Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2020

01-12-2020 | Parkinson's Disease | Research

Angiotensin AT1 and AT2 receptor heteromer expression in the hemilesioned rat model of Parkinson’s disease that increases with levodopa-induced dyskinesia

Authors: Rafael Rivas-Santisteban, Ana I. Rodriguez-Perez, Ana Muñoz, Irene Reyes-Resina, José Luis Labandeira-García, Gemma Navarro, Rafael Franco

Published in: Journal of Neuroinflammation | Issue 1/2020

Login to get access

Abstract

Background/aims

The renin-angiotensin system (RAS) is altered in Parkinson’s disease (PD), a disease due to substantia nigra neurodegeneration and whose dopamine-replacement therapy, using the precursor levodopa, leads to dyskinesias as the main side effect. Angiotensin AT1 and AT2 receptors, mainly known for their role in regulating water homeostasis and blood pressure and able to form heterodimers (AT1/2Hets), are present in the central nervous system. We assessed the functionality and expression of AT1/2Hets in Parkinson disease (PD).

Methods

Immunocytochemistry was used to analyze the colocalization between angiotensin receptors; bioluminescence resonance energy transfer was used to detect AT1/2Hets. Calcium and cAMP determination, MAPK activation, and label-free assays were performed to characterize signaling in homologous and heterologous systems. Proximity ligation assays were used to quantify receptor expression in mouse primary cultures and in rat striatal sections.

Results

We confirmed that AT1 and AT2 receptors form AT1/2Hets that are expressed in cells of the central nervous system. AT1/2Hets are novel functional units with particular signaling properties. Importantly, the coactivation of the two receptors in the heteromer reduces the signaling output of angiotensin. Remarkably, AT1/2Hets that are expressed in both striatal neurons and microglia make possible that candesartan, the antagonist of AT1, increases the effect of AT2 receptor agonists. In addition, the level of striatal expression increased in the unilateral 6-OH-dopamine lesioned rat PD model and was markedly higher in parkinsonian-like animals that did not become dyskinetic upon levodopa chronic administration if compared with expression in those that became dyskinetic.

