Summary

Rating L-DOPA-Induced Dyskinesias in the Unilaterally 6-OHDA-Lesioned Rat Model of Parkinson's Disease

Published: October 04, 2021
doi:

Summary

Rodent models of L-DOPA-induced dyskinesias are invaluable tools to identify therapeutic interventions to attenuate the development or alleviate the manifestations that emerge due to the repeated administration of L-DOPA. This protocol demonstrates how to induce and analyze dyskinetic-like movements in the unilaterally 6-OHDA-lesioned rat model of Parkinson’s disease.

Abstract

L-DOPA-induced dyskinesias (LIDs) refer to motor complications that arise from prolonged L-DOPA administration to patients with Parkinson’s disease (PD). The most common pattern observed in the clinic is the peak-dose dyskinesia which consists of clinical manifestations of choreiform, dystonic, and ballistic movements. The 6-hydroxydopamine (6-OHDA) rat model of PD mimics several characteristics of LIDs. After repeated L-DOPA administration, 6-OHDA-lesioned rats exhibit dyskinetic-like movements (e.g., abnormal involuntary movements, AIMs). This protocol demonstrates how to induce and analyze AIMs in 6-OHDA-lesioned rats with 90%-95% dopaminergic depletion in the nigrostriatal pathway. Repeated administration (3 weeks) of L-DOPA (5 mg/kg, combined with 12.5 mg/kg of benserazide) can induce the development of AIMs. The time course analysis reveals a significant increase in AIMs at 30-90 min (peak-dose dyskinesia). Rodent models of LIDs are an important preclinical tool to identify effective antidyskinetic interventions.

Introduction

The dopamine precursor L-3,4-dihydroxyphenylalanine (L-DOPA) represents the most effective treatment for the motor symptoms of Parkinson's disease (PD)1. L-DOPA therapy may ameliorate motor symptoms associated with PD but loses effectiveness with time. Motor fluctuations such as "wearing-off fluctuation" or "end-of-dose deterioration" manifest clinically as a shortened duration of the effect of single L-DOPA doses2. In other cases, clinical manifestations consist of slow twisting movements and abnormal postures (dystonia)3 and occur when dopamine levels are low (off-period dystonia)4. On the other hand, L-DOPA-induced dyskinesias (LIDs) appear when dopamine levels in the plasma and the brain are high5.

LIDs produce debilitating side effects that include motor complications such as choreiform, dystonic, and ballistic6 movements. Once established, LIDs occur after every L-DOPA administration. Motor complications occur in 40%-50% of PD patients undergoing L-DOPA therapy for 5 years, and the incidence increases over the years7. Although the pathophysiological mechanisms involved in the development of LIDs in PD patients are not yet fully elucidated, the extent of dopaminergic denervation, pulsatile L-DOPA administration, downstream changes in striatal proteins and genes, and abnormalities in non-dopamine transmitter systems are factors that contribute to the development of these unwanted side effects6,8,9,10.

The neurotoxin 6-hydroxydopamine (6-OHDA) is a well-characterized tool to study PD in rodents11,12,13,14. Since 6-OHDA does not cross the blood-brain barrier, it must be injected directly into the nigrostriatal pathway. 6-OHDA-induced dopaminergic depletion is concentration- and site-dependent15. Unilateral administration of 6-OHDA at the medial forebrain bundle (MFB) can produce severe (>90%) nigrostriatal damage in rodents16,17,18,19. Chronic administration of L-DOPA to severe unilaterally 6-OHDA-lesioned rodents causes the appearance of dyskinetic-like movements named abnormal involuntary movements (AIMs). Dyskinetic-like movements in rodents share similar molecular, functional, and pharmacological mechanisms related to LIDs in PD patients5. Therefore, 6-OHDA-lesioned rats20 and mice21 are valuable preclinical models to study LIDs. When treated chronically (7-21 days) with therapeutic doses of L-DOPA (5-20 mg/kg), unilaterally 6-OHDA-lesioned rats and mice show a gradual development of AIMs that affect the forelimb, trunk, and orofacial muscles contralateral to the lesion17,18,19,20,22,23,24. These movements are presented at a time course similar to L-DOPA-induced peak-dose dyskinesias in PD patients25 and are characterized by hyperkinetic movements and dystonia5. AIMs are usually scored based on their severity (e.g., when a specific AIM is present) and amplitude (e.g., characterized by the amplitude of each movement)5,23,25.

6-OHDA-lesioned rodent models of LIDs present face validity (i.e., the model has several characteristics that look like the human condition)5,11,26,27,28. Rodent AIMs, similar to what occurs in PD patients, are seen as hyperkinetic (forelimb and orolingual) and dystonic (axial) movements29 and mimics peak-dose dyskinesia. At the molecular and functional level, rodent models share many pathological characteristics with PD patients5, such as upregulation of FosB/ΔFosB19,26,30,31,32,33 and serotonin transporter (SERT)34,35. Concerning predictive validity, drugs that reduce LIDs in PD patients (e.g., the N-methyl-D-aspartate (NMDA) receptor antagonist amantadine) present antidyskinetic efficacy in the rodent model22,36,37,38,39.

The rodent AIMs rating scale was created based on four AIMs subtypes that include AIMs affecting the head, neck, and trunk (axial AIMs), hyperkinetic forelimb movements (limb AIMs), and dyskinetic-like orolingual movements (orolingual AIMs). Although contralateral rotation (locomotive AIMs) is also present in unilaterally lesioned rodents20,22,23,25,40, it has not been scored as a dyskinetic-like movement since it may not represent a specific measure of LIDs22,37,41.

Here, we will describe how to induce and analyze dyskinetic-like movements (axial, limb, and orolingual AIMs) in the severe (>90%) unilaterally 6-OHDA-lesioned rat model of PD. We organized our protocol based on the previous literature and our laboratory expertise.

Protocol

All experiments were performed in accordance with The Ethics Committee of the Faculty of Philosophy, Sciences, and Letters of Ribeirão Preto (CEUA/FFCLRP 18.5.35.59.5). 1. 6-OHDA lesion Use Sprague-Dawley male rats weighing 200-250 g at the beginning of the experiments (6 weeks). House the animals (2-3 per cage) under standard laboratory conditions (12:12 h light/dark cycle, lights on at 06:00 h, temperature-controlled facilities (22-24 °C), with foo…

Representative Results

Although the AIMs patterns observed in rats are simpler and limited compared to those observed in humans and nonhuman primates, this model reproduces both hyperkinetic and dystonic-like movements induced by chronic L-DOPA administration. Here we present data collected from a group (n = 10) of unilaterally 6-OHDA-lesioned rats chronically treated with L-DOPA (5 mg/kg combined with 12.5 mg/kg of benserazide) for 3 weeks (Monday to Friday). Note that the data presented in Figure 2, <strong clas…

Discussion

This protocol demonstrates how to induce and analyze AIMs in the rat model of PD induced by unilateral microinjection of 6-OHDA in the MFB. Chronic daily administration of low doses of L-DOPA (5 mg/kg, combined with 12.5 mg/kg of benserazide) produced the development of AIMs over the 3 weeks of treatment. Temporal analysis revealed a significant increase of AIMs, and the peak-dose dyskinesia is observed between 30 and 90 min after L-DOPA administration. AIMs are repetitive and purposeless movements affecting axial, limb,…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was supported by São Paulo Research Foundation (FAPESP, grant 2017/00003-0). We are grateful for the Coordination for the Improvement of Higher Education Personnel (CAPES). We thank Dr. Anthony R. West, Dr. Heinz Steiner, and Dr. Kuei Y. Tseng for support and mentoring.

