Skip to main content
Log in

PARK14 (D331Y) PLA2G6 Causes Early-Onset Degeneration of Substantia Nigra Dopaminergic Neurons by Inducing Mitochondrial Dysfunction, ER Stress, Mitophagy Impairment and Transcriptional Dysregulation in a Knockin Mouse Model

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

PARK14 patients with homozygous (D331Y) PLA2G6 mutation display motor deficits of pure early-onset Parkinson’s disease (PD). The aim of this study is to investigate the pathogenic mechanism of mutant (D331Y) PLA2G6-induced PD. We generated knockin (KI) mouse model of PARK14 harboring homozygous (D331Y) PLA2G6 mutation. Then, we investigated neuropathological and neurological phenotypes of PLA2G6D331Y/D331Y KI mice and molecular pathogenic mechanisms of (D331Y) PLA2G6-induced degeneration of substantia nigra (SN) dopaminergic neurons. Six-or nine-month-old PLA2G6D331Y/D331Y KI mice displayed early-onset cell death of SNpc dopaminergic neurons. Lewy body pathology was found in the SN of PLA2G6D331Y/D331Y mice. Six-or nine-month-old PLA2G6D331Y/D331Y KI mice exhibited early-onset parkinsonism phenotypes. Disrupted cristae of mitochondria were found in SNpc dopaminergic neurons of PLA2G6D331Y/D331Y mice. PLA2G6D331Y/D331Y mice displayed mitochondrial dysfunction and upregulated ROS production, which may lead to activation of apoptotic cascade. Upregulated protein levels of Grp78, IRE1, PERK, and CHOP, which are involved in activation of ER stress, were found in the SN of PLA2G6D331Y/D331Y mice. Protein expression of mitophagic proteins, including parkin and BNIP3, was downregulated in the SN of PLA2G6D331Y/D331Y mice, suggesting that (D331Y) PLA2G6 mutation causes mitophagy dysfunction. In the SN of PLA2G6D331Y/D331Y mice, mRNA levels of eight genes that are involved in neuroprotection/neurogenesis were decreased, while mRNA levels of two genes that promote apoptotic death were increased. Our results suggest that PARK14 (D331Y) PLA2G6 mutation causes degeneration of SNpc dopaminergic neurons by causing mitochondrial dysfunction, elevated ER stress, mitophagy impairment, and transcriptional abnormality.

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
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Dickson DW, Braak H, Duda JE, Duyckaerts C, Gasser T, Halliday GM, Hardy J, Leverenz JB et al (2009) Neuropathological assessment of Parkinson’s disease: Refining the diagnostic criteria. Lancet Neurol 8(12):1150–1157. https://doi.org/10.1016/S1474-4422(09)70238-8S1474-4422(09)70238-8

    Article  CAS  PubMed  Google Scholar 

  2. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M (1998) alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with Lewy bodies. Proc Natl Acad Sci U S A 95(11):6469–6473

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Schapira AH, Jenner P (2011) Etiology and pathogenesis of Parkinson’s disease. Mov Disord 26(6):1049–1055. https://doi.org/10.1002/mds.23732

    Article  PubMed  Google Scholar 

  4. Menzies FM, Fleming A, Caricasole A, Bento CF, Andrews SP, Ashkenazi A, Fullgrabe J, Jackson A et al (2017) Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities. Neuron 93(5):1015–1034. https://doi.org/10.1016/j.neuron.2017.01.022

    Article  CAS  PubMed  Google Scholar 

  5. Houlden H, Singleton AB (2012) The genetics and neuropathology of Parkinson’s disease. Acta Neuropathol 124(3):325–338. https://doi.org/10.1007/s00401-012-1013-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hayflick SJ, Kurian MA, Hogarth P (2018) Neurodegeneration with brain iron accumulation. Handb Clin Neurol 147:293–305. https://doi.org/10.1016/B978-0-444-63233-3.00019-1

    Article  PubMed  PubMed Central  Google Scholar 

  7. Lu CS, SC Lai RMW, Weng YH, Huang CL, Chen RS, Chang HC, Wu-Chou YH, Yeh TH (2012) PLA2G6 mutations in PARK14-linked young-onset parkinsonism and sporadic Parkinson’s disease. Am J Med Genet B Neuropsychiatr Genet 159B(2):183–191. https://doi.org/10.1002/ajmg.b.32012

    Article  CAS  PubMed  Google Scholar 

  8. Shi CH, Tang BS, Wang L, Lv ZY, Wang J, Luo LZ, Shen L, Jiang H et al (2011) PLA2G6 gene mutation in autosomal recessive early-onset parkinsonism in a Chinese cohort. Neurology 77(1):75–81

    Article  PubMed  Google Scholar 

  9. Xie F, Cen Z, Ouyang Z, Wu S, Xiao J, Luo W (2015) Homozygous p.D331Y mutation in PLA2G6 in two patients with pure autosomal-recessive early-onset parkinsonism: further evidence of a fourth phenotype of PLA2G6-associated neurodegeneration. Parkinsonism Relat Disord 21(4):420–422. https://doi.org/10.1016/j.parkreldis.2015.01.012S1353-8020(15)00032-2

    Article  CAS  PubMed  Google Scholar 

  10. Ramanadham S, Ali T, Ashley JW, Bone RN, Hancock WD, Lei X (2015) Calcium-independent phospholipases A2 and their roles in biological processes and diseases. J Lipid Res 56(9):1643–1668. https://doi.org/10.1194/jlr.R058701jlr.R058701

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ong WY, Farooqui T, Farooqui AA (2010) Involvement of cytosolic phospholipase A(2), calcium independent phospholipase A(2) and plasmalogen selective phospholipase A(2) in neurodegenerative and neuropsychiatric conditions. Curr Med Chem 17(25):2746–2763

    Article  CAS  PubMed  Google Scholar 

  12. Xie Z, MC Gong WS, Turk J, Guo Z (2007) Group VIA phospholipase A2 (iPLA2beta) participates in angiotensin II-induced transcriptional up-regulation of regulator of g-protein signaling-2 in vascular smooth muscle cells. J Biol Chem 282(35):25278–25289. https://doi.org/10.1074/jbc.M611206200

    Article  CAS  PubMed  Google Scholar 

  13. Kinghorn KJ, Castillo-Quan JI, Bartolome F, Angelova PR, Li L, Pope S, Cocheme HM, Khan S et al (2015) Loss of PLA2G6 leads to elevated mitochondrial lipid peroxidation and mitochondrial dysfunction. Brain 138(Pt 7):1801–1816. https://doi.org/10.1093/brain/awv132awv132

    Article  PubMed  PubMed Central  Google Scholar 

  14. Beck G, Sugiura Y, Shinzawa K, Kato S, Setou M, Tsujimoto Y, Sakoda S, Sumi-Akamaru H (2011) Neuroaxonal dystrophy in calcium-independent phospholipase A2beta deficiency results from insufficient remodeling and degeneration of mitochondrial and presynaptic membranes. J Neurosci 31(31):11411–11420. https://doi.org/10.1523/JNEUROSCI.0345-11.201131/31/11411

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zhou Q, Yen A, Rymarczyk G, Asai H, Trengrove C, Aziz N, Kirber MT, Mostoslavsky G et al (2016) Impairment of PARK14-dependent Ca(2+) signalling is a novel determinant of Parkinson’s disease. Nat Commun 7:10332. https://doi.org/10.1038/ncomms10332ncomms10332

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gao L, Hidalgo-Figueroa M, Escudero LM, Diaz-Martin J, Lopez-Barneo J, Pascual A (2013) Age-mediated transcriptomic changes in adult mouse substantia nigra. PLoS One 8(4):e62456. https://doi.org/10.1371/journal.pone.0062456

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Masuda-Suzukake M, Nonaka T, Hosokawa M, Oikawa T, Arai T, Akiyama H, Mann DM, Hasegawa M (2013) Prion-like spreading of pathological alpha-synuclein in brain. Brain 136(Pt 4):1128–1138. https://doi.org/10.1093/brain/awt037

    Article  PubMed  PubMed Central  Google Scholar 

  18. Henderson MX, Chung CH, Riddle DM, Zhang B, Gathagan RJ, Seeholzer SH, Trojanowski JQ, Lee VMY (2017) Unbiased proteomics of early Lewy body formation model implicates active microtubule affinity-regulating kinases (MARKs) in synucleinopathies. J Neurosci 37(24):5870–5884. https://doi.org/10.1523/JNEUROSCI.2705-16.2017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Paisan-Ruiz C, Li A, Schneider SA, Holton JL, Johnson R, Kidd D, Chataway J, Bhatia KP et al (2012) Widespread Lewy body and tau accumulation in childhood and adult onset dystonia-parkinsonism cases with PLA2G6 mutations. Neurobiol Aging 33(4):814–823. https://doi.org/10.1016/j.neurobiolaging.2010.05.009S0197-4580(10)00223-X

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Paisan-Ruiz C, Bhatia KP, Li A, Hernandez D, Davis M, Wood NW, Hardy J, Houlden H et al (2009) Characterization of PLA2G6 as a locus for dystonia-parkinsonism. Ann Neurol 65(1):19–23. https://doi.org/10.1002/ana.21415

    Article  PubMed  PubMed Central  Google Scholar 

  21. Schapira AH, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD (1990) Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem 54(3):823–827

    Article  CAS  PubMed  Google Scholar 

  22. Kagan VE, Tyurin VA, Jiang J, Tyurina YY, Ritov VB, Amoscato AA, Osipov AN, Belikova NA et al (2005) Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors. Nat Chem Biol 1(4):223–232. https://doi.org/10.1038/nchembio727

    Article  CAS  PubMed  Google Scholar 

  23. Remondelli P, Renna M (2017) The endoplasmic reticulum unfolded protein response in neurodegenerative disorders and its potential therapeutic significance. Front Mol Neurosci 10:187. https://doi.org/10.3389/fnmol.2017.00187

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Celardo I, Costa AC, Lehmann S, Jones C, Wood N, Mencacci NE, Mallucci GR, Loh SH et al (2016) Mitofusin-mediated ER stress triggers neurodegeneration in pink1/parkin models of Parkinson’s disease. Cell Death Dis 7(6):e2271. https://doi.org/10.1038/cddis.2016.173cddis2016173

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lei X, Zhang S, Bohrer A, Bao S, Song H, Ramanadham S (2007) The group VIA calcium-independent phospholipase A2 participates in ER stress-induced INS-1 insulinoma cell apoptosis by promoting ceramide generation via hydrolysis of sphingomyelins by neutral sphingomyelinase. Biochemistry 46(35):10170–10185. https://doi.org/10.1021/bi700017z

    Article  CAS  PubMed  Google Scholar 

  26. Rodolfo C, Campello S, Cecconi F (2017) Mitophagy in neurodegenerative diseases. Neurochem Int. https://doi.org/10.1016/j.neuint.2017.08.004

    Article  CAS  PubMed  Google Scholar 

  27. Thomas RL, Kubli DA, Gustafsson AB (2011) Bnip3-mediated defects in oxidative phosphorylation promote mitophagy. Autophagy 7(7):775–777

    Article  PubMed  Google Scholar 

  28. Yu W, Polepalli J, Wagh D, Rajadas J, Malenka R, Lu B (2012) A critical role for the PAR-1/MARK-tau axis in mediating the toxic effects of Abeta on synapses and dendritic spines. Hum Mol Genet 21(6):1384–1390. https://doi.org/10.1093/hmg/ddr576ddr576

    Article  CAS  PubMed  Google Scholar 

  29. Liston P, Fong WG, Kelly NL, Toji S, Miyazaki T, Conte D, Tamai K, Craig CG et al (2001) Identification of XAF1 as an antagonist of XIAP anti-caspase activity. Nat Cell Biol 3(2):128–133. https://doi.org/10.1038/35055027

    Article  CAS  PubMed  Google Scholar 

  30. Kowalczyk A, RK Filipkowski MR, Wilczynski GM, FA Konopacki JJ, Ciemerych MA, Sicinski P, Kaczmarek L (2004) The critical role of cyclin D2 in adult neurogenesis. J Cell Biol 167(2):209–213. https://doi.org/10.1083/jcb.200404181

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. IM Ethell FI, Kalo MS, Couchman JR, Pasquale EB, Yamaguchi Y (2001) EphB/syndecan-2 signaling in dendritic spine morphogenesis. Neuron 31(6):1001–1013

    Article  Google Scholar 

  32. Cavanaugh JE, Jaumotte JD, Lakoski JM, Zigmond MJ (2006) Neuroprotective role of ERK1/2 and ERK5 in a dopaminergic cell line under basal conditions and in response to oxidative stress. J Neurosci Res 84(6):1367–1375. https://doi.org/10.1002/jnr.21024

    Article  CAS  PubMed  Google Scholar 

  33. Du J, Zhu Y, Chen X, Fei Z, Yang S, Yuan W, Zhang J, Zhu T (2007) Protective effect of bone morphogenetic protein-6 on neurons from H2O2 injury. Brain Res 1163:10–20. https://doi.org/10.1016/j.brainres.2007.06.002

    Article  CAS  PubMed  Google Scholar 

  34. Liu Y, Hao S, Yang B, Fan Y, Qin X, Chen Y, Hu J (2017) Wnt/beta-catenin signaling plays an essential role in alpha7 nicotinic receptor-mediated neuroprotection of dopaminergic neurons in a mouse Parkinson's disease model. Biochem Pharmacol 140:115–123. https://doi.org/10.1016/j.bcp.2017.05.017

    Article  CAS  PubMed  Google Scholar 

  35. Duffy AM, Schaner MJ, Wu SH, Staniszewski A, Kumar A, Arevalo JC, Arancio O, Chao MV et al (2011) A selective role for ARMS/Kidins220 scaffold protein in spatial memory and trophic support of entorhinal and frontal cortical neurons. Exp Neurol 229(2):409–420. https://doi.org/10.1016/j.expneurol.2011.03.008S0014-4886(11)00091-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Meyer RC, Giddens MM, Coleman BM, Hall RA (2014) The protective role of prosaposin and its receptors in the nervous system. Brain Res 1585:1–12. https://doi.org/10.1016/j.brainres.2014.08.022S0006-8993(14)01086-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Park HK, Cho AR, Lee SC, Ban JY (2012) MPTP-induced model of Parkinson’s disease in heat shock protein 70.1 knockout mice. Mol Med Rep 5(6):1465–1468. https://doi.org/10.3892/mmr.2012.839

    Article  CAS  PubMed  Google Scholar 

  38. Yoshino H, Tomiyama H, Tachibana N, Ogaki K, Li Y, Funayama M, Hashimoto T, Takashima S et al (2010) Phenotypic spectrum of patients with PLA2G6 mutation and PARK14-linked parkinsonism. Neurology 75(15):1356–1361. https://doi.org/10.1212/WNL.0b013e3181f7364975/15/1356

    Article  CAS  PubMed  Google Scholar 

  39. Bonifati V (2014) Genetics of Parkinson’s disease—state of the art, 2013. Parkinsonism Relat Disord 20(Suppl 1):S23–S28. https://doi.org/10.1016/S1353-8020(13)70009-9S1353-8020(13)70009-9

    Article  PubMed  Google Scholar 

  40. Miki Y, Yoshizawa T, Morohashi S, Seino Y, Kijima H, Shoji M, Mori A, Yamashita C et al (2017) Neuropathology of PARK14 is identical to idiopathic Parkinson’s disease. Mov Disord 32(5):799–800. https://doi.org/10.1002/mds.26952

    Article  CAS  PubMed  Google Scholar 

  41. Beck G, Shinzawa K, Hayakawa H, Baba K, Sumi-Akamaru H, Tsujimoto Y, Mochizuki H (2016) Progressive axonal degeneration of nigrostriatal dopaminergic neurons in calcium-independent phospholipase A2beta knockout mice. PLoS One 11(4):e0153789. https://doi.org/10.1371/journal.pone.0153789

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sumi-Akamaru H, Beck G, Kato S, Mochizuki H (2015) Neuroaxonal dystrophy in PLA2G6 knockout mice. Neuropathology 35(3):289–302. https://doi.org/10.1111/neup.12202

    Article  CAS  PubMed  Google Scholar 

  43. Blanchard H, Taha AY, Cheon Y, Kim HW, Turk J, Rapoport SI (2014) iPLA2beta knockout mouse, a genetic model for progressive human motor disorders, develops age-related neuropathology. Neurochem Res 39(8):1522–1532. https://doi.org/10.1007/s11064-014-1342-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sumi-Akamaru H, Beck G, Shinzawa K, Kato S, Riku Y, Yoshida M, Fujimura H, Tsujimoto Y et al (2016) High expression of alpha-synuclein in damaged mitochondria with PLA2G6 dysfunction. Acta Neuropathol Commun 4:27. https://doi.org/10.1186/s40478-016-0298-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Michel PP, Hirsch EC, Hunot S (2016) Understanding dopaminergic cell death pathways in Parkinson disease. Neuron 90(4):675–691. https://doi.org/10.1016/j.neuron.2016.03.038S0896-6273(16)30058-7

    Article  CAS  PubMed  Google Scholar 

  46. Esteves AR, Arduino DM, Silva DF, Oliveira CR, Cardoso SM (2011) Mitochondrial dysfunction: the road to alpha-synuclein oligomerization in PD. Parkinsons Dis 2011(693761):1–20. https://doi.org/10.4061/2011/693761

    Article  CAS  Google Scholar 

  47. Takahashi M, Ko LW, Kulathingal J, Jiang P, Sevlever D, Yen SH (2007) Oxidative stress-induced phosphorylation, degradation and aggregation of alpha-synuclein are linked to upregulated CK2 and cathepsin D. Eur J Neurosci 26(4):863–874. https://doi.org/10.1111/j.1460-9568.2007.05736.x

    Article  PubMed  Google Scholar 

  48. Mercado G, Castillo V, Vidal R, Hetz C (2015) ER proteostasis disturbances in Parkinson’s disease: novel insights. Front Aging Neurosci 7:39. https://doi.org/10.3389/fnagi.2015.00039

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Urra H, Dufey E, Lisbona F, Rojas-Rivera D, Hetz C (2013) When ER stress reaches a dead end. Biochim Biophys Acta 1833(12):3507–3517. https://doi.org/10.1016/j.bbamcr.2013.07.024S0167-4889(13)00311-X

    Article  CAS  PubMed  Google Scholar 

  50. 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 alpha-synucleinopathy in vivo. J Neurosci 32(10):3306–3320. https://doi.org/10.1523/JNEUROSCI.5367-11.201232/10/3306

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Martinez-Vicente M (2017) Neuronal mitophagy in neurodegenerative diseases. Front Mol Neurosci 10:64. https://doi.org/10.3389/fnmol.2017.00064

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Crews L, Adame A, Patrick C, Delaney A, Pham E, Rockenstein E, Hansen L, Masliah E (2010) Increased BMP6 levels in the brains of Alzheimer’s disease patients and APP transgenic mice are accompanied by impaired neurogenesis. J Neurosci 30(37):12252–12262. https://doi.org/10.1523/JNEUROSCI.1305-10.201030/37/12252

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Jordan J, Bottner M, Schluesener HJ, Unsicker K, Krieglstein K (1997) Bone morphogenetic proteins: neurotrophic roles for midbrain dopaminergic neurons and implications of astroglial cells. Eur J Neurosci 9(8):1699–1709

    Article  CAS  PubMed  Google Scholar 

  54. Garthe A, Huang Z, Kaczmarek L, Filipkowski RK, Kempermann G (2014) Not all water mazes are created equal: cyclin D2 knockout mice with constitutively suppressed adult hippocampal neurogenesis do show specific spatial learning deficits. Genes Brain Behav 13(4):357–364. https://doi.org/10.1111/gbb.12130

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Luo SX, Huang EJ (2016) Dopaminergic neurons and brain reward pathways: from neurogenesis to circuit assembly. Am J Pathol 186(3):478–488. https://doi.org/10.1016/j.ajpath.2015.09.023S0002-9440(15)00646-X

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Joksimovic M, Awatramani R (2014) Wnt/beta-catenin signaling in midbrain dopaminergic neuron specification and neurogenesis. J Mol Cell Biol 6(1):27–33. https://doi.org/10.1093/jmcb/mjt043mjt043

    Article  CAS  PubMed  Google Scholar 

  57. Neubrand VE, Cesca F, Benfenati F, Schiavo G (2012) Kidins220/ARMS as a functional mediator of multiple receptor signalling pathways. J Cell Sci 125(Pt 8):1845–1854. https://doi.org/10.1242/jcs.102764jcs.102764

