Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2120 2022-06-07 17:18:13 |
2 format correct -3 word(s) 2117 2022-06-08 04:00:15 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kawahata, I.; , .; Fukunaga, K. α-Synuclein Phosphorylation and Its Kinases. Encyclopedia. Available online: https://encyclopedia.pub/entry/23793 (accessed on 24 April 2024).
Kawahata I,  , Fukunaga K. α-Synuclein Phosphorylation and Its Kinases. Encyclopedia. Available at: https://encyclopedia.pub/entry/23793. Accessed April 24, 2024.
Kawahata, Ichiro, , Kohji Fukunaga. "α-Synuclein Phosphorylation and Its Kinases" Encyclopedia, https://encyclopedia.pub/entry/23793 (accessed April 24, 2024).
Kawahata, I., , ., & Fukunaga, K. (2022, June 07). α-Synuclein Phosphorylation and Its Kinases. In Encyclopedia. https://encyclopedia.pub/entry/23793
Kawahata, Ichiro, et al. "α-Synuclein Phosphorylation and Its Kinases." Encyclopedia. Web. 07 June, 2022.
α-Synuclein Phosphorylation and Its Kinases
Edit

α-Synuclein is a protein with a molecular weight of 14.5 kDa and consists of 140 amino acids encoded by the SNCA gene. Missense mutations and gene duplications in the SNCA gene cause hereditary Parkinson’s disease. Highly phosphorylated and abnormally aggregated α-synuclein is a major component of Lewy bodies found in neuronal cells of patients with sporadic Parkinson’s disease, dementia with Lewy bodies, and glial cytoplasmic inclusion bodies in oligodendrocytes with multiple system atrophy. Aggregated α-synuclein is cytotoxic and plays a central role in the pathogenesis of the above-mentioned synucleinopathies. In a healthy brain, most α-synuclein is unphosphorylated; however, more than 90% of abnormally aggregated α-synuclein in Lewy bodies of patients with Parkinson’s disease is phosphorylated at Ser129, which is presumed to be of pathological significance. Several kinases catalyze Ser129 phosphorylation, but the role of phosphorylation enzymes in disease pathogenesis and their relationship to cellular toxicity from phosphorylation are not fully understood in α-synucleinopathy. G-protein-coupled receptor kinases, casein kinase II, and polo-like kinase possess the ability to phosphorylate α-synuclein protein. On this point, inhibition of these kinases is able to prevent α-synuclein phosphorylation, which indicates the potential therapeutic targets and availability of drug development for α-synucleinopathies. α-Synuclein phosphorylation can clinically be an accompanying event in the brains of patients with Parkinson’s disease rather than the critical factor for α-synuclein aggregation and toxicity. Nevertheless, increasing phosphorylated α-synuclein and the accumulation with disease progression is useful as a therapeutic target and biomarker.

α-synuclein phosphorylation Parkinson’s disease dementia with Lewy bodies multiple system atrophy α-synucleinopathy

1. Characteristics and Distribution of α-Synuclein

α-Synuclein is a protein with a molecular weight of 14.5 kDa and consists of 140 amino acids encoded by the SNCA gene. The protein consists of three domains: the N-terminal amphipathic region, the non-amyloid component region, and the C-terminal acidic region (Figure 1). In the central nervous system, it is particularly abundant in presynaptic terminals and is presumed to be involved in regulating synaptic function and neural plasticity. Synuclein was first identified in cholinergic synaptic vesicles derived from the electric organ of the northeastern Pacific electric ray (Torpedo californica) [1]. The name “synuclein” originated from its distribution because its expression was observed not only in synaptic vesicles in the presynaptic terminal but also in the nucleus following immunostaining. Owing to this, α-synuclein was presumed to be a molecule related to the nervous system, especially in synaptic function.
Figure 1. Structure and summary of post-translational modification sites of α-synuclein. The schematic represents α-synuclein protein [2] with the N-terminal amphipathic region, non-amyloid component region, and C-terminal acidic region. Phosphorylation sites are indicated in red letters, and ubiquitination is shown in purple. The indicated Ser129 phosphorylation site is the potential region mostly associated with α-synuclein-induced neurodegeneration.
α-Synuclein is particularly highly expressed in the central nervous system in human adults—accounting for approximately 1% of soluble proteins in the brain—and is abundant in presynaptic terminals [3]. α-Synuclein protein is also abundant in erythrocytes [4]. Most α-synuclein is distributed in the cytoplasm of cells and its carboxyterminal region, which is flexible in monomeric and fibrillar states and plays an essential role in the interaction with other partners [5]. Some of the protein is associated with biological membranes, such as synaptic vesicles [6]. In addition, α-synuclein is observed in the mitochondrial inner membrane, the mitochondria-associated endoplasmic reticulum membranes, the Golgi apparatus, and endosomes [7][8][9][10]. It also interacts with various proteins, including tau, ATPases, lipid membranes, lymphocyte-activation gene 3, flotillin, and fatty acid-binding protein 3 [11][12][13][14][15][16][17][18][19][20][21], as well as metal ions [22][23]. Nuclear α-synuclein presumably functions as a DNA-binding protein and is thought to be involved in regulating various gene expressions [24][25][26].