Conclusion

The results indicate that boosting the action of neuroprotective AT2 receptors using an AT1 receptor antagonist constitutes a promising therapeutic strategy in PD.
Appendix
Available only for authorised users
Literature
5.
go back to reference Benito C, Núñez E, Tolón RM, Carrier EJ, Rábano A, Hillard CJ, Romero J. Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively overexpressed in neuritic plaque-associated glia in Alzheimer’s disease brains. J Neurosci. 2003;23:11136–41.CrossRef Benito C, Núñez E, Tolón RM, Carrier EJ, Rábano A, Hillard CJ, Romero J. Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively overexpressed in neuritic plaque-associated glia in Alzheimer’s disease brains. J Neurosci. 2003;23:11136–41.CrossRef
9.
go back to reference Birkmayer W, Hornykiewicz O. Additional experimental studies on L-DOPA in Parkinson’s syndrome and reserpine parkinsonism. Arch Psychiatr Nervenkr. 1964;206:367–81.CrossRef Birkmayer W, Hornykiewicz O. Additional experimental studies on L-DOPA in Parkinson’s syndrome and reserpine parkinsonism. Arch Psychiatr Nervenkr. 1964;206:367–81.CrossRef
12.
13.
go back to reference Farré D, Muñoz A, Moreno E, Reyes-Resina I, Canet-Pons J, Dopeso-Reyes IG, Rico AJ, Lluís C, Mallol J, Navarro G, Canela EI, Cortés A, Labandeira-García JL, Casadó V, Lanciego JL, Franco R. Stronger dopamine D1 receptor-mediated neurotransmission in dyskinesia. Mol Neurobiol. 2015;52:1408–20. https://doi.org/10.1007/s12035-014-8936-x.CrossRefPubMed Farré D, Muñoz A, Moreno E, Reyes-Resina I, Canet-Pons J, Dopeso-Reyes IG, Rico AJ, Lluís C, Mallol J, Navarro G, Canela EI, Cortés A, Labandeira-García JL, Casadó V, Lanciego JL, Franco R. Stronger dopamine D1 receptor-mediated neurotransmission in dyskinesia. Mol Neurobiol. 2015;52:1408–20. https://​doi.​org/​10.​1007/​s12035-014-8936-x.CrossRefPubMed
14.
go back to reference Ferrão FM, Lara LS, Axelband F, Dias J, Carmona AK, Reis RI, Costa-Neto CM, Vieyra A, Lowe J. Exposure of luminal membranes of LLC-PK 1 cells to ANG II induces dimerization of AT 1 /AT 2 receptors to activate SERCA and to promote Ca 2+ mobilization. Am J Physiol Physiol. 2012;302:F875–83. https://doi.org/10.1152/ajprenal.00381.2011.CrossRef Ferrão FM, Lara LS, Axelband F, Dias J, Carmona AK, Reis RI, Costa-Neto CM, Vieyra A, Lowe J. Exposure of luminal membranes of LLC-PK 1 cells to ANG II induces dimerization of AT 1 /AT 2 receptors to activate SERCA and to promote Ca 2+ mobilization. Am J Physiol Physiol. 2012;302:F875–83. https://​doi.​org/​10.​1152/​ajprenal.​00381.​2011.CrossRef
18.
go back to reference Franco, R., Navarro, G., Rivas Santisteban, R., Awad Alkozi, H., 2019. Potency of melatonin at G-protein-coupled MT1 and MT2 receptors. Available at: osf.io/w7qxh. Franco, R., Navarro, G., Rivas Santisteban, R., Awad Alkozi, H., 2019. Potency of melatonin at G-protein-coupled MT1 and MT2 receptors. Available at: osf.​io/​w7qxh.
20.
23.
go back to reference Hinz S, Navarro G, Borroto-Escuela D, Seibt BF, Ammon C, De Filippo E, Danish A, Lacher SK, Červinková B, Rafehi M, Fuxe K, Schiedel AC, Franco R, Müller CE, Ammon YC, de Filippo E, Danish A, Lacher SK, Červinková B, Rafehi M, Fuxe K, Schiedel AC, Franco R, Müller CE, Ammon C, De Filippo E, Danish A, Lacher SK, Červinková B, Rafehi M, Fuxe K, Schiedel AC, Franco R, Müller CE. Adenosine A2A receptor ligand recognition and signaling is blocked by A2B receptors. Oncotarget. 2018;9:13593–611. https://doi.org/10.18632/oncotarget.24423.CrossRefPubMedPubMedCentral Hinz S, Navarro G, Borroto-Escuela D, Seibt BF, Ammon C, De Filippo E, Danish A, Lacher SK, Červinková B, Rafehi M, Fuxe K, Schiedel AC, Franco R, Müller CE, Ammon YC, de Filippo E, Danish A, Lacher SK, Červinková B, Rafehi M, Fuxe K, Schiedel AC, Franco R, Müller CE, Ammon C, De Filippo E, Danish A, Lacher SK, Červinková B, Rafehi M, Fuxe K, Schiedel AC, Franco R, Müller CE. Adenosine A2A receptor ligand recognition and signaling is blocked by A2B receptors. Oncotarget. 2018;9:13593–611. https://​doi.​org/​10.​18632/​oncotarget.​24423.CrossRefPubMedPubMedCentral
28.
go back to reference Kirik D, Winkler C, Björklund A. Growth and functional efficacy of intrastriatal nigral transplants depend on the extent of nigrostriatal degeneration. J Neurosci. 2001;21:2889–96. Kirik D, Winkler C, Björklund A. Growth and functional efficacy of intrastriatal nigral transplants depend on the extent of nigrostriatal degeneration. J Neurosci. 2001;21:2889–96.
35.
go back to reference Leonhardt J, Villela DC, Teichmann A, Münter L-M, Mayer MC, Mardahl M, Kirsch S, Namsolleck P, Lucht K, Benz V, Alenina N, Daniell N, Horiuchi M, Iwai M, Multhaup G, Schülein R, Bader M, Santos RA, Unger T, Steckelings UM. Evidence for Heterodimerization and Functional Interaction of the Angiotensin Type 2 Receptor and the Receptor MASNovelty and Significance. Hypertension. 2017;69:1128–35. https://doi.org/10.1161/HYPERTENSIONAHA.116.08814.CrossRefPubMed Leonhardt J, Villela DC, Teichmann A, Münter L-M, Mayer MC, Mardahl M, Kirsch S, Namsolleck P, Lucht K, Benz V, Alenina N, Daniell N, Horiuchi M, Iwai M, Multhaup G, Schülein R, Bader M, Santos RA, Unger T, Steckelings UM. Evidence for Heterodimerization and Functional Interaction of the Angiotensin Type 2 Receptor and the Receptor MASNovelty and Significance. Hypertension. 2017;69:1128–35. https://​doi.​org/​10.​1161/​HYPERTENSIONAHA.​116.​08814.CrossRefPubMed
