Animal models of pain: Diversity and benefits

https://doi.org/10.1016/j.jneumeth.2020.108997Get rights and content

Highlights

  • Evaluation tools to assess chronic pain and its comorbidities

  • inventory of a large diversity of pain models including rodents, non-human primates pain models and simple organisms

  • Focus on inflammatory and neuropathic pain models

  • Limitations of the predictive validity of animal models of pain

  • Translational successes from animal model studies to develop new analgesics

Abstract

Chronic pain is a maladaptive neurological disease that remains a major health problem. A deepening of our knowledge on mechanisms that cause pain is a prerequisite to developing novel treatments. A large variety of animal models of pain has been developed that recapitulate the diverse symptoms of different pain pathologies. These models reproduce different pain phenotypes and remain necessary to examine the multidimensional aspects of pain and understand the cellular and molecular basis underlying pain conditions.

In this review, we propose an overview of animal models, from simple organisms to rodents and non-human primates and the specific traits of pain pathologies they model. We present the main behavioral tests for assessing pain and investing the underpinning mechanisms of chronic pathological pain. The validity of animal models is analysed based on their ability to mimic human clinical diseases and to predict treatment outcomes. Refine characterization of pathological phenotypes also requires to consider pain globally using specific procedures dedicated to study emotional comorbidities of pain. We discuss the limitations of pain models when research findings fail to be translated from animal models to human clinics. But we also point to some recent successes in analgesic drug development that highlight strategies for improving the predictive validity of animal models of pain. Finally, we emphasize the importance of using assortments of preclinical pain models to identify pain subtype mechanisms, and to foster the development of better analgesics.

Introduction

Pain is a vital physiological function that protects organisms against potential damage. Acute nociceptive pain is a normal function of the nervous system that provides important sensory information about the environment and reacts to harmful stimuli such as noxious heat, extreme cold, chemical irritants, and mechanical tissue damage. These noxious stimuli activate peripheral nociceptors, triggering action potentials that propagate along sensory axons to the dorsal horn of the spinal cord where nociceptive inputs are processed and relayed to the brain. In turn, the activation of specific brain areas produces a broad array of sensory, emotional, autonomic, and motor responses that shape our experience and perception of pain (Basbaum et al., 2009; Burma et al., 2017).

In contrast to acute pain, chronic pain is a maladaptive disease that heightens the sensitivity to sensory stimulation (Woolf and Salter, 2000; Costigan et al., 2009). Chronic pain results from abnormal functioning of the nervous system, with pain persisting far beyond the resolution of the primary injury. Pain hypersensitivity manifests as spontaneous pain (pain in the absence of an external stimulus), allodynia (pain resulting from an innocuous stimulus), and/or hyperalgesia (an exaggerated pain response to a noxious stimulus). Chronic pain is a major health problem that negatively impacts the quality-of-life of sufferers and exacts enormous socio-economic costs with a prevalence of around 8 % of the general population (Bouhassira et al., 2008). In the European Union, it is estimated that 20 % of the population would suffer from chronic pain during lifespan (Breivik et al., 2006; Alshami, 2014), and it is among the most significant risk factors for suicide.

Chronic pain drastically diminishes quality of life and causes enormous socio-economic costs. Besides high costs for disease management, chronic pain is associated with major impacts on daily activities and quality of life (Groenewald and Palermo, 2015) and high productivity losses due to work absences (Mayer et al., 2019) partly due to common co-morbidities such as depression (Phillips, 2006). The estimated direct and indirect healthcare costs for chronic pain disorders in European Member States vary between two and three percent of GDP across the EU (Breivik et al., 2013). For 2016, this estimate would result in up to €441 billion. In the USA, ∼100 million people suffer from pain costing ∼$600 billion/year in health care and lost productivity (Walker et al., 2014). These costs are reported to exceed those estimated for heart disease, cancer and diabetes (Breivik et al., 2013). However, chronic pain is poorly managed with treatment success rates around 30 % (Ossipov et al., 2014), mainly because chronic pain mechanisms remain poorly understood (Basbaum et al., 2009; Dolique et al., 2010; Cordero-Erausquin et al., 2016; Kuner and Flor, 2016), and patients often suffer from comorbid disorders such as anxiety and depression (Attal et al., 2011).