2. Kinases That Phosphorylate α-Synuclein and Their Association to Cytotoxicity

Exogenous α-synuclein that is propagated and taken up into the cell facilitates aggregation with intracellular proteins to form inclusions, which are immunopositive for α-synuclein phosphorylated at Ser129 (Figure 2). Nonetheless, the question of how α-synuclein obtains these aggregation characteristics and enhances cytotoxicity arises. Earlier investigations reported that α-synuclein within Lewy bodies is subject to post-translational modifications, including phosphorylation and ubiquitination. Notably, among these modifications, the phosphorylation at residue Ser129 and its possible implication on α-synucleinopathy have had significant research focus and are postulated to play a key role in the regulation of α-synuclein aggregation and neurotoxicity [27][28][29]. In particular, a small portion of α-synuclein (~4%) is constitutively phosphorylated at Ser129 in the brains of healthy individuals [30][31][32], whereas >90% of the insoluble α-synuclein is phosphorylated in the brain of patients with synucleinopathies [31][33][34][35]. The accumulation of phosphorylated α-synuclein at Ser129 was also observed in animal models of Parkinson’s disease [36][37][38][39][40]. These observations indicate the importance of α-synuclein phosphorylation at Ser129 in regulating its aggregation and neurotoxicity.
Figure 2. α-Synuclein Ser129 phosphorylation in cultured dopaminergic neurons. Exposure to exogenous α-synuclein fibrils induces the formation of phosphorylated α-synuclein-positive intracellular aggregates. Cells were exposed to ATTO550-labeled α-synuclein fibrils for 48 h. Scale bar = 10 μm, and 1 μm in magnified images of ROI i, ii, and iii. Images were originally obtained for this entry.
The carboxy-terminal region of α-synuclein is flexible in monomeric and fibrillar forms as well as in membrane-bound states [5][19][41][42][43]. Interestingly, the α-synuclein C-terminal tail contains the majority of phosphorylation sites (Figure 3). Phosphorylation of α-synuclein Ser129 is mediated by several kinases such as G-protein-coupled receptor kinases (GRKs) [44][45][46], casein kinase II [46][47][48], polo-like kinases [49][50][51], and leucine-rich repeat kinase 2 (LRRK2) [52][53]. Below, researchers summarize the α-synuclein phosphorylation kinases and describe the associated pathogenic neurotoxicity.
Figure 3. Summary of representative phosphorylation sites, kinases, and mutation sites of α-synuclein [44][45][46][47][48][49][50][51][52][53][54]. GRK: G-protein-coupled receptor kinase; CKII: casein kinase II; PLK: polo-like kinase; LRRK2: leucine-rich repeat kinase 2; NAC: non-amyloid component.

2.1. Impact of G-Protein-Coupled Receptor Kinases on α-Synuclein Phosphorylation

GRKs are a serine/threonine kinase family that regulate G-protein-coupled receptors. Mammalian GRKs have seven isoforms that can be divided into three subfamilies based on their structural organization and homology: GRK1 (rhodopsin kinase) and GRK7; GRK2 (βARK1) and GRK3 (βARK2); and GRK4, GRK5, and GRK6. Among these isoforms, GRK2 preferentially phosphorylates α-synuclein, while GRK5 prefers α-isoform as a substrate [44][45]. Interestingly, GRK-mediated authentic phosphorylation and mimicking of the phosphorylation state (S129E) reduce α-synuclein affinity to binding phospholipids [44][55]. Furthermore, phosphomimetic mutation (S129D) inhibits its association with membranes [56][57]. These data indicate an inhibitory effect of Ser129 phosphorylation on the α-synuclein binding to the cellular membrane. Following the loss of ability to bind to plasma membrane, phosphorylated α-synuclein preferentially localizes to mitochondria and suppresses cell respiration [58][59], which also induces endoplasmic reticulum stress and cell death [36][60][61]. These data indicate that α-synuclein phosphorylation is linked to mitochondrial dysfunction and the production of reactive oxygen species. GRK5 is immunohistochemically co-localized with phosphorylated α-synuclein in Lewy bodies [45], indicating its pathogenic significance in α-synucleinopathies.

2.2. Impact of Casein Kinase II on α-Synuclein Phosphorylation

Another kinase of α-synuclein phosphorylation, casein kinase II (CK2), is a serine/threonine-selective protein kinase. CK2 is a tetrameric enzyme assembled from two catalytic subunits (CK2α and CK2α′) and a regulatory subunit (CK2β dimer). Casein kinase II is necessary for cell survival and is a crucial suppressor of apoptosis [62]. Wu et al. reported that CK2α can be S-nitrosylated by nitric oxide (NO). Intriguingly, S-nitrosylation of CK2α enhanced the kinase activity of this catalytic subunit towards the phosphorylation of α-synuclein at Ser129 [46]. These data indicate that CK2α can enhance the phosphorylation of pSer129 α-synuclein. In addition, Sano et al. found that α-synuclein aggregates are predominantly phosphorylated at Y136 in brains with DLB, which is mediated by the activity of CK2 [48]. Furthermore, aggregate formation with Ser129 phosphorylation was significantly attenuated by CK2 inhibition, which increased Y136 phosphorylation in cultured neuroblastoma SH-SY5Y cells [48]. Moreover, CK2β regulatory subunits are detected in the halo region of Lewy bodies in the substantia nigra of patients with Parkinson’s disease [60]. These data indicate the involvement of CK2-related α-synuclein phosphorylation in the pathogenesis of Lewy-body-associated diseases.

2.3. Impact of Polo-like Kinase on α-Synuclein Phosphorylation

The third phosphorylation candidate, polo-like kinase, is also a regulatory serine/threonine kinase containing an N-terminal kinase catalytic domain and a C-terminal polo-box domain (PBD) that is involved in substrate binding and regulation of kinase activity. Five mammalian PLK family members have been identified, viz. PLK1–5 [61]. The polo-like kinases PLK2 and PLK3 have phosphorylation activity against α-synuclein that is better than that of PLK1 and PLK4 and are capable of completely phosphorylating α-synuclein [49][50]. PLK5 lacks a functional kinase domain due to a premature stop codon in exon 6 and is thus unable to phosphorylate α-synuclein. Interestingly, a significant reduction in α-synuclein phosphorylation has been observed in primary cortical neurons and the cerebral cortex of PLK2 knockout mice [49]. Bergeron et al. further demonstrated the ability of PLK2 to phosphorylate α-synuclein at Ser129 in the brain of mice in vivo [51]. These data show that polo-like kinase is critical for α-synuclein phosphorylation in the pathogenesis of α-synucleinopathy. Furthermore, dephosphorylated α-synuclein binding inhibits the activity of tyrosine hydroxylase (TH) [63], the rate-limiting enzyme in dopamine biosynthesis. Interestingly, phosphorylation of α-synuclein at Ser129 by PLK2 in vitro reduced the ability of α-synuclein to inhibit TH [64]. Active phosphorylated TH is easily aggregated [65][66] and degraded by the ubiquitin-proteasome system [67][68], indicating the attenuation of dopaminergic function. These data suggest the relevance of PLK2 in the reduced dopamine levels observed in Parkinson’s disease in an α-synuclein phosphorylation-dependent manner.

3. Pathological Impact of Phosphorylated Synuclein and Its Potential as a Novel Therapeutic Target

Previous investigations involving immunohistochemical and biochemical analyses have accumulated much data on Lewy bodies, showing the deposition of α-synuclein phosphorylated at Ser129 in postmortem brains of patients with Parkinson’s disease and DLB. Thus, it is worth investigating when the accumulation of phosphorylated protein begins in the various stages of Lewy body diseases. In this section, researchers summarize when phosphorylation at Ser129 occurs during the pathogenesis of Lewy body diseases and propose potential therapeutic targets to modulate α-synuclein phosphorylation.
Previously, Walker et al. reported that a biochemical analysis revealed progressive accumulation of p-Ser129 immunoreactivities as well as a positive correlation between phosphorylation level and the severity of symptoms of Parkinson’s disease and DLB [69]. Moreover, the accumulation of insoluble phosphorylated forms and the formation of p-Ser129-positive insoluble species became detectable only at the late stages of the diseases, stages IV and V, according to the Unified Staging System [69][70]. Zhou et al. also reported a dramatic accumulation of p-Ser129-positive inclusions in various brain regions at the late stages of the disease using postmortem brain samples from patients with Parkinson’s disease [71]. These findings revealed that pathogenic deposition of insoluble α-synuclein phosphorylated at Ser129 primarily occurs at the advanced stages of synucleinopathies.
Many kinases are activated in Parkinson’s disease and related disorders and are associated to the diseases’ pathogenesis. Focusing on the kinases that have the ability to phosphorylate α-synuclein (Figure 3), GRK, CK2, and PLK indeed alter the activity by their modification in Lewy body diseases. GRK6 and CK2α can be S-nitrosylated by NO, with S-nitrosylation enhancing their kinase activity towards the phosphorylation of α-synuclein at Ser129 [46]. Notably, in an A53T α-synuclein transgenic mouse, a familial Parkinson’s disease model, Wu et al. demonstrated that increased GRK6 and CK2α S-nitrosylation was age-dependent and associated with an increased level of p-Ser129 α-synuclein, which suggests GRK6 and CK2α S-nitrosylation as a potential therapeutic target of α-synucleinopathy. In fact, treatment of A53T α-synuclein transgenic mice with Nω-nitro-L-arginine (L-NNA), a NO synthase inhibitor, significantly reduced the S-nitrosylation of GRK6 and CK2α in the brain [46]. Additionally, Chau et al. reported that pramipexole (PPX) decreases the phosphorylation of CK2 to reduce its activity, thereby inhibiting the phosphorylation of α-synuclein at Ser129 [72]. Observation of LRRK2 phosphorylation by casein kinase 1α [73] also raises the possible development of another therapeutic agent.
PLK is also associated with the pathogenesis of Parkinson’s disease. Oxidative stress generated as a consequence of mitochondrial dysfunction can activate PLK2 [74]. PLK2 knockout reduces presynaptic terminal α-synuclein Ser129 phosphorylation [75]. In this condition, PLK2 deletion has no effect on the phosphorylation levels in Lewy-body-like inclusions [75], suggesting that the formation of inclusions is α-synuclein-phosphorylation-independent. Furthermore, PLK2 deletion slows the rate of neuronal death [75], indicating that inhibition of PLK2 is a potential therapeutic target. Treatment with the PLK inhibitor BI2536 inhibited α-synuclein phosphorylation at Ser129 [49]. Furthermore, treatment with a designed highly selective inhibitor for PLK2, compound 37, reduced α-synuclein phosphorylation in rat brain [76][77]. However, the inhibitor had no effect on α-synuclein aggregation, p-Ser129, or inter-neuronal spreading of the aggregated α-synuclein in the S129A preformed fibril injection model [78]. These findings suggest that PLK2 could be a potential target for the development of novel therapies, although further optimization is still required at the current stage (Figure 4).
Figure 4. Schematic pathways of α-synuclein phosphorylation by kinases in the neurodegeneration process and potential pharmacotherapeutic targets [46][72][74][75][76][77][78]. NO: nitric oxide; NOS: NO synthase; ROS: reactive oxygen species; L-NNA: N(omega)-nitro-L-arginine, a competitive inhibitor of nitric oxide synthase; GRK: G-protein-coupled receptor kinase; CKII: casein kinase II; PLK: polo-like kinase; PPX: pramipexole.

References

  1. Maroteaux, L.; Campanelli, J.T.; Scheller, R.H. Synuclein: A neuron-specific protein localized to the nucleus and presynaptic nerve terminal. J. Neurosci. 1988, 8, 2804–2815.
  2. Ulmer, T.S.; Bax, A.; Cole, N.B.; Nussbaum, R.L. Structure and dynamics of micelle-bound human alpha-synuclein. J. Biol. Chem. 2005, 280, 9595–9603.
  3. Goedert, M. