistological studies of patellar cartilage in an ovine model of osteoarthritis induced by lateral meniscectomy. Osteoarthr Cartilage 1999;7:281–94.Search in Google Scholar

[38] Muehleman C, Green J, Williams JM, Kuettner KE, Thonar E.J.-M.A., Sumner DR. The effect of bone remodeling inhibition by zoledronic acid in an animal model of cartilage matrix damage. Osteoarthr Cartilage 2002;10:226–33.Search in Google Scholar

[39] Pap G, Eberhardt R, Stürmer I, Machner A, Schwarzberg H, Roessner A, Neumann W. Development of osteoarthritis in the knee joints of wistar rats after strenuous running exercise in a running wheel by intracranial selfstimulation. Pathol Res Pract 1998;194:41–7.Search in Google Scholar

[40] Simmons EJ, Bertone AL, Weisbrode SE. Instability-induced osteoarthritis in the metacarpophalangeal joint of horses. Am J Vet Res 1999;60:7–13.Search in Google Scholar

[41] Helminen HJ, Säämänen A-M, Salminen H, Hyttinen MM. Transgenic mouse models for studying the role of cartilage macromolecules in osteoarthritis. Rheumatology 2002;41:848–56.Search in Google Scholar

[42] Glasson SS. In vivo osteoarthritis target validation utilizing geneticallymodified mice. Curr Drug Targ 2007;8:367–76.Search in Google Scholar

[43] Lascelles BDX. Feline degenerative joint disease. Vet Surg 2010;39:2–13.Search in Google Scholar

[44] Sokoleff L, Crittenden LB, Yamamoto RS, Jay Jr GE. The genetics of degenerative joint disease in mice. Arthritis Rheum 1962;5:531–46.Search in Google Scholar

[45] Walton M. Degenerative joint disease in the mouse knee: histological observations. J Pathol 1977;123:109–22.Search in Google Scholar

[46] Nordling C, Karlsson-Parra A, Jansson L, Holmdahl R, Klareskog L. Characterisation of spontaneously occurring arthritis in male DBA/1 mice. Arthritis Rheum 1992;35:717–22.Search in Google Scholar

[47] Anderson-Mackenzie JM, Hulmes DJS, Thorp BH. Degenerative joint disease in poultry – differences in composition and morphology of articular cartilage are associated with strain susceptibility. Res Vet Sci 1997;63:29–33.Search in Google Scholar

[48] Bendele AM, Hulman JF. Spontaneous cartilage degeneration in guinea pigs. Arthritis Rheum 1988;31:561–5.Search in Google Scholar

[49] Bendele AM, White SL, Hulman JF. Osteoarthrosis in guinea pigs: histopathologic and scanning electron microscopic features. Lab Anim Sci 1989;39:115–21.Search in Google Scholar

[50] de Bri E, Reinholt FP, Svensson O. Primary osteoarthrosis in guinea pigs: a stereological study. J Orthop Res 1995;13:769–76.Search in Google Scholar

[51] Silberberg R, Saxton J, Sperling G. Degenerative joint disease in Syrian hamsters. Fed Proc 1952;11:427–32.Search in Google Scholar

[52] Pritzker KPH, Chateauvert J, Grynpas MD, Renlund RC, Turnquist J, Kessler MJ. Rhesus Macaques as an experimental model for degenerative arthritis. P R Health Sci J 1989;8:99–102.Search in Google Scholar

[53] Carlson CS, Loeser RF, Jayo MJ, Weaver DS, Adams MR, Jerome CP. Osteoarthritis in cynomolgus macaques: a primate model of naturally occurring disease. J Orthop Res 1994;12:321–39.Search in Google Scholar

[54] Godfrey DR. Osteoarthritis in cats: a retrospective radiological study. J Small Anim Pract 2005;46:425–9.Search in Google Scholar

[55] Rychel JK. Diagnosis and treatment of osteoarthritis. Top Companion Anim Med 2010;25:20–5.Search in Google Scholar

[56] Johnston SA. Osteoarthritis, joint anatomy, physiology, and pathobiology. Osteoarthritis 1997;27:699–723.Search in Google Scholar

[57] Schaible HG, Richter F, Ebersberger A, Boettger MK, Vanegas H, Natura G, Vazquez E, von Banchet GS. Joint pain. Exp Brain Res 2009;196:153–62.Search in Google Scholar

[58] Riser WH. The dog as a model for the study of hip dysplasia: growth, form, and development of the normal and dysplastic hip joint: introduction. Vet Pathol 1975;12:235–8.Search in Google Scholar

[59] Aragon CL, Hofmeister EH, Budsberg SC. Systematic review of clinical trials of treatments for osteoarthritis in dogs. J Am Vet Med Assoc 2007;230:514–21.Search in Google Scholar

[60] Vaughan-Scott T, Taylor JH. The pathophysiology and medical management of canine osteoarthritis. J S Afr Vet Med Assoc 1997;68:21–5.Search in Google Scholar

[61] McLaughlin R. Management of chronic osteoarthritic pain. Vet Clin North Am Small Anim Pract 2000;30:933–49.Search in Google Scholar

[62] Innes J. Diagnosis and treatment of osteoarthritis in dogs. In Practice 1995;17:102–9.Search in Google Scholar

[63] Henrotin Y, Sanchez C, Balligand M. Pharmaceutical and nutraceutical management of canine osteoarthritis: present and future perspectives. Vet J 2005;170:113–23.Search in Google Scholar

[64] Marshall WG, Bockstahler BA, Hulse DA, Carmichael S. Areview of osteoarthritis and obesity: current understanding of the relationship and benefit of obesity treatment and prevention in the dog. Vet Comp Orthop Traumatol 2009;5:339–45.Search in Google Scholar

[65] Sanderson RO, Beata C, Flipo R-M, Genevois J-P, Macias C, Tacke S, Vezzoni A, Innes JF. Systematic review of the management of canine osteoarthritis. Vet Rec 2009;164:418–24.Search in Google Scholar

[66] Hampshire VA, Doddy FM, Post LO, Koogler TL, Burgess TM, Batten PO, Hudson R, McAdams DR, Brown MA. Adverse drug event reports at the United States Food and Drug Administration center for veterinary medicine. J Am Vet Med Assoc 2004;225:533–6.Search in Google Scholar

[67] Bonnett BN, Egenvall A. Age patterns of disease and death in insured Swedish dogs, cats and horses. J Comp Pathol 2010;142:S33–8.Search in Google Scholar

[68] Waarsing JH, Kloppenburg M, Slagboom PE, Kroon HM, Houwing-Duistermaat JJ, Weinans H, Meulenbelt I. Osteoarthritis susceptibility genes influence the association between hip morphology and osteoarthritis. Arthritis Rheum 2011;63:1349–54.Search in Google Scholar

[69] Valdes AM, Spector TD. Genetic epidemiology of hip and knee osteoarthritis. Nat Rev Rheumatol 2011;7:23–32.Search in Google Scholar

[70] Cachon T, Genevois JP, Remy D, Carozzo C, Viguier E, Maitre P, Arnault F, Fau D. Risk of simultaneous phenotypic expression of hip and elbow dysplasia in dogs. Vet Comp Orthop Traumatol 2010;23:28–30.Search in Google Scholar

[71] Guo G, Zhou Z, Wang Y, Zhao K, Zhu L, Lust G, Hunter L, Friedenberg S, Li J, Zhang Y, Harris S, Jones P, Sandler J, Krotscheck U, Todhunter R, Zhang Z. Canine hip dysplasia is predictable by genotyping. Osteoarthr Cartilage 2011;19:420–9.Search in Google Scholar

[72] Stock KF, Klein SB, Tellhelm B, Distl O. Genetic analyses of elbow and hip dysplasia in the German shepherd dogs. J Anim Breed Genet 2011;128:219–29.Search in Google Scholar

[73] Zhou Z, Sheng X, Zhang Z, Zhao K, Zhu L, Guo G, Friedenberg SG, Hunter LS, Vandenberg-Foels WS, Hornbuckle WE, Krotscheck U, Corey E, Moise NS, Dykes NL, Li J, Xu S, Du L, Wang Y, Sandler J, Acland GM, Lust G, Todhunter RJ. Differential genetic regulation of canine hip dysplasia and osteoarthritis. PLoS One 2010;5, 10.1371/journal.pone.0013219 [available online 11 October 2010].10.1371/journal.pone.0013219Search in Google Scholar

[74] Valdes AM, Loughlin J, Timms KM, van Meurs JJB, Southam L, Wilson SG, Doherty S, Lories RJ, Luyten FP, Gutin A, Abkevich V, Ge D, Hofman A, Uitterlinden AG, Hart DJ, Zhang F, Zhai G, Egli RJ, Doherty M, Lanchbury J, Spector TD. Genome-wide association scan identifies a prostaglandin-endoperoxide synthase 2 variant involved in risk of knee osteoarthritis. Am J Hum Genet 2008;82:1231–40.Search in Google Scholar