It is important to recognize that there is not one overarching, singular condition called chronic pain but rather, there are multiple aetiologies of pain, each resulting from different pathologies and differing in the clinical presentation of signs and symptoms (Burma et al., 2017). Pain is usually subdivided in different categories as a function of the mechanism of injury. Nociceptive pain represents the normal response to noxious insult or injury of tissues such as skin, muscles, visceral organs, joints, tendons, or bones. Inflammatory pain results of activation and sensitization (peripheral and/or central) of the nociceptive pain pathway by a variety of mediators released at a site of tissue inflammation. Neuropathic pain arises from damage to the nervous system itself, central or peripheral, either from disease, injury, or pinching. Other types of pain were characterized, e.g. cancer pain or dysfunctional pain when no biological cause is identified. To address differences in the presentation of pain symptoms across chronic pain conditions, an array of preclinical animal models has been developed to recapitulate the underlying pathology, duration, and comorbidities of pain phenotypes (Mogil, 2009). This variety of preclinical pain models is essential for understanding the molecular and cellular mechanisms that underlie distinct pain conditions. The development of novel and more efficacious therapies requires a thorough understanding of the mechanistic underpinnings of chronic pain and the design and testing of new drugs.

Rodents are employed in an large majority of preclinical pain studies (Mogil, 2009). However, the use of alternate vertebrates and invertebrates, such as zebrafish, fruit flies (Drosophila sp.) and nematodes (Caenorhabditis elegans) can also be advantageous for screening assays and for studying the genetic and molecular mechanisms of acute and chronic pain (Way and Chalfie, 1989; Gonzalez-Nunez and Rodríguez, 2009; Milinkeviciute et al., 2012). Each organism confers a distinct advantage for studying pain; the behavioral complexity of the rodents allows the analysis of the affective components of pain (Johansen et al., 2001; Panksepp and Lahvis, 2011), whereas simpler organisms, such as Drosophila, can facilitate the discovery of novel molecular players involved in the detection of noxious stimuli (Caldwell and Tracey, 2010; Mogil et al., 2010).

Another important aspect for designing animal models is to recognize that pain is a multi-dimensional experience. Indeed, pain is processed not only at the peripheral and spinal levels, but also in higher brain structures including cortical areas underlying the affective component of pain (Liu and Chen, 2014). As pain transitions from acute to chronic and becomes pathological, the associated negative emotional state not only exacerbates sensory modalities, but also worsens the comorbidities. Indeed, anxiety is a highly common comorbidity of pain where the interactions between pain and anxiety have been demonstrated in human. In addition, chronic pain and depression are complex disorders that often coexist and increase the risk of one another (Radat et al., 2013; Steel et al., 2014; Zhu et al., 2018a). Investigations of pain and psychiatric disorders are mostly conducted separately, but more knowledge of the overlap and interactions between affective and pain circuits is key to better treatments. Refined analysis of animal models of pain and depression has become mandatory to understand interactions between pain and emotional comorbidities (Kremer et al., 2020).

The present review proposes an overview of rodent and alternate models of pain. A large panel of evaluation devices used to characterize pain-like behavior in animal models has been developed over the years, and a summary description of these tools is presented here. The review focuses mainly on the broadly studied neuropathic and inflammatory types of pain. It also highlights the necessity of specific procedures dedicated to studying emotional comorbidities to pain. Finally, it summarizes the main limitations of using animal models to mimic clinical pain in humans, but also provides examples of successful translational applications of using animal models.

Section snippets

Assessment of pain

The classical pain evaluation devices aim to assess the sensory component of pain, usually by measuring a withdrawal reflex. However, more recently, specific efforts have been made to assess pain perception and to evaluate the emotional component of pain and comorbid affections. Unlike humans, animals are incapable of verbally describing pain, therefore a battery of behavioral tests has been developed in order to assess pain-like behavior in animals. These assays can be divided into

Models of inflammatory pain

Two important parameters to be considered in animal models of pain are the method of injury and the endpoint measurement. The most appropriate models, whether an injury, application of chemical agents, or other manipulations, should be based on 1) understanding the clinical disease presentation and pathology (i.e. face validity); 2) producing nociception by recapitulating the mechanisms of specific clinical conditions (i.e. construct validity). Measures of nociceptive behavior must not only

Models of migraine pain

Migraine has not classically been considered an inflammatory disease probably because it is not obviously associated with heat, redness, and swelling. Instead, a vascular aetiology was proposed and the prevailing theory of migraine for most of the twentieth century, held that pain results from an abnormal dilatation of intracranial blood vessels, leading to mechanical excitation of sensory fibers. However, in recent years, advancements in the neurobiology of migraine headache have shifted the

Models of neuropathic pain

Neuropathic pain (NP) is a painful syndrome caused by central or peripheral lesion of the nervous system. It is highly disabling, affecting 7–8 % of general population (Bouhassira et al., 2008). NP elicits sensory alteration including dysesthesia and paresthesia, spontaneous pain, increase pain sensation for innocuous stimuli (allodynia) and increase pain sensation for noxious stimuli (hyperalgesia). Animal models of NP are developed, mainly in rodents, to recapitulate one or several symptoms

Models of cancer pain

Several mammalian and non-mammalian animal models have been developed to improve our understanding of cancer biology (Schachtschneider et al., 2017). However, cancer pain, which is experienced by human patients and animals in advanced stages of cancer, has only been addressed in some of these models.

Immunocompromised and immunocompetent rodents (mouse and rat) are commonly used to study cancer pain. Because of its large size, rat model is better suited for manipulations and injections into

Models of visceral pain

Visceral pain includes pain emanating from organs localized into the thoracic, pelvic and abdominal regions. This type of pain is poorly localized, often affecting two or more visceral organs. In particular, gastrointestinal (GI) pain is common in various disorders including irritable bowel syndrome, inflammatory bowel diseases (IBD) (Crohn’s disease and ulcerative colitis), pancreatitis, kidney stones, biliary disorders or associated with cancer. Several factors have been identified as

Models of musculoskeletal pain

Similarly to cancer pain or visceral pain, muscle pain results in a diffuse, aching pain that contrasts with sharp and localized cutaneous pain. Models specific to studying muscle pain are scarce in the literature although muscle pain is a major clinical problem. Indeed, musculoskeletal pain is a chronic widespread pain manifestation that affect between one quarter and one third of the US population (Gaskin and Richard, 2012). In chronic conditions like lowback pain, myositis, myofascial pain,

The particular case of non-human primates

Although rodents are mammals, their behavior and the organization of their nervous system remains different from humans. In contrast, studying pain in non-human primates offers the opportunity to investigate mechanisms, and response to analgesic treatment, that are close to what is expected in humans. However, using non-human primates undoubtedly poses acute ethical problems and little exploration of pain-related mechanisms has been done in non-human primates as compared to rodent.

Few attempts

Pain models in simple organisms

To reduce the use of rodents in pain studies and carry out high-throughput screening studies, new models have started to appear in pain research, using lower vertebrate species such as zebrafish or Xenopus and invertebrates such as Drosophila and Caenorhabditis elegans (C. elegans).

The shared advantages of these models reside in their simplicity of use including low cost and easy maintenance, large brood and egg size and rapid external development. They are genetically similar to humans and

Limitations of animal models of pain

The study of pain always relied on preclinical animal models that attempted to explore the complex physiological and sensory implications of the condition. Various animal models of chronic pain aim to emulate different types of pain and they have been instrumental in the discovery and development of analgesic agents. While there is no doubt about the necessity of such models in chronic pain research, there are several limitations to their use. This is especially true when considering their

Translational lessons learned from animal model studies

Translation is defined by the National center for advancing translation science as “the process of turning observations in the laboratory, clinic, and community into interventions that improve the health of individuals and populations – from diagnostics and therapeutics to medical procedures and behavioral interventions” (Nakao, 2019). It is worth noting that despite the aforementioned limitations, a growing number of examples demonstrate the potential to convert new agents into widely used

Conclusion

Animal models of pain were historically viewed as reliable tools that have served to advance research in the field over the past decades. They have been instrumental in constructing a global picture of how key proteins, signalling systems, and neural circuits contribute to pain-like behaviors. However, there are limitations and caveats to these models that must be acknowledged when considering the translation of research findings from the bench to the bedside. There is not one “best model” for

CRediT authorship contribution statement

Cynthia Abboud: Writing - original draft. Alexia Duveau: Writing - original draft. Rabia Bouali-Benazzouz: Writing - original draft. Karine Massé: Writing - original draft. Joseph Mattar: Writing - original draft. Louison Brochoire: Writing - original draft, Writing - review & editing. Pascal Fossat: Writing - original draft. Eric Boué-Grabot: Writing - original draft. Walid Hleihel: Funding acquisition, Supervision, Writing - original draft. Marc Landry: Funding acquisition, Supervision,

Declaration of Competing Interest

The authors declare they have no competing interests.

Acknowledgements

This work is supported by an ANR grant (Relax, n° 193992). CA is the recipient of a scholarship from the CNRSL (Lebanon). We thank Dr. Najib NAJJAR, Adjunct Instructor at Holy Spirit University of Kaslik – USEK, Chairperson at American University of Technology – AUT, for English language editing.

References (507)

  • A. Berić

    Central pain and dysesthesia syndrome

    Neurol. Clin.

    (1998)
  • T. Bleehen et al.

    Observations on the algogenic actions of adenosine compounds on the human blister base preparation

    Pain

    (1977)
  • G.D. Bossé et al.

    Development of an opioid self-administration assay to study drug seeking in zebrafish

    Behav. Brain Res.

    (2017)
  • D. Bouhassira et al.

    Prevalence of chronic pain with neuropathic characteristics in the general population

    Pain

    (2008)
  • M. Bourin et al.

    The mouse light/dark box test

    Eur. J. Pharmacol.

    (2003)
  • S.E. Bove et al.

    Surgically induced osteoarthritis in the rat results in the development of both osteoarthritis-like joint pain and secondary hyperalgesia

    Osteoarthr. Cartil.

    (2006)
  • J.M. Boyce-Rustay et al.

    Comparison of mechanical allodynia and the affective component of inflammatory pain in rats

    Neuropharmacology

    (2010)
  • H. Breivik et al.

    Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment

    Eur. J. Pain Lond. Engl.

    (2006)
  • J. Brodkin et al.

    Validation and implementation of a novel high-throughput behavioral phenotyping instrument for mice

    J. Neurosci. Methods

    (2014)
  • S.H. Buck et al.

    Thermal analgesia and substance P depletion induced by capsaicin in guinea-pigs

    Neuroscience

    (1981)
  • G. Burnstock

    Purinergic mechanisms and pain

    Adv. Pharmacol. San Diego Calif

    (2016)
  • M.G. Buzzi et al.

    Dihydroergotamine and sumatriptan attenuate levels of CGRP in plasma in rat superior sagittal sinus during electrical stimulation of the trigeminal ganglion

    Neuropharmacology

    (1991)
  • M. Calvo et al.

    The Genetics of Neuropathic Pain from Model Organisms to Clinical Application

    Neuron

    (2019)
  • J.N. Campbell et al.

    Mechanisms of neuropathic pain

    Neuron

    (2006)
  • S.M. Carlton et al.

    Behavioral manifestations of an experimental model for peripheral neuropathy produced by spinal nerve ligation in the primate

    Pain

    (1994)
  • R.B. Carter et al.

    Capsaicin desensitization to plasma extravasation evoked by antidromic C-fiber stimulation is not associated with antinociception in the rat

    Neurosci. Lett.

    (1991)
  • J.L. Cash et al.

    Chapter 17. Zymosan-induced peritonitis as a simple experimental system for the study of inflammation

    Methods Enzymol.

    (2009)
  • A. Catanzaro et al.

    Preliminary study on attitudes, opinions and knowledge of Italian veterinarians with regard to abdominal visceral pain in dogs

    Vet. Anaesth. Analg.

    (2016)
  • J.-P. Chae et al.

    Evaluation of developmental toxicity and teratogenicity of diclofenac using Xenopus embryos

    Chemosphere

    (2015)
  • S.R. Chaplan et al.

    Quantitative assessment of tactile allodynia in the rat paw

    J. Neurosci. Methods

    (1994)
  • I.P. Chessell et al.

    Disruption of the P2X7 purinoceptor gene abolishes chronic inflammatory and neuropathic pain

    Pain

    (2005)
  • A.R. Chopade et al.

    Pharmacological characterization of carrageenan induced heat muscle hyperalgesia in rats using non-selective, preferential and selective COX-2 inhibitors

    Pharmacol. Rep. PR

    (2014)
  • M.D. Christensen et al.

    Mechanical and thermal allodynia in chronic central pain following spinal cord injury

    Pain

    (1996)
  • E. Cohen et al.

    Caenorhabditis elegans nicotinic acetylcholine receptors are required for nociception

    Molecular and Cellular Neurosciences

    (2014)
  • R.W. Colburn et al.

    Attenuated cold sensitivity in TRPM8 null mice

    Neuron

    (2007)
  • M. Cordero-Erausquin et al.

    Neuronal networks and nociceptive processing in the dorsal horn of the spinal cord

    Neuroscience

    (2016)
  • B. Costa et al.

    Effect of the cannabinoid CB1 receptor antagonist, SR141716, on nociceptive response and nerve demyelination in rodents with chronic constriction injury of the sciatic nerve

    Pain

    (2005)
  • F.V. Costa et al.

    Understanding nociception-related phenotypes in adult zebrafish: behavioral and pharmacological characterization using a new acetic acid model

    Behav. Brain Res.

    (2019)
  • J. Crawley et al.

    Preliminary report of a simple animal behavior model for the anxiolytic effects of benzodiazepines

    Pharmacol. Biochem. Behav.

    (1980)
  • Y. Cui et al.

    A voxel-based analysis of brain activity in high-order trigeminal pathway in the rat induced by cortical spreading depression

    NeuroImage

    (2015)
  • I. Abdus-Saboor et al.

    Development of a mouse pain scale using sub-second behavioral mapping and statistical modeling

    Cell Rep.

    (2019)
  • M.Sde Abreu et al.

    Zebrafish as a model of neurodevelopmental disorders

    Neuroscience

    (2020)
  • N. Agarwal et al.

    Evoked hypoalgesia is accompanied by tonic pain and immune cell infiltration in the dorsal root ganglia at late stages of diabetic neuropathy in mice

    Mol. Pain

    (2018)
  • S. Akerman et al.

    Pearls and pitfalls in experimental in vivo models of migraine: dural trigeminovascular nociception

    Cephalalgia Int. J. Headache

    (2013)
  • B. Aldrich et al.

    The amnesiac gene is involved in the regulation of thermal nociception in Drosophila melanogaster

    Journal of Neurogenetics

    (2010)
  • A.J. Allchorne et al.

    Detection of cold pain, cold allodynia and cold hyperalgesia in freely behaving rats

    Mol. Pain

    (2005)
  • A.J. Alonso-Castro et al.

    Synergism between Naproxen and Rutin in a Mouse Model of Visceral Pain

    Pain. Drug Dev. Res.

    (2017)
  • A.M. Alshami

    Knee osteoarthritis related pain: a narrative review of diagnosis and treatment

    Int. J. Health Sci.

    (2014)
  • P. Anand et al.

    Rational treatment of chemotherapy-induced peripheral neuropathy with capsaicin 8% patch: from pain relief towards disease modification

    J. Pain Res.

    (2019)
  • C.K. Appel et al.

    Decitabine attenuates nociceptive behavior in a murine model of bone cancer pain

    Pain

    (2019)
  • Cited by (52)

    View all citing articles on Scopus
    1

    Share seniority.

    View full text