Skip to main content

Advertisement

Log in

Current disease modifying approaches to treat Parkinson’s disease

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Parkinson’s disease (PD is a progressive neurological disorder characterized by the degeneration and death of midbrain dopamine and non-dopamine neurons in the brain leading to motor dysfunctions and other symptoms, which seriously influence the quality of life of PD patients. The drug L-dopa can alleviate the motor symptoms in PD, but so far there are no rational therapies targeting the underlying neurodegenerative processes. Despite intensive research, the molecular mechanisms causing neuronal loss are not fully understood which has hampered the development of new drugs and disease-modifying therapies. Neurotrophic factors are by virtue of their survival promoting activities attract candidates to counteract and possibly halt cell degeneration in PD. In particular, studies employing glial cell line-derived neurotrophic factor (GDNF) and its family member neurturin (NRTN), as well as the recently described cerebral dopamine neurotrophic factor (CDNF) and the mesencephalic astrocyte-derived neurotrophic factor (MANF) have shown positive results in protecting and repairing dopaminergic neurons in various models of PD. Other substances with trophic actions in dopaminergic neurons include neuropeptides and small compounds that target different pathways impaired in PD, such as increased cell stress, protein handling defects, dysfunctional mitochondria and neuroinflammation. In this review, we will highlight the recent developments in this field with a focus on trophic factors and substances having the potential to beneficially influence the viability and functions of dopaminergic neurons as shown in preclinical or in animal models of PD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Klein C, Westenberger A (2012) Genetics of Parkinson’s disease. Cold Spring Harb Perspect Med. 2:a008888

    Article  PubMed  PubMed Central  Google Scholar 

  2. Mullin S, Schapira A (2015) The genetics of Parkinson’s disease. Br Med Bull 114:39–52

    Article  PubMed  Google Scholar 

  3. Henchcliffe C, Beal MF (2008) Mitochondrial biology and oxidative stress in Parkinson disease pathogenesis. Nat Clin Pract Neurol 4:600–609

    Article  CAS  PubMed  Google Scholar 

  4. Gupta A, Dawson VL, Dawson TM (2008) What causes cell death in Parkinson’s disease? Ann Neurol 64(Suppl 2):S3–S15

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Perier C, Vila M (2012) Mitochondrial biology and Parkinson’s disease. Cold Spring Harb Perspect Med 2:a009332

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Guzman JN, Sanchez-Padilla J, Wokosin D, Kondapalli J, Ilijic E, Schumacker PT, Surmeier DJ (2010) Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature 468:696–700

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pacelli C, Giguere N, Bourque M-J, Lévesque M, Slack RS, Trudeau LE (2015) Elevated mitochondrial mioenergetics and axonal arborization size are key contributors to the vulnerability of dopamine neurons. Curr Biol 25:2349–2360. doi:10.1016/j.cub.2015.07.050

    Article  CAS  PubMed  Google Scholar 

  8. Goedert M, Spillantini MG, Del Tredici K, Braak H (2013) 100 years of Lewy pathology. Nat Rev Neurol 9:13–24

    Article  CAS  PubMed  Google Scholar 

  9. Brundin P, Atkin G, Lamberts JT (2015) Basic science breaks through: new therapeutic advances in Parkinson’s disease. Mov Disord 30:1521–1527. doi:10.1002/mds.26332

    Article  PubMed  Google Scholar 

  10. Betzer C, Movius AJ, Shi M, Gai WP, Zhang J, Jensen PH (2015) Identification of synaptosomal proteins binding to monomeric and oligomeric α-synuclein. PLoS One 10:e0116473

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D (2004) Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 305:1292–1295

    Article  CAS  PubMed  Google Scholar 

  12. Chu Y, Dodiya H, Aebischer P, Olanow CW, Kordower JH (2009) Alterations in lysosomal and proteasomal markers in Parkinson’s disease: relationship to alpha-synuclein inclusions. Neurobiol Dis 35:385–398

    Article  CAS  PubMed  Google Scholar 

  13. Decressac M, Mattsson B, Weikop P, Lundblad M, Jakobsson J, Björklund A (2013) TFEB-mediated autophagy rescues midbrain dopamine neurons from α-synuclein toxicity. Proc Natl Acad Sci U S A 110:E1817–E1826

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Schapira AH (2015) Glucocerebrosidase and Parkinson disease: recent advances. Mol Cell Neurosci 66:37–42

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gegg ME, Burke D, Heales SJ, Cooper JM, Hardy J, Wood NW, Schapira AH (2012) Glucocerebrosidase deficiency in substantia nigra of parkinson disease brains. Ann Neurol 72:455–463

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gegg ME, Sweet L, Wang BH, Shihabuddin LS, Sardi SP, Schapira AH (2015) No evidence for substrate accumulation in Parkinson brains with GBA mutations. Mov Disord 30:1085–1089

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, Kolodziej P, Kahn I, Saftig P, Woulfe J, Rochet JC, Glicksman MA, Cheng SH, Grabowski GA, Shihabuddin LS, Schlossmacher MG (2011) Acid β-glucosidase mutants linked to Gaucher disease, Parkinson disease, and Lewy body dementia alter α-synuclein processing. Ann Neurol 69:940–953

    Article  CAS  PubMed  Google Scholar 

  18. Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN, Braak E (2003) Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 24:197–211

    Article  PubMed  Google Scholar 

  19. Braak H, Bohl JR, Müller CM, Rüb U, de Vos RA, Del Tredici K (2006) Stanley Fahn Lecture 2005: the staging procedure for the inclusion body pathology associated with sporadic Parkinson’s disease reconsidered. Mov Disord 21:2042–2051

    Article  PubMed  Google Scholar 

  20. Dehay B, Vila M, Bezard E, Brundin P, Kordower JH (2015) Alpha-synuclein propagation: New insights from animal models. Mov Disord. doi:10.1002/mds.26370

  21. Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, Haapaniemi E, Kaakkola S, Eerola-Rautio J, Pohja M, Kinnunen E, Murros K, Auvinen P (2015) Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord 30:350–358

    Article  PubMed  Google Scholar 

  22. Schapira AH, Olanow CW, Greenamyre JT, Bezard E (2014) Slowing of neurodegeneration in Parkinson’s disease and Huntington’s disease: future therapeutic perspectives. Lancet 384:545–555

    Article  CAS  PubMed  Google Scholar 

  23. Lindström V, Ihse E, Fagerqvist T, Bergström J, Nordström E, Möller C, Lannfelt L, Ingelsson M (2014) Immunotherapy targeting α-synuclein, with relevance for future treatment of Parkinson’s disease and other Lewy body disorders. Immunotherapy 6:141–153

    Article  PubMed  CAS  Google Scholar 

  24. Walter P, Ron D (2011) The unfolded protein response: from stress pathway to homeostatic regulation. Science 334:1081–1086

    Article  CAS  PubMed  Google Scholar 

  25. Lindholm D, Wootz H, Korhonen L (2006) ER stress and neurodegenerative diseases. Cell Death Differ 13:385–392

    Article  CAS  PubMed  Google Scholar 

  26. Hetz C, Mollereau B (2014) Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci 15:233–249

    Article  CAS  PubMed  Google Scholar 

  27. Ryu EJ, Harding HP, Angelastro JM, Vitolo OV, Ron D, Greene LA (2002) Endoplasmic reticulum stress and the unfolded protein response in cellular models of Parkinson’s disease. J Neurosci 22:10690–10698

    CAS  PubMed  Google Scholar 

  28. Baek JH, Whitfield D, Howlett D, Francis P, Bereczki E, Ballard C, Hortobágyi T, Attems J, Aarsland D (2015) Unfolded protein response is activated in Lewy body dementias. Neuropathol Appl Neurobiol. doi:10.1111/nan.12260

    PubMed  Google Scholar 

  29. Chung CY, Khurana V, Auluck PK, Tardiff DF, Mazzulli JR, Soldner F, Baru V, Lou Y, Freyzon Y, Cho S, Mungenast AE, Muffat J, Mitalipova M, Pluth MD, Jui NT, Schüle B, Lippard SJ, Tsai LH, Krainc D, Buchwald SL, Jaenisch R, Lindquist S (2013) Identification and rescue of α-synuclein toxicity in Parkinson patient-derived neurons. Science 342:983–987

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mercado G, Valdés P, Hetz C (2013) An ERcentric view of Parkinson’s disease. Trends Mol Med 19:165–715

    Article  CAS  PubMed  Google Scholar 

  31. Mercado G, Castillo V, Vidal R, Hetz C (2015) ER proteostasis disturbances in Parkinson’s disease: novel insights. Front Aging Neurosci. 7:39

    Article  PubMed  PubMed Central  Google Scholar 

  32. Imai Y, Takahashi R (2004) How do Parkin mutations result in neurodegeneration? Curr Opin Neurobiol 14:384–389

    Article  CAS  PubMed  Google Scholar 

  33. Kitao Y, Imai Y, Ozawa K, Kataoka A, Ikeda T, Soda M, Nakimawa K, Kiyama H, Stern DM, Hori O, Wakamatsu K, Ito S, Itohara S, Takahashi R, Ogawa S (2007) Pael receptor induces death of dopaminergic neurons in the substantia nigra via endoplasmic reticulum stress and dopamine toxicity, which is enhanced under condition of parkin inactivation. Hum Mol Genet 16:50–60

    Article  CAS  PubMed  Google Scholar 

  34. Thayanidhi N, Helm JR, Nycz DC, Bentley M, Liang Y, Hay JC (2010) Alpha-synuclein delays endoplasmic reticulum (ER)-to-Golgi transport in mammalian cells by antagonizing ER/Golgi SNAREs. Mol Biol Cell 21:1850–1863

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Colla E, Coune P, Liu Y, Pletnikova O, Troncoso JC, Iwatsubo T, Schneider BL, Lee MK (2012) Endoplasmic reticulum stress is important for the manifestations of α-synucleinopathy in vivo. J Neurosci 32:3306–3320

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hayashi T, Rizzuto R, Hajnoczky G, Su TP (2009) MAM: more than just a housekeeper. Trends Cell Biol 19:81–88

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Guardia-Laguarta C, Area-Gomez E, Rüb C, Liu Y, Magrané J, Becker D, Voos W, Schon EA, Przedborski S (2014) α-Synuclein is localized to mitochondria-associated ER membranes. J Neurosci 34:249–259

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Francardo V, Bez F, Wieloch T, Nissbrandt H, Ruscher K, Cenci MA (2014) Pharmacological stimulation of sigma-1 receptors has neurorestorative effects in experimental parkinsonism. Brain 137:1998–2014

    Article  PubMed  Google Scholar 

  39. Hyrskyluoto A, Pulli I, Törnqvist K, Ho TH, Korhonen L, Lindholm D (2013) Sigma-1 receptor agonist PRE084 is protective against mutant huntingtin-induced cell degeneration: involvement of calpastatin and the NF-κB pathway. Cell Death Dis 4:e646

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Nunnari J, Suomalainen A (2012) Mitochondria: in sickness and in health. Cell 148:1145–1159

    Article  CAS  PubMed  Google Scholar 

  41. Rugarli EI, Langer T (2012) Mitochondrial quality control: a matter of life and death for neurons. EMBO J 31:1336–1349

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kalia LV, Kalia SK, Lang AE (2015) Disease-modifying strategies for Parkinson’s disease. Mov Disord. doi:10.1002/mds.26354

  43. Puigserver P, Spiegelman BM (2003) Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulator. Endocr Rev 24:78–90

    Article  CAS  PubMed  Google Scholar 

  44. Houten SM, Auwerx J (2004) PGC-1alpha: turbocharging mitochondria. Cell 119:5–7

    Article  CAS  PubMed  Google Scholar 

  45. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jäger S, Handschin C, Zheng K, Lin J, Yang W, Simon DK, Bachoo R, Spiegelman BM (2006) Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 127:397–408

    Article  CAS  PubMed  Google Scholar 

  46. Mudò G, Mäkelä J, Di Liberto V, Tselykh TV, Olivieri M, Piepponen P, Eriksson O, Mälkiä A, Bonomo A, Kairisalo M, Aguirre JA, Korhonen L, Belluardo N, Lindholm D (2012) Transgenic expression and activation of PGC-1α protect dopaminergic neurons in the MPTP mouse model of Parkinson’s disease. Cell Mol Life Sci 69:1153–1165

    Article  PubMed  CAS  Google Scholar 

  47. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA, Grünblatt E, Moran LB, Mandel SA, Riederer P, Miller RM, Federoff HJ, Wüllner U, Papapetropoulos S, Youdim MB, Cantuti-Castelvetri I, Young AB, Vance JM, Davis RL, Hedreen JC, Adler CH, Beach TG, Graeber MB, Middleton FA, Rochet JC, Scherzer CR, Global PD Gene Expression (GPEX) Consortium (2010) PGC-1α, a potential therapeutic target for early intervention in Parkinson’s disease. Sci Transl Med 2: 52ra73

  48. Ciron C, Lengacher S, Dusonchet J, Aebischer P, Schneider BL (2012) Sustained expression ofPGC-1a in the rat nigrostriatal system selectively impairs dopaminergic function. Hum Mol Genet 21:1861–1876

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Clark J, Silvaggi JM, Kiselak T, Zheng K, Clore EL, DaiY Bass CE, Simon DK (2012) Pgc-1a overexpression downregulates Pitx3 and increases susceptibility to MPTP toxicity associated with decreased Bdnf. PLoS One 7:e48925

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lindholm D, Eriksson O, Makela J, Belluardo N, Korhonen L (2012) PGC-1alpha: a master gene that is hard to master. Cell Mol Life Sci 69:2465–2468

    Article  CAS  PubMed  Google Scholar 

  51. Soyal SM, Felder TK, Auer S, Hahne P, Oberkofler H, Witting A, Paulmichl M, Landwehrmeyer GB, Weydt P, Patsch W, Network European Huntington Disease (2012) A greatly extended PPARGC1A genomic locus encodes several new brain-specific isoforms and influences Huntington disease age of onset. Hum Mol Genet 21:3461–3473

    Article  CAS  PubMed  Google Scholar 

  52. Aviles-Olmos I, Limousin P, Lees A, Foltynie T (2013) Parkinson’s disease, insulin resistance and novel agents of neuroprotection. Brain 136:374–384

    Article  PubMed  Google Scholar 

  53. Patrone C, Eriksson O, Lindholm D (2014) Diabetes drugs and neurological disorders: new views and therapeutic possibilities. Lancet Diabetes Endocrinol 2:256–262

    Article  CAS  PubMed  Google Scholar 

  54. Schintu N, Frau L, Ibba M, Caboni P, Garau A, Carboni E, Carta AR (2009) PPAR-gamma-mediated neuroprotection in a chronic mouse model of Parkinson’s disease. Eur J Neurosci 29:954–963

    Article  PubMed  Google Scholar 

  55. Carta AR, Frau L, Pisanu A, Wardas J, Spiga S, Carboni E (2011) Rosiglitazone decreases peroxisome proliferator receptor-gamma levels in microglia and inhibits TNF-alpha production: new evidences on neuroprotection in a progressive Parkinson’s disease model. Neuroscience 194:250–261

    Article  CAS  PubMed  Google Scholar 

  56. Swanson CR, Joers V, Bondarenko V, Brunner K, Simmons HA, Ziegler TE, Kemnitz JW, Johnson JA, Emborg ME (2011) The PPAR-γ agonist pioglitazone modulates inflammation and induces neuroprotection in parkinsonian monkeys. J Neuroinflammation 8:91. doi:10.1186/1742-2094-8-91

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Brauer R, Bhaskaran K, Chaturvedi N, Dexter DT, Smeeth L, Douglas I (2015) Glitazone treatment and incidence of Parkinson’s disease among people with diabetes: a retrospective cohort study. PLoS Med 12:e1001854

    Article  PubMed  PubMed Central  Google Scholar 

  58. NINDS Exploratory Trials in Parkinson Disease (NET-PD) FS-ZONE Investigators (2015) Pioglitazone in early Parkinson’s disease: a phase 2, multicentre, double-blind, randomised trial. Lancet Neurol 14:795–803

    Article  CAS  Google Scholar 

  59. Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, Powers K, Shen H, Egan JM, Sambamurti K, Brossi A, Lahiri DK, Mattson MP, Hoffer BJ, Wang Y, Greig NH (2009) GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci USA 106:1285–1290

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Ell P, Soderlund T, Whitton P, Wyse R, Isaacs T, Lees A, Limousin P, Foltynie T (2013) Exenatide and the treatment of patients with Parkinson’s disease. J Clin Invest 123:2730–2736

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Kahan J, Ell P, Whitton P, Wyse R, Isaacs T, Lees A, Limousin P, Foltynie T (2014) Motor and cognitive advantages persist 12 months after exenatide exposure in Parkinson’s disease. J Parkinsons Dis 4:337–344

    Article  CAS  PubMed  Google Scholar 

  62. McGeer PL, Itagaki S, Boyes BE, McGeer EG (1988) Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 38:1285–1291

    Article  CAS  PubMed  Google Scholar 

  63. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, Eggert K, Oertel W, Banati RB, Brooks DJ (2006) In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol Dis 21:404–412

    Article  CAS  PubMed  Google Scholar 

  64. Ouchi Y, Yagi S, Yokokura M, Sakamoto M (2009) Neuroinflammation in the living brain of Parkinson’s disease. Parkinsonism Relat Disord 15:S200–S204

    Article  PubMed  Google Scholar 

  65. Sica A, Mantovani A (2012) Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 122:787–795

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kwilasz AJ, Grace PM, Serbedzija P, Maier SF, Watkins LR (2015) The therapeutic potential of interleukin-10 in neuroimmune diseases. Neuropharmacology 96:55–69

    Article  CAS  PubMed  Google Scholar 

  67. Fernandes A, Miller-Fleming L, Pais TF (2014) Microglia and inflammation: conspiracy, controversy or control? Cell Mol Life Sci 71:3969–3985

    Article  CAS  PubMed  Google Scholar 

  68. Bakshi R, Zhang H, Logan R, Joshi I, Xu Y, Chen X, Schwarzschild MA (2015) Neuroprotective effects of urate are mediated by augmenting astrocytic glutathione synthesis and release. Neurobiol Dis. doi:10.1016/j.nbd.2015.08.022

    PubMed  Google Scholar 

  69. Weisskopf MG, O’Reilly E, Chen H, Schwarzschild MA, Ascherio A (2007) Plasma urate and risk of Parkinson’s disease. Am J Epidemiol 166:561–567

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ascherio A, LeWitt PA, Xu K, Eberly S, Watts A, Matson WR, Marras C, Kieburtz K, Rudolph A, Bogdanov MB, Schwid SR, Tennis M, Tanner CM, Beal MF, Lang AE, Oakes D, Fahn S, Shoulson I, Schwarzschild MA, Parkinson Study Group DATATOP Investigators (2009) Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol 66:1460–1468

  71. Parkinson Study Group SURE-PD Investigators, Schwarzschild MA, Ascherio A, Beal MF, Cudkowicz ME, Curhan GC, Hare JM, Hooper DC, Kieburtz KD, Macklin EA, Oakes D, Rudolph A, Shoulson I, Tennis MK, Espay AJ, Gartner M, Hung A, Bwala G, Lenehan R, Encarnacion E, Ainslie M, Castillo R, Togasaki D, Barles G, Friedman JH, Niles L, Carter JH, Murray M, Goetz CG, Jaglin J, Ahmed A, Russell DS, Cotto C, Goudreau JL, Russell D, Parashos SA, Ede P, Saint-Hilaire MH, Thomas CA, James R, Stacy MA, Johnson J, Gauger L, Antonelle de Marcaida J, Thurlow S, Isaacson SH, Carvajal L, Rao J, Cook M, Hope-Porche C, McClurg L, Grasso DL, Logan R, Orme C, Ross T, Brocht AF, Constantinescu R, Sharma S, Venuto C, Weber J, Eaton K (2014) Inosine to increase serum and cerebrospinal fluid urate in Parkinson disease: a randomized clinical trial. JAMA Neurol 71:141–150

  72. Schwarzschild MA, Macklin EA, Ascherio A, Parkinson Study Group SURE-PD Investigators (2014) Urate and neuroprotection trials. Lancet Neurol 13:758

  73. Chamorro A, Amaro S, Castellanos M, Segura T, Arenillas J, Martí-Fábregas J, Gállego J, Krupinski J, Gomis M, Cánovas D, Carné X, Deulofeu R, Román LS, Oleaga L, Torres F, Planas AM, Investigators URICO-ICTUS (2014) Safety and efficacy of uric acid in patients with acute stroke (URICO-ICTUS): a randomised, double-blind phase 2b/3 trial. Lancet Neurol 13:453–460

    Article  CAS  PubMed  Google Scholar 

  74. Lin LF, Doherty DH, Lile JD, Bektesh S, Collins F (1993) GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 260:1130–1132

    Article  CAS  PubMed  Google Scholar 

  75. Airaksinen MS, Saarma M (2002) The GDNF family: signalling, biological functions and therapeutic value. Nat Rev Neurosci 3:383–394

    Article  CAS  PubMed  Google Scholar 

  76. Domanskyi A, Saarma M, Airavaara M (2015) Prospects of neurotrophic factors for Parkinson’s disease: comparison of protein and gene therapy. Hum Gene Ther 26:550–559

    Article  CAS  PubMed  Google Scholar 

  77. Pascual A, Hidalgo-Figueroa M, Piruat JI, Pintado CO, Gómez-Díaz R, López-Barneo J (2008) Absolute requirement of GDNF for adult catecholaminergic neuron survival. Nat Neurosci 11:755–761

    Article  CAS  PubMed  Google Scholar 

  78. Kopra J, Vilenius C, Grealish S, Harma M-A, Varendi K, Lindholm J, Castren E, Voikar V, Bjorklund A, Piepponen TP, Saarma M, Andressoo J-O (2015) GDNF is not required for catecholaminergic neuron survival in vivo. Nat Neurosci 18:319–322

    Article  CAS  PubMed  Google Scholar 

  79. Kramer ER, Aron L, Ramakers GM, Seitz S, Zhuang X, Beyer K, Smidt MP, Klein R (2007) Absence of Ret signaling in mice causes progressive and late degeneration of the nigrostriatal system. PLoS Biol 5:e39

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Aron L, Klein R (2011) Repairing the parkinsonian brain with neurotrophic factors. Trends Neurosci 34:88–100

    Article  CAS  PubMed  Google Scholar 

  81. Decressac M, Kadkhodaei B, Mattsson B, Laguna A, Perlmann T, Björklund A (2012) α-Synuclein-induced down-regulation of Nurr1 disrupts GDNF signaling in nigral dopamine neurons. Sci Transl Med 4:163ra156

  82. Bartus RT, Weinberg MS, Samulski RJ (2014) Parkinson’s disease gene therapy: successby design meets failure by efficacy. Mol Ther 22:487–497

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Nutt JG, Burchiel KJ, Comella CL, Jankovic J, Lang AE, Laws ER Jr, Lozano AM, Penn RD, Simpson RK Jr, Stacy M, Wooten GF; ICV GDNF Study Group (2003) Implanted intracerebroventricular. Glial cell line-derived neurotrophic factor. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology 6:69–73

  84. Gill SS, Patel NK, Hotton GR, O’Sullivan K, McCarter R, Bunnage M, Brooks DJ, Svendsen CN, Heywood P (2003) Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat Med 9:589–595

    Article  CAS  PubMed  Google Scholar 

  85. Lang AE, Gill S, Patel NK, Lozano A, Nutt JG, Penn R, Brooks DJ, Hotton G, Moro E, Heywood P, Brodsky MA, Burchiel K, Kelly P, Dalvi A, Scott B, Stacy M, Turner D, Wooten VG, Elias WJ, Laws ER, Dhawan V, Stoessl AJ, Matcham J, Coffey RJ, Traub M (2006) Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann Neurol 59:459–466

    Article  CAS  PubMed  Google Scholar 

  86. Marks WJ Jr, Bartus RT, Siffert J, Davis CS, Lozano A, Boulis N, Vitek J, Stacy M, Turner D, Verhagen L, Bakay R, Watts R, Guthrie B, Jankovic J, Simpson R, Tagliati M, Alterman R, Stern M, Baltuch G, Starr PA, Larson PS, Ostrem JL, Nutt J, Kieburtz K, Kordower JH, Olanow CW (2010) Gene delivery of AAV2-neurturin for Parkinson’s disease: a double-blind, randomised, controlled trial. Lancet Neurol 9:1164–1172

    Article  CAS  PubMed  Google Scholar 

  87. Petrova P, Raibekas A, Pevsner J, Vigo N, Anafi M, Moore MK, Peaire AE, Shridhar V, Smith DI, Kelly J, Durocher Y, Commissiong JW (2003) MANF: a new mesencephalic, astrocyte-derived neurotrophic factor with selectivity for dopaminergic neurons. J Mol Neurosci 20:173–188

    Article  CAS  PubMed  Google Scholar 

  88. Lindholm P, Voutilainen MH, Laurén J, Peränen J, Leppänen V-M, Andressoo J-O, Lindahl M, Janhunen S, Kalkkinen N, Timmusk T, Tuominen RK, Saarma M (2007) Novel neurotrophic factor CDNF protects and rescues midbrain dopaminergic neurons in vivo. Nature 448:73–77

    Article  CAS  PubMed  Google Scholar 

  89. Lindholm P, Saarma M (2010) Novel CDNF/MANF family of neurotrophic factors. Dev Neurobiol 70:360–371

    CAS  PubMed  Google Scholar 

  90. Lindahl M, Danilova T, Palm E, Pulkkila P, Voikar V, Hakonen E, Ustinov J, Andressoo J-O, Harvery B, Otonkoski T, Rossi J, Saarma M (2014) MANF is indispensable for the proliferation and survival of pancreatic β-cells. Cell Reports 7:366–375

    Article  CAS  PubMed  Google Scholar 

  91. Latge C, Cabral KM, Johanson L, Romão LF, Herrmann T, Almeida MS, Foguel D (2015) The solution structure and dynamics of full-length human cerebral dopamine neurotrophic factor and its neuroprotective role against α-synuclein oligomers. J Biol Chem 290:20527–20540

    Article  CAS  PubMed  Google Scholar 

  92. Voutilainen MH, Bäck S, Pörsti E, Toppinen L, Lindgren L, Lindholm P, Peränen J, Saarma M, Tuominen RK (2009) Mesencephalic astrocyte-derived neurotrophic factor is neurorestorative in rat model of Parkinson’s disease. J Neurosci 29:9651–9659

    Article  CAS  PubMed  Google Scholar 

  93. Voutilainen MH, Bäck S, Peränen J, Lindholm P, Raasmaja A, Männistö PT, Saarma M, Tuominen RK (2011) Chronic infusion of CDNF prevents 6-OHDA-induced deficits in a rat model of Parkinson’s disease. Exp Neurol 228:99–108

    Article  CAS  PubMed  Google Scholar 

  94. Piltonen M, Planken A, Leskelä O, Myöhänen TT, Hänninen AL, Auvinen P, Alitalo K, Andressoo JO, Saarma M, Männistö PT (2011) Vascular endothelial growth factor C acts as a neurotrophic factor for dopamine neurons in vitro and in vivo. Neuroscience 192:550–563

    Article  CAS  PubMed  Google Scholar 

  95. Peterziel H, Unsicker K, Krieglstein K (2002) TGFbeta induces GDNF responsiveness in neurons by recruitment of GFRalpha1 to the plasma membrane. J Cell Biol 59:157–167

    Article  CAS  Google Scholar 

  96. Heldin CH, Westermark B (1999) Mechanism of action and in vivo role of platelet-derived growth factor. Physiol Rev 79:1283–1316

    CAS  PubMed  Google Scholar 

  97. Pietz K, Odin P, Funa K, Lindvall O (1996) Protective effect of platelet-derived growth factor against 6-hydroxydopamine-induced lesion of rat dopaminergic neurons in culture. Neurosci Lett 204:101–104

    Article  CAS  PubMed  Google Scholar 

  98. Funa K, Yamada N, Brodin G, Pietz K, Ahgren A, Wictorin K, Lindvall O, Odin P (1996) Enhanced synthesis of platelet-derived growth factor following injury induced by 6-hydroxydopamine in rat brain. Neuroscience 74:825–833

    Article  CAS  PubMed  Google Scholar 

  99. Zachrisson O, Zhao M, Andersson A, Dannaeus K, Häggblad J, Isacson R, Nielsen E, Patrone C, Rönnholm H, Wikstrom L, Delfani K, McCormack AL, Palmer T, Di Monte DA, Hill MP, Janson Lang AM, Haegerstrand A (2011) Restorative effects of platelet derived growth factor-BB in rodent models of Parkinson’s disease. J Parkinsons Dis 1:49–63

    PubMed  Google Scholar 

  100. Paul G, Zachrisson O, Varrone A, Almqvist P, Jerling M, Lind G, Rehncrona S, Linderoth B, Bjartmarz H, Shafer LL, Coffey R, Svensson M, Mercer KJ, Forsberg A, Halldin C, Svenningsson P, Widner H, Frisén J, Pålhagen S, Haegerstrand A (2015) Safety and tolerability of intracerebroventricular PDGF-BB in Parkinson’s disease patients. J Clin Invest. 125:1339–1346

    Article  PubMed  PubMed Central  Google Scholar 

  101. Foltynie T (2015) Can Parkinson’s disease be cured by stimulating neurogenesis? J Clin Invest 125:978–980

    Article  PubMed  PubMed Central  Google Scholar 

  102. Ohmachi S, Mikami T, Konishi M, Miyake A, Itoh N (2003) Preferential neurotrophic activity of fibroblast growth factor-20 for dopaminergic neurons through fibroblast growth factor receptor-1c. J Neurosci Res 72:436–443

    Article  CAS  PubMed  Google Scholar 

  103. Murase S, McKay RD (2006) A specific survival response in dopamine neurons at most risk in Parkinson’s disease. J Neurosci 26:9750–9760

    Article  CAS  PubMed  Google Scholar 

  104. Sleeman IJ, Boshoff EL, Duty S (2012) Fibroblast growth factor-20 protects against dopamine neuron loss in vitro and provides functional protection in the 6-hydroxydopamine-lesioned rat model of Parkinson’s disease. Neuropharmacology 63:1268–1277

    Article  CAS  PubMed  Google Scholar 

  105. Correia AS, Anisimov SV, Li JY, Brundin P (2008) Growth factors and feeder cells promote differentiation of human embryonic stem cells into dopaminergic neurons: a novel role for fibroblast growth factor-20. Front Neurosci 2:26–34

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. van der Walt JM, Noureddine MA, Kittappa R, Hauser MA, Scott WK, McKay R, Zhang F, Stajich JM, Fujiwara K, Scott BL, Pericak-Vance MA, Vance JM, Martin ER (2004) Fibroblast growth factor 20 polymorphisms and haplotypes strongly influence risk of Parkinson disease. Am J Hum Genet 74:1121–1127

    Article  PubMed  PubMed Central  Google Scholar 

  107. Xu X, Wang N, Xu H, Xie A, Jiang H, Xie J (2013) Fibroblast growth factor 20 polymorphism in sporadic Parkinson’s disease in Northern Han Chinese. J Clin Neurosci 20:1588–1590

    Article  CAS  PubMed  Google Scholar 

  108. Grothe C, Timmer M (2007) The physiological and pharmacological role of basic fibroblast growth factor in the dopaminergic nigrostriatal system. Brain Res Rev 54:80–91

    Article  CAS  PubMed  Google Scholar 

  109. Cantó C, Auwerx J (2012) Cell biology. FGF21 takes a fat bite. Science 336:675–676

    Article  PubMed  PubMed Central  Google Scholar 

  110. Zhang F, Yu L, Lin X, Cheng P, He L, Li X, Lu X, Tan Y, Yang H, Cai L, Zhang C (2015) Minireview: roles of fibroblast growth factors 19 and 21 in metabolic regulation and chronic diseases. Mol Endocrinol 26:me20151155

  111. Kuro-o M (2008) Endocrine FGFs and Klothos: emerging concepts. Trends Endocrinol Metab 19:239–245

    Article  CAS  PubMed  Google Scholar 

  112. Suomalainen A, Elo JM, Pietiläinen KH, Hakonen AH, Sevastianova K, Korpela M, Isohanni P, Marjavaara SK, Tyni T, Kiuru-Enari S, Pihko H, Darin N, Õunap K, Kluijtmans LA, Paetau A, Buzkova J, Bindoff LA, Annunen-Rasila J, Uusimaa J, Rissanen A, Yki-Järvinen H, Hirano M, Tulinius M, Smeitink J, Tyynismaa H (2011) FGF-21 as a biomarker for muscle-manifesting mitochondrial respiratory chain deficiencies: a diagnostic study. Lancet Neurol 10:806–818

    Article  CAS  PubMed  Google Scholar 

  113. Hsuchou H, Pan W, Kastin AJ (2007) The fasting polypeptide FGF21 can enter brain from blood. Peptides 28:2382–2386

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Zhang Y, Xie Y, Berglund ED, Coate KC, He TT, Katafuchi T, Xiao G, Potthoff MJ, Wei W, Wan Y, Yu RT, Evans RM, Kliewer SA, Mangelsdorf DJ (2012) The starvation hormone, fibroblast growth factor-21, extends lifespan in mice. Elife 1:e00065

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Mäkelä J, Tselykh TV, Maiorana F, Eriksson O, Do HT, Mudò G, Korhonen LT, Belluardo N, Lindholm D (2014) Fibroblast growth factor-21 enhances mitochondrial functions and increases the activity of PGC-1α in human dopaminergic neurons via Sirtuin-1. Springerplus 3:2. doi:10.1186/2193-1801-3-2

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Miyata A, Arimura A, Dahl RR, Minamino N, Uehara A, Jiang L, Culler MD, Coy DH (1989) Isolation of a novel 38 residue-hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells. Biochem Biophys Res Commun 164:567–574

    Article  CAS  PubMed  Google Scholar 

  117. Arimura A, Somogyvari-Vigh A, Weill C, Fiore RC, Tatsuno I, Bay V, Brenneman DE (1994) PACAP functions as a neurotrophic factor. Ann N Y Acad Sci 739:228–243

    Article  CAS  PubMed  Google Scholar 

  118. Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BK, Hashimoto H, Galas L, Vaudry H (2009) Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 61:283–357

    Article  CAS  PubMed  Google Scholar 

  119. Reglodi D, Kiss P, Lubics A, Tamas A (2011) Review on the protective effects of PACAP in models of neurodegenerative diseases in vitro and in vivo. Curr Pharm Des 17:962–972

    Article  CAS  PubMed  Google Scholar 

  120. Spengler D, Waeber C, Pantaloni C, Holsboer F, Bockaert J, Seeburg PH, Journot L (1993) Differential signal transduction by five splice variants of the PACAP receptor. Nature 365:170–175

    Article  CAS  PubMed  Google Scholar 

  121. Wang G, PanJ TanYY, SunXK ZhangYF, ZhouHY RenRJ, WangXJ Chen SD (2008) Neuroprotective effects of PACAP27 in mice model of Parkinson’s disease involved in the modulation of K(ATP) subunits and D2 receptors in the striatum. Neuropeptides 42:267–276

    Article  CAS  PubMed  Google Scholar 

  122. Lee EH, Seo SR (2014) Neuroprotective roles of pituitary adenylate cyclase-activating polypeptide in neurodegenerative diseases. BMB Rep 47:369–375

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Lee FS, Rajagopal R, Kim AH, Chang PC, Chao MV (2002) Activation of Trk neurotrophin receptor signaling by pituitary adenylate cyclase-activating polypeptides. J Biol Chem 277:9096–9102

    Article  CAS  PubMed  Google Scholar 

  124. Takei N, Skoglosa Y, Lindholm D (1998) Neurotrophic and neuroprotective effects of pituitary adenylate cyclase-activating polypeptide (PACAP) on mesencephalic dopaminergic neurons. J Neurosci Res 54:698–706

    Article  CAS  PubMed  Google Scholar 

  125. Chung CY, Seo H, Sonntag KC, Brooks A, Lin L, Isacson O (2005) Cell type-specific gene expression of midbrain dopaminergic neurons reveals molecules involved in their vulnerability and protection. Hum Mol Genet 14:1709–1725

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Reglodi D, Kiss P, Szabadfi K, Atlasz T, Gabriel R, Horvath G, SzakalyP Sandor B, Lubics A, Laszlo E, Farkas J, Matkovits A, Brubel R, Hashimoto H, Ferencz A, Vincze A, Helyes Z, Welke L, Lakatos A, Tamas A (2012) PACAP is an endogenous protective factor-insights from PACAP-deficient mice. J Mol Neurosci 48:482–492

    Article  CAS  PubMed  Google Scholar 

  127. Watson MB, Nobuta H, Abad C, Lee SK, Bala N, Zhu C, Richter F, Chesselet MF, Waschek JA (2013) PACAP deficiency sensitizes nigrostriatal dopaminergic neurons to paraquat-induced damage and modulates central and peripheral inflammatory activation in mice. Neuroscience 240:277–286

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Delgado M, Leceta J, Ganea D (2003) Vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide inhibit the production of inflammatory mediators by activated microglia. J Leukoc Biol 73:155–164

    Article  CAS  PubMed  Google Scholar 

  129. Mäkelä J, Koivuniemi R, Korhonen L, Lindholm D (2010) Interferon-gamma produced by microglia and the neuropeptide PACAP have opposite effects on the viability of neural progenitor cells. PLoS One 5:e11091

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Banks WA, Kastin AJ, Komaki G, Arimura A (1993) Passage of pituitary adenylate cyclase activating polypeptide1-27 and pituitary adenylate cyclase activating polypeptide1-38 across the blood-brain barrier. J Pharmacol Exp Ther 267:690–696

    CAS  PubMed  Google Scholar 

  131. Dogrukol-Ak D, Tore F, Tuncel N (2004) Passage of VIP/PACAP/secretin family across the blood-brain barrier: therapeutic effects. Curr Pharm Des 10:1325–1340

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The work in the groups is supported by Academy of Finland (DL, MS), Jane and Aatos Erkko Foundation (MS), Michael J Fox Foundation for Parkinson’s Research (MS), Finnish Parkinson Foundation (DL, JM), Minerva Foundation (DL, JM), University of Helsinki (DL, JM, OE, MS) and by Progetti di Ateneo, University of Palermo (VD, GM, NB).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dan Lindholm.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lindholm, D., Mäkelä, J., Di Liberto, V. et al. Current disease modifying approaches to treat Parkinson’s disease. Cell. Mol. Life Sci. 73, 1365–1379 (2016). https://doi.org/10.1007/s00018-015-2101-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-015-2101-1

Keywords

Navigation