[75] Karlsson C, Dehne T, Lindahl A, Brittberg M, Pruss A, Sittinger M, Ringe J. Genome-wide expression profiling reveals new candidate genes associated with osteoarthritis. Osteoarthr Cartilage 2010;18:581–92.Search in Google Scholar

[76] Dworkin RH, Turk DC, Wyrwich KW, Beaton D, Cleeland CS, Farrar JT, Haythornthwaite JA, Jensen MP, Kerns RD, Ader DN, Brandenburg N, Burke LB, Cella D, Chandler J, Cowan P, Dimitrova R, Dionne R, Hertz S, Jadad AR, Katz NP, Kehlet H, Kramer LD, Manning DC, McCormick C, McDermott MP, McQuay HJ, Patel S, Porter L, Quessy S, Rappaport BA, Rauschkolb C, Revicki DA, Rothman M, Schmader KE, Stacey BR, Stauffer JW, von Stein T, White RE, Witter J, Zavisic S. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. J Pain 2008;9:105–21.Search in Google Scholar

[77] Brown DC, Boston RC, Coyne JC, Farrar JT. Ability of the canine brief pain inventory to detect response to treatment in dogs with osteoarthritis. J Am Vet Med Assoc 2008;233:1278–83.Search in Google Scholar

[78] Hercock CA, Pinchbeck G, Giejda A, Clegg PD, Innes JF. Validation of a clientbased clinical metrology instrument for the evaluation of canine elbow osteoarthritis. J Small Anim Pract 2009;50:266–71.Search in Google Scholar

[79] Hielm-Björkman AK, Kuusela E, Liman A, Markkola A, Saarto E, Huttunen P, Leppäluoto J, Tulamo R-M, Raekallio M. Evaluation of methods for assessment of pain associated with chronic osteoarthritis in dogs. J Am Vet Med Assoc 2003;222:1552–8.Search in Google Scholar

[80] Hielm-Björkman AK, Rita H, Tulamo RM. Psychometric testing of Helsinki chronic pain index by completion of a questionnaire in Finnish by owners of dogs with chronic signs of pain caused by osteoarthritis. Am J Vet Res 2009;70:727–34.Search in Google Scholar

[81] Hielm-Björkman AK, Kapatkin AS, Rita HJ. Reliability and validity of a visual analogue scale used by owners to measure chronic pain attributable to osteoarthritis in their dogs. Am J Vet Res 2011;72:601–7.Search in Google Scholar

[82] Griffon DJ, McLaughlin RM, Roush JK. Vertical ground reaction force redistribution during experimentally induced shoulder lameness in dogs. Vet Comp Orthop Traumatol 1994;7:154–7.Search in Google Scholar

[83] McLaughlin R. Kinetic and kinematic gait analysis in dogs. Vet Clin North Am Small Anim Pract 2001;31:193–201.Search in Google Scholar

[84] Poole R, Blake S, Buschmann M, Goldring S, Laverty S, Lockwood S, Matyas J, McDouglas J, Pritzker K, Rudolphi K, van der Berg W, Yaksh T. Recommendations for the use of preclinical models in the study and treatment of osteoarthritis. Osteoarthr Cartilage 2010;18:S10–6.Search in Google Scholar

[85] Budsberg SC, Verstraete MC, Soutas-Little RW. Force plate analysis of the walking gait in healthy dogs. Am J Vet Res 1987;48:915–8.Search in Google Scholar

[86] Budsberg SC, Jevens DJ, Brown J, Foutz TL, DeCamp CE, Reece L. Evaluation of limb symmetry indices using ground reaction forces in healthy dogs. AmJ Vet Res 1993;54:1569–74.Search in Google Scholar

[87] Budsberg SC, Verstraete MC, Brown J, Reece L. Vertical loading rates in clinically normal dogs at a trot. Am J Vet Res 1995;56:1275–80.Search in Google Scholar

[88] Lascelles BDX, Roe SC, Smith E, Reynolds L, Markham J, Marcellin-Little D, Bergh MS, Budsberg SC. Evaluation of a pressure walkway system for measurement of vertical limb forces in clinically normal dogs. Am J Vet Res 2006;67:277–82.Search in Google Scholar

[89] Hansen BD, Lascelles BDX, Keene BW, Adams AK, Thomson AE. Evaluation of an accelerometer for at-homemonitoring of spontaneous activity in dogs. Am J Vet Res 2007;68:468–75.Search in Google Scholar

[90] Light VA, Steiss JE, Montgomery RD, Rumph PF, Wright JC. Temporal spatial gait analysis by use of a portable walkway system in healthy Labrador retrievers at a walk. Am J Vet Res 2010;71:997–1002.Search in Google Scholar

[91] Besancon MF, Conzemius MG, Evans RB, Ritter MJ. Distribution of vertical forces in the pads of greyhounds and Labrador retrievers during walking. Am J Vet Res 2004;65:1497–501.Search in Google Scholar

[92] Mölsä SH, Hielm-Björkman AK, Laitinen-Vapaavuori OM. Force platform analysis in clinical healthy rottweilers: comparison with Labrador retrievers. Vet Surg 2010;39:701–7.Search in Google Scholar

[93] Voss K, Imhof J, Kaestner S, Montavon PM. Force plate gait analysis at the walk and trot in dogs with low-grade hindlimb lameness. Vet Comp Orthop Traumatol 2007;20:299–304.Search in Google Scholar

[94] Rumph PF, Kincaid SA, Baird DK, Kammermann JR, Visco DM, Goetze LF. Vertical ground reaction force distribution during experimentally induced acute synovitis in dogs. Am J Vet Res 1993;54:365–9.Search in Google Scholar

[95] Rumph PF, Kincaid SA, Visco DM, Baird DK, Kammermann JR, West MS. Redistribution of vertical ground reaction force in dogs with experimentally induced chronic hind limb lameness. Vet Surg 1995;24:384–9.Search in Google Scholar

[96] Beraurd R, Moreau M, Lussier B. Effect of exercise on kinetic gait analysis of dogs afflicted by osteoarthritis. Vet Comp Orthop Traumatol 2010;2:87–92.Search in Google Scholar

[97] Ballagas AJ, Montgomery RD, Henderson RA, Gillette R. Pre- and postoperative force plate analysis of dogs with experimentally transacted cranial cruciate ligaments treated using tibial plateau levelling osteotomy. Vet Surg 2004;33:187–90.Search in Google Scholar

[98] Budsberg SC, Verstraete MC, Soutas-Little RW, Flo GL, Probst CW. Force plate analysis before and after stabilization of canine stifles for cruciate injury. Am J Vet Res 1988;49:1522–4.Search in Google Scholar

[99] Conzemius MG, Evans RB, Becanson MF, Gordon WJ, Horstman CL, Hoefle WD, Nieves MA, Wagner SD. Effect of surgical technique on limb function after surgery for rupture of the cranial cruciate ligament in dogs. J Am Vet Med Assoc 2005;226:232–6.Search in Google Scholar

[100] Horstman CL, Conzemius MG, Evans R, Gordon WJ. Assessing efficacy of perioperative oral carprofen after cranial cruciate surgery using noninvasive, objective pressure platform gait analysis. Vet Surg 2004;33:286–92.Search in Google Scholar

[101] Fu Y-C, Torres BT, Budsberg SC. Evaluation of a three-dimensional kinematic model for canine gait analysis. Am J Vet Res 2010;71:1118–22.Search in Google Scholar

[102] Quessy SN. The challenges of translational research for analgesics: the state of knowledge needs upgrading and some uncomfortable deficiencies remain to be urgently addressed. J Pain 2010;11:698–700.Search in Google Scholar

[103] Paster ER, LaFond E, Biery DN, Iriye A, Gregor TP, Shofer FS, Smith GK. Estimates of prevalence of hip dysplacia in golden retrievers and rottweilers and influence of bias on published prevalence figures. J Am Vet Med Assoc 2005;226:387–92.Search in Google Scholar

[104] http://www.avma.org/reference/marketstats/ownership.asp [accessed 23. 03.11].Search in Google Scholar

[105] (http://www.fci.be/uploaded_files/Statistics_Europe_Oct27.pdf [accessed 31. 03.11].Search in Google Scholar

[106] Directive 2010/63/EU of the European Parliament and of the Council of 22 September 2010 on the protection of animals used for scientific purposes. Off J Eur Union 2010; L276/33–L276/79. http://eurlex.europa.eu/LexUriServ/LexUriServ.do_uri=OJ:L:2010:276:0033:0079:EN:PDF [accessed 04.04.11].Search in Google Scholar

[107] U.S. Department of Health & Human Services, Office of Extramural Research, National Institutes of Health. Office of Laboratory Animal Welfare. Health research extension act of 1985; public law 99-158, November 20, 1985. “Animals in Research”. http://grants.nih.gov/grants/olaw/references/hrea1985.htm [accessed 04. 03.11].Search in Google Scholar

[108] European Medicines Agency (EMEA), EudraLex – volume 5 – pharmaceutical legislation medicinal products for veterinary use. http://ec.europa.eu/health/documents/eudralex/vol-5/index_en.htm [accessed 04.04.11].Search in Google Scholar

[109] U.S. Food and Drug Agency (FDA). Animal & veterinary, development & approval process. http://www.fda.gov/AnimalVeterinary/Development_Approval_Process/default.htm [accessed 04.04.11].Search in Google Scholar

[110] Proudfoot AG, McAuley DF, Hind M, Griffiths MJ. Translational research: what does it mean, what has it delivered and what might it deliver? Curr Opin Crit Care 2011 [Epub ahead of print].Search in Google Scholar

[111] Rowell JL, McCarthy DO, Alvarez CE. Dog models of naturally occurring cancer. Trends MolMed. 10.1016/j.molmed.2011.02.004 [available online 24 March 2011].10.1016/j.molmed.2011.02.004Search in Google Scholar

[112] Wehling M. Drug development in the light of translational science: shine or shade? Drug Discov Today 2011, 10.1016/j.drudis.2011.07.008 [Epub ahead of print].10.1016/j.drudis.2011.07.008Search in Google Scholar

[113] Bonnett BN, Egenvall A, Hedhammar Å, Olson P. Mortality in over 350,000 insured Swedish dogs from 1995-2000: I. breed-, gender-, age- and causespecific rates. Acta Vet Scand 2005:105–20.Search in Google Scholar

[114] Lawler DF, Evans RH, Larson BT, Spitznagel EL, Ellersieck MR, Kealy RD. Influence of lifetime food restriction on causes, time, and predictors of death in dogs. J Am Vet Med Assoc 2005;226:199–265.Search in Google Scholar

[115] Merskey H, Albe-Fessard DG, Bonica JJ, Carmon A, Dubner R, Kerr FWL, Lindblom U, Mumford JM, Nathan PW, Noordenbos W, Pagni CA, Renaer MJ, Sternbach RA, Sunderland S. Pain terms: a list with definitions and notes on usage. Recommended by the IASP Subcommittee of Taxonomy. Pain 1979;6:249–52.Search in Google Scholar

[116] http://www.iasp-pain.org//AM/Template.cfm_Section=Home [accessed 23. 09.11].Search in Google Scholar

[117] American College of Veterinary Anesthesiologists’ position paper on the treatment of pain in animals. J Am Vet Med Assoc 1998;213:628–30.Search in Google Scholar

[118] Molony V, Kent JE. Assessment of acute pain in farm animals using behavioural and physiological measurements. J Anim Sci 1997;75:266–72.Search in Google Scholar

[119] Johnson CB, Sylvester SP, Stafford KJ, Mitchinson SL, Ward N, Mellor DJ. Effects of age on the electroencephalographic response to castration in lambs anaesthetized with halothane in oxygen from birth to 6 weeks old. Vet Anaesth Analg 2009;36:273–9.Search in Google Scholar

[120] Hare B, Tomasello M. Human-like social skills in dogs. Trends Cogn Sci 2005;9:439–44.Search in Google Scholar

[121] Tomasello M, Kaminski J. Like infant, like dog. Science 2009;325:1213–4.Search in Google Scholar

[122] Miklósi A. Evolutionary approach to communication between humans and dogs. Vet Res Commun 2009;33 (Suppl 1):S53–9.Search in Google Scholar

[123] Udell MAR, Wynne CDL. A review of domestic dogs’ (Canine familiaris) human-like behaviors: or why behavior analysts should stop worrying and love their dogs. J Exp Anal Behav 2008;89:247–61.Search in Google Scholar

[124] Reid PJ. Adapting to the human world: dogs’ responsiveness to our social cues. Behav Processes 2009;80:325–33.Search in Google Scholar

Received: 2011-07-01
Revised: 2011-11-02
Accepted: 2011-11-18
Published Online: 2012-04-01
Published in Print: 2012-04-01

© 2011 Scandinavian Association for the Study of Pain

Downloaded on 7.6.2024 from https://www.degruyter.com/document/doi/10.1016/j.sjpain.2011.11.007/html
Scroll to top button