Materials

6-hydroxydopamine hydrobromide Sigma-Aldrich, USA H6507 Neurotoxin that produces degeneration of catecholaminergic terminals
Benzerazide hydrochloride Sigma B7283 Peripheral dopa-decarboxylase inhibitor
Camera Bullet IR Turbo HD (HD-TVI)  2.8mm B HIKVISION DS-2CE16C0T-IRP Camera used to record all behavior
Imipramine hidrochloride Alfa Aesar J63723 Norepinephrine transporter inhibitor (NET) used to protect noradrenergic neurons from 6-OHDA
Ketamine hydrochloride Ceva Animal Health Anesthesia for surgical intervention
L-3,4-dihydroxyphenylalanine (L-DOPA) methyl ester (hydrochloride) Cayman Chemical Company 16149 Dopamine precursor
Mirrors Used to observe the behavior of animals during experiments in all directions
Needles 0.30 x 13 mm PrecisionGlide Needles used to inject drugs
Sodium chloride (NaCl) Samtec Salt
Syringes 1 ml Sterile BD Plastipak Syringes used to inject drugs
Transparent cylinders Used to record animal behavior during experiments
Xylazine hydrochloride Ceva Animal Health Sedative, analgesic and muscle relaxant for surgical intervention

References

  1. Jenner, P. Molecular mechanisms of L-DOPA-induced dyskinesia. Nature Reviews. Neuroscience. 9 (9), 665-677 (2008).
  2. Nutt, J. G. Levodopa-induced dyskinesia: review, observations, and speculations. Neurology. 40 (2), 340-345 (1990).
  3. Luquin, M. R., Scipioni, O., Vaamonde, J., Gershanik, O., Obeso, J. A. Levodopa-induced dyskinesias in Parkinson’s disease: clinical and pharmacological classification. Movement Disorders: Official Journal of the Movement Disorder Society. 7 (2), 117-124 (1992).
  4. Fabbrini, G., Brotchie, J. M., Grandas, F., Nomoto, M., Goetz, C. G. Levodopa-induced dyskinesias. Movement Disorders: Official Journal of the Movement Disorder Society. 22 (10), 1379 (2007).
  5. Cenci, M. A., Crossman, A. R. Animal models of l-dopa-induced dyskinesia in Parkinson’s disease. Movement Disorders: Official Journal of the Movement Disorder Society. 33 (6), 889-899 (2018).
  6. Bastide, M. F., et al. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson’s disease. Progress in Neurobiology. 132, 96-168 (2015).
  7. Manson, A., Stirpe, P., Schrag, A. Levodopa-induced-dyskinesias clinical features, incidence, risk factors, management and impact on quality of life. Journal of Parkinson’s Disease. 2 (3), 189-198 (2012).
  8. Grandas, F., Galiano, M. L., Tabernero, C. Risk factors for levodopa-induced dyskinesias in Parkinson’s disease. Journal of Neurology. 246 (12), 1127-1133 (1999).
  9. Schrag, A., Quinn, N. Dyskinesias and motor fluctuations in Parkinson’s disease. A community-based study. Brain: A Journal of Neurology. 123, 2297-2305 (2000).
  10. Bezard, E., Brotchie, J. M., Gross, C. E. Pathophysiology of levodopa-induced dyskinesia: potential for new therapies. Nature Reviews. Neuroscience. 2 (8), 577-588 (2001).
  11. Tronci, E., Shin, E., Björklund, A., Carta, M. Amphetamine-induced rotation and L-DOPA-induced dyskinesia in the rat 6-OHDA model: a correlation study. Neuroscience Research. 73 (2), 168-172 (2012).
  12. Duty, S., Jenner, P. Animal models of Parkinson’s disease: a source of novel treatments and clues to the cause of the disease. British Journal of Pharmacology. 164 (4), 1357-1391 (2011).
  13. Ungerstedt, U. 6-Hydroxy-dopamine induced degeneration of central monoamine neurons. European Journal of Pharmacology. 5 (1), 107-110 (1968).
  14. Ungerstedt, U. Postsynaptic supersensitivity after 6-hydroxy-dopamine induced degeneration of the nigro-striatal dopamine system. Acta Physiologica Scandinavica. Supplementum. 367, 69-93 (1971).
  15. Kirik, D., Rosenblad, C., Bjorklund, A. Characterization of behavioral and neurodegenerative changes following partial lesions of the nigrostriatal dopamine system induced by intrastriatal 6-hydroxydopamine in the rat. Experimental Neurology. 152, 259-277 (1998).
  16. Zahm, D. S. Compartments in rat dorsal and ventral striatum revealed following injection of 6-hydroxydopamine into the ventral mesencephalon. Brain Research. 552 (1), 164-169 (1991).
  17. Padovan-Neto, F. E., Echeverry, M. B., Tumas, V., Del-Bel, E. A. Nitric oxide synthase inhibition attenuates L-DOPA-induced dyskinesias in a rodent model of Parkinson’s disease. Neuroscience. 159 (3), 927-935 (2009).
  18. Padovan-Neto, F. E., et al. Antidyskinetic effect of the neuronal nitric oxide synthase inhibitor is linked to decrease of FosB/deltaFosB expression. Neuroscience Letters. 541, 126-131 (2013).
  19. Padovan-Neto, F. E., Cavalcanti-Kiwiatkoviski, R., Carolino, R. O., Anselmo-Franci, J., Del Bel, E. Effects of prolonged neuronal nitric oxide synthase inhibition on the development and expression of L-DOPA-induced dyskinesia in 6-OHDA-lesioned rats. Neuropharmacology. 89, 87-99 (2015).
  20. Winkler, C., Kirik, D., Björklund, A., Cenci, M. A. L-DOPA-induced dyskinesia in the intrastriatal 6-hydroxydopamine model of parkinson’s disease: relation to motor and cellular parameters of nigrostriatal function. Neurobiology of Disease. 10 (2), 165-186 (2002).
  21. Francardo, V., et al. Impact of the lesion procedure on the profiles of motor impairment and molecular responsiveness to l-DOPA in the 6-hydroxydopamine mouse model of Parkinson’s disease. Neurobiology of Disease. 42, 327-340 (2011).
  22. Lundblad, M., et al. Pharmacological validation of behavioural measures of akinesia and dyskinesia in a rat model of Parkinson’s disease. The European Journal of Neuroscience. 15 (1), 120-132 (2002).
  23. Cenci, M. A., Lee, C. S., Björklund, A. L-DOPA-induced dyskinesia in the rat is associated with striatal overexpression of prodynorphin- and glutamic acid decarboxylase mRNA. The European Journal of Neuroscience. 10 (8), 2694-2706 (1998).
  24. Henry, B., Crossman, A. R., Brotchie, J. M. Characterization of enhanced behavioral responses to L-DOPA following repeated administration in the 6-hydroxydopamine-lesioned rat model of Parkinson’s disease. Experimental Neurology. 151 (2), 334-342 (1998).
  25. Lee, C. S., Cenci, M. A., Schulzer, M., Björklund, A. Embryonic ventral mesencephalic grafts improve levodopa-induced dyskinesia in a rat model of Parkinson’s disease. Brain: A Journal of Neurology. 123, 1365-1379 (2000).
  26. Andersson, M., Hilbertson, A., Cenci, M. A. Striatal fosB expression is causally linked with l-DOPA-induced abnormal involuntary movements and the associated upregulation of striatal prodynorphin mRNA in a rat model of Parkinson’s disease. Neurobiology of Disease. 6 (6), 461-474 (1999).
  27. Lindgren, H. S., Rylander, D., Ohlin, K. E., Lundblad, M., Cenci, M. A. The “motor complication syndrome” in rats with 6-OHDA lesions treated chronically with L-DOPA: relation to dose and route of administration. Behavioural Brain Research. 177 (1), 150-159 (2007).
  28. Lane, E. L., Dunnett, S. . Animal Models of Movement Disorders: volume I, Neuromethods. 61, (2011).
  29. Carlsson, T., Carta, M., Winkler, C., Björklund, A., Kirik, D. Serotonin neuron transplants exacerbate l-DOPA-induced dyskinesias in a rat model of Parkinson’s disease. Journal of Neuroscience. 27 (30), 8011-8022 (2007).
  30. Lundblad, M., Picconi, B., Lindgren, H., Cenci, M. A. A model of L-DOPA-induced dyskinesia in 6-hydroxydopamine lesioned mice: relation to motor and cellular parameters of nigrostriatal function. Neurobiology of Disease. 16 (1), 110-123 (2004).
  31. Fasano, S., et al. Inhibition of Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1) signaling in the striatum reverts motor symptoms associated with L-dopa-induced dyskinesia. Proceedings of the National Academy of Sciences of the United States of America. 107 (50), 21824-21829 (2010).
  32. Berton, O., et al. Striatal overexpression of DeltaJunD resets L-DOPA-induced dyskinesia in a primate model of Parkinson disease. Biological Psychiatry. 66 (6), 554-561 (2009).
  33. Lindgren, H. S., et al. Putaminal upregulation of FosB/ΔFosB-like immunoreactivity in Parkinson’s disease patients with dyskinesia. Journal of Parkinson’s Disease. 1 (4), 347-357 (2011).
  34. Rylander, D., et al. Maladaptive plasticity of serotonin axon terminals in levodopa-induced dyskinesia. Annals of Neurology. 68 (5), 619-628 (2010).
  35. Smith, R., et al. The role of pallidal serotonergic function in Parkinson’s disease dyskinesias: a positron emission tomography study. Neurobiology of Aging. 36 (4), 1736-1742 (2015).
  36. Lundblad, M., et al. Pharmacological validation of a mouse model of l-DOPA-induced dyskinesia. Experimental Neurology. 194 (1), 66-75 (2005).
  37. Dekundy, A., Lundblad, M., Danysz, W., Cenci, M. A. Modulation of L-DOPA-induced abnormal involuntary movements by clinically tested compounds: further validation of the rat dyskinesia model. Behavioural Brain Research. 179 (1), 76-89 (2007).
  38. Blanchet, P. J., Konitsiotis, S., Chase, T. N. Amantadine reduces levodopa-induced dyskinesias in parkinsonian monkeys. Movement Disorders: Official Journal of the Movement Disorder Society. 13 (5), 798-802 (1998).
  39. Verhagen Metman, I., et al. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology. 50 (5), 1323-1326 (1998).
  40. Cenci, M. A., Lundblad, M. Ratings of L-DOPA-induced dyskinesia in the unilateral 6-OHDA lesion model of Parkinson’s disease in rats and mice. Current Protocols in Neuroscience. , 25 (2007).
  41. Dupre, K. B., et al. Effects of coincident 5- HT1A receptor stimulation and NMDA receptor antagonism on L-DOPA-induced dyskinesia and rotational behaviors in the hemi-parkinsonian rat. Psychopharmacology (Berl). 199 (1), 99-108 (2008).
  42. Guimarães, P. R., Bariotto-dos-Santos, K., Ribeiro, D. L., Padovan-Neto, F. E. The 6-hydroxydopamine (6-OHDA) rat model of Parkinson’s disease. Journal of Visualized Experiments: JoVE. , (2021).
  43. Paxinos, G., Watson, C. . The Rat Brain in Stereotaxic Coordinates. , (1998).
  44. Tseng, K. Y., et al. Inhibition of striatal soluble guanylyl cyclase-cGMP signaling reverses basal ganglia dysfunction and akinesia in experimental parkinsonism. PloS One. 6 (11), 27187 (2011).
  45. Padovan-Neto, F. E., et al. Selective regulation of 5-HT1B serotonin receptor expression in the striatum by dopamine depletion and repeated L-DOPA treatment: Relationship to L-DOPA-induced dyskinesias. Molecular Neurobiology. 57 (2), 736-751 (2020).
  46. Olsson, M., Nikkhah, G., Bentlage, C., Bjorklund, A. Forelimb akinesia in the rat Parkinson model: Differential effects of dopamine agonists and nigral transplants as assessed by a new stepping test. Journal of Neuroscience. 15, 3863-3875 (1995).
  47. Chang, J. W., Wachtel, S. R., Young, D., Kang, U. J. Biochemical and anatomical characterization of forepaw adjusting steps in rat models of Parkinson’s disease lesions. Neuroscience. 88, 617-628 (1995).
  48. Jayasinghe, V. R., Flores-Barrera, E., West, A. R., Tseng, K. Y. Frequency-dependent corticostriatal disinhibition resulting from chronic dopamine depletion: Role of local striatal cGMP and GABAAR signaling. Cerebral Cortex. 27, 625-634 (2017).
  49. Picconi, B., et al. Loss of bidirectional striatal synaptic plasticity in L-DOPA-induced dyskinesia. Nature Neuroscience. 6 (5), 501-506 (2003).
  50. Carta, M., et al. Role of striatal L-DOPA in the production of dyskinesia in 6-hydroxydopamine lesioned rats. Journal of Neurochemistry. 96 (6), 1718-1727 (2006).
  51. Westin, J. E., Andersson, M., Lundblad, M., Cenci, M. A. Persistent changes in striatal gene expression induced by long-term L-DOPA treatment in a rat model of Parkinson’s disease. The European Journal of Neuroscience. 14 (7), 1171-1176 (2001).
  52. Bariotto-Dos-Santos, K., et al. Repurposing an established drug: an emerging role for methylene blue in L-DOPA-induced dyskinesia. The European Journal of Neuroscience. 49 (6), 869-882 (2018).
  53. Lindgren, H. S., Lane, E. L., Lane, E., Dunnett, S. Rodent Models of l-DOPA-Induced Dyskinesia. Animal Models of Movement Disorders. Neuromethods. 61, (2011).
  54. Doucet, J., et al. Drug-drug interactions related to hospital admissions in older adults: A prospective study of 1000 patients. Journal of the American Geriatrics Society. 44, 944-948 (1996).
  55. Ohlin, K. E., et al. Vascular endothelial growth factor is upregulated by L-dopa in the parkinsonian brain: implications for the development of dyskinesia. Brain: A Journal of Neurology. 134, 2339-2357 (2011).
  56. Jourdain, V. A., et al. Increased putamen hypercapnic vasoreactivity in levodopa-induced dyskinesia. JCI insight. 2 (20), 96411 (2017).
  57. Thiele, S. L., et al. Generation of a model of l-DOPA-induced dyskinesia in two different mouse strains. Journal of Neuroscience Methods. 197 (2), 193-208 (2011).
  58. Dos-Santos-Pereira, M., et al. Contributive role of TNF-α to L-DOPA-induced dyskinesia in a unilateral 6-OHDA lesion model of Parkinson’s disease. Frontiers in Pharmacology. 11, 617085 (2021).
  59. Cenci, M. A., Ohlin, K. E., Odin, P. Current options and future possibilities for the treatment of dyskinesia and motor fluctuations in Parkinson’s disease. CNS & Neurological Disorders Drug Targets. 10 (6), 670-684 (2011).
  60. Cenci, M. A. Presynaptic mechanisms of l-DOPA-induced dyskinesia: The findings, the debate, and the therapeutic implications. Frontiers in Neurology. 5, 242 (2014).
  61. Langston, J. W., Quik, M., Petzinger, G., Jakowec, M., Di Monte, D. A. Investigating levodopa-induced dyskinesias in the parkinsonian primate. Annals of Neurology. 47 (4), 79-89 (2000).
  62. Chase, T. N. Levodopa therapy: consequences of the nonphysiologic replacement of dopamine. Neurology. 50 (5), 17-25 (1998).
check_url/62924?article_type=t

Play Video

Cite This Article
Bariotto-dos-Santos, K., Ribeiro, D. L., Guimarães, R. P., Padovan-Neto, F. E. Rating L-DOPA-Induced Dyskinesias in the Unilaterally 6-OHDA-Lesioned Rat Model of Parkinson’s Disease. J. Vis. Exp. (176), e62924, doi:10.3791/62924 (2021).

View Video