    Article  CAS  PubMed  Google Scholar 

  58. Scholz-Starke J, Cesca F, Schiavo G, Benfenati F, Baldelli P (2012) Kidins220/ARMS is a novel modulator of short-term synaptic plasticity in hippocampal GABAergic neurons. PLoS One 7(4):e35785. https://doi.org/10.1371/journal.pone.0035785

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Li N, Sarojini H, An J, Wang E (2010) Prosaposin in the secretome of marrow stroma-derived neural progenitor cells protects neural cells from apoptotic death. J Neurochem 112(6):1527–1538. https://doi.org/10.1111/j.1471-4159.2009.06565.xJNC6565

    Article  CAS  PubMed  Google Scholar 

  60. Gao HL, Li C, Nabeka H, Shimokawa T, Saito S, Wang ZY, Cao YM, Matsuda S (2013) Attenuation of MPTP/MPP(+) toxicity in vivo and in vitro by an 18-mer peptide derived from prosaposin. Neuroscience 236:373–393. https://doi.org/10.1016/j.neuroscience.2013.01.007S0306-4522(13)00023-7

    Article  CAS  PubMed  Google Scholar 

  61. Naz F, Anjum F, Islam A, Ahmad F, Hassan MI (2013) Microtubule affinity-regulating kinase 4: structure, function, and regulation. Cell Biochem Biophys 67(2):485–499. https://doi.org/10.1007/s12013-013-9550-7

    Article  CAS  PubMed  Google Scholar 

  62. West T, Stump M, Lodygensky G, Neil JJ, Deshmukh M, Holtzman DM (2009) Lack of X-linked inhibitor of apoptosis protein leads to increased apoptosis and tissue loss following neonatal brain injury. ASN Neuro 1(1):AN20090005. https://doi.org/10.1042/AN20090005e00004

    Article  Google Scholar 

Download references

Acknowledgements

The authors are grateful to Microscopy Core Laboratory and Center for Advanced Molecular Imaging and Translation, Department of Nuclear Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan. We thank Han Chiu and Chia-Chen Hsu for assistance and technical support. We would like to thank Chih-Wei Hu and Wen-Ai Wu of Transgenic Mouse Core Laboratory of Experimental Animal Center for help with ES cells microinjection.

Funding

This work was supported by the Ministry of Science and Technology, Taiwan (MOST 104-2314-B-182A-35-, MOST 105-2314-B-038-092-MY3 and MOST 105-2314-B-182A-013-MY3 to TH Yeh; MOST 105-2314-B-182A-003- and MOST 106-2314-B-182A-012-MY3 to CC Chiu; MOST104-2320-B-182-014-MY3 to HL Wang), Taipei Medical University (TMU106-AE1-B20 to TH Yeh), and the Chang Gung Medical Foundation (grants CMRPG3C1482, CMRPG3C0783, CMRPG3C1491, CMRPG3C1492, CMRPG3D0382, CRRPG3C0023, CRRPG3C0033 to TH Yeh.; CMRPG3F1821 to CC Chiu; CMRPD1B0332, CMRPD1C0623, CRRPD1C0013, CMRPD180433, and EMRPD1F0251 to HL Wang).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Tu-Hsueh Yeh or Hung-Li Wang.

Ethics declarations

Animal experiments were performed in accordance with protocols approved by Institutional Animal Care and Use Committee (IACUC) of Chang Gung University.

Conflict of Interest

The authors declare that they have no competing financial interests.

Electronic Supplementary Material

ESM 1

(DOCX 3469 kb)

ESM 2

(CEL 9838 kb)

ESM 3

(CEL 9828 kb)

ESM 4

(CEL 9829 kb)

ESM 5

(CEL 9829 kb)

ESM 6

(CEL 9831 kb)

ESM 7

(CEL 9831 kb)

ESM 8

(CEL 9830 kb)

ESM 9

(CEL 9826 kb)

ESM 10

(XLSX 185 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chiu, CC., Lu, CS., Weng, YH. et al. PARK14 (D331Y) PLA2G6 Causes Early-Onset Degeneration of Substantia Nigra Dopaminergic Neurons by Inducing Mitochondrial Dysfunction, ER Stress, Mitophagy Impairment and Transcriptional Dysregulation in a Knockin Mouse Model. Mol Neurobiol 56, 3835–3853 (2019). https://doi.org/10.1007/s12035-018-1118-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1118-5

Keywords

Navigation