39.
go back to reference Navarro G, Borroto-Escuela D, Angelats E, Etayo I, Reyes-Resina I, Pulido-Salgado M, Rodríguez-Pérez AAI, Canela EIE, Saura J, Lanciego JJL, Labandeira-García JLJ, Saura CA, Fuxe K, Franco R. Receptor-heteromer mediated regulation of endocannabinoid signaling in activated microglia. Relevance for Alzheimer’s disease and levo-dopa-induced dyskinesia. Brain Behav Immun. 2018;67:139–51. https://doi.org/10.1016/j.bbi.2017.08.015.CrossRefPubMed Navarro G, Borroto-Escuela D, Angelats E, Etayo I, Reyes-Resina I, Pulido-Salgado M, Rodríguez-Pérez AAI, Canela EIE, Saura J, Lanciego JJL, Labandeira-García JLJ, Saura CA, Fuxe K, Franco R. Receptor-heteromer mediated regulation of endocannabinoid signaling in activated microglia. Relevance for Alzheimer’s disease and levo-dopa-induced dyskinesia. Brain Behav Immun. 2018;67:139–51. https://​doi.​org/​10.​1016/​j.​bbi.​2017.​08.​015.CrossRefPubMed
42.
go back to reference Olanow CW, Agid Y, Mizuno Y, Albanese A, Bonuccelli U, Bonucelli U, Damier P, De Yebenes J, Gershanik O, Guttman M, Grandas F, Hallett M, Hornykiewicz O, Jenner P, Katzenschlager R, Langston WJ, LeWitt P, Melamed E, Mena MA, Michel PP, Mytilineou C, Obeso JA, Poewe W, Quinn N, Raisman-Vozari R, Rajput AH, Rascol O, Sampaio C, Stocchi F. Levodopa in the treatment of Parkinson’s disease: current controversies. Mov Disord. 2004;19:997–1005. https://doi.org/10.1002/mds.20243.CrossRefPubMed Olanow CW, Agid Y, Mizuno Y, Albanese A, Bonuccelli U, Bonucelli U, Damier P, De Yebenes J, Gershanik O, Guttman M, Grandas F, Hallett M, Hornykiewicz O, Jenner P, Katzenschlager R, Langston WJ, LeWitt P, Melamed E, Mena MA, Michel PP, Mytilineou C, Obeso JA, Poewe W, Quinn N, Raisman-Vozari R, Rajput AH, Rascol O, Sampaio C, Stocchi F. Levodopa in the treatment of Parkinson’s disease: current controversies. Mov Disord. 2004;19:997–1005. https://​doi.​org/​10.​1002/​mds.​20243.CrossRefPubMed
47.
go back to reference Pinna A, Bonaventura J, Farré D, Sánchez M, Simola N, Mallol J, Lluís C, Costa G, Baqi Y, Müller CE, Cortés A, McCormick P, Canela EI, Martínez-Pinilla E, Lanciego JL, Casadó V, Armentero M-TT, Franco R. l-DOPA disrupts adenosine A2A–cannabinoid CB1–dopamine D2 receptor heteromer cross-talk in the striatum of hemiparkinsonian rats: Biochemical and behavioral studies. Exp Neurol. 2014;253:180–91. https://doi.org/10.1016/j.expneurol.2013.12.021.CrossRefPubMed Pinna A, Bonaventura J, Farré D, Sánchez M, Simola N, Mallol J, Lluís C, Costa G, Baqi Y, Müller CE, Cortés A, McCormick P, Canela EI, Martínez-Pinilla E, Lanciego JL, Casadó V, Armentero M-TT, Franco R. l-DOPA disrupts adenosine A2A–cannabinoid CB1–dopamine D2 receptor heteromer cross-talk in the striatum of hemiparkinsonian rats: Biochemical and behavioral studies. Exp Neurol. 2014;253:180–91. https://​doi.​org/​10.​1016/​j.​expneurol.​2013.​12.​021.CrossRefPubMed
57.
go back to reference Schallert T, Kozlowski DA, Humm JL, Cocke R. Use-dependent structural events in recovery of function. Adv Neurol. 1997;73:229–38.PubMed Schallert T, Kozlowski DA, Humm JL, Cocke R. Use-dependent structural events in recovery of function. Adv Neurol. 1997;73:229–38.PubMed
62.
go back to reference Valero-Esquitino V, Lucht K, Namsolleck P, Monnet-Tschudi F, Stubbe T, Lucht F, Liu M, Ebner F, Brandt C, Danyel LA, Villela DC, Paulis L, Thoene-Reineke C, Dahlöf B, Hallberg A, Unger T, Sumners C, Steckelings UM. Direct angiotensin type 2 receptor (AT 2 R) stimulation attenuates T-cell and microglia activation and prevents demyelination in experimental autoimmune encephalomyelitis in mice. Clin Sci. 2015;128:95–109. https://doi.org/10.1042/CS20130601.CrossRefPubMed Valero-Esquitino V, Lucht K, Namsolleck P, Monnet-Tschudi F, Stubbe T, Lucht F, Liu M, Ebner F, Brandt C, Danyel LA, Villela DC, Paulis L, Thoene-Reineke C, Dahlöf B, Hallberg A, Unger T, Sumners C, Steckelings UM. Direct angiotensin type 2 receptor (AT 2 R) stimulation attenuates T-cell and microglia activation and prevents demyelination in experimental autoimmune encephalomyelitis in mice. Clin Sci. 2015;128:95–109. https://​doi.​org/​10.​1042/​CS20130601.CrossRefPubMed
Metadata
Title
Angiotensin AT1 and AT2 receptor heteromer expression in the hemilesioned rat model of Parkinson’s disease that increases with levodopa-induced dyskinesia
Authors
Rafael Rivas-Santisteban
Ana I. Rodriguez-Perez
Ana Muñoz
Irene Reyes-Resina
José Luis Labandeira-García
Gemma Navarro
Rafael Franco
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2020
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-020-01908-z

Other articles of this Issue 1/2020

Journal of Neuroinflammation 1/2020 Go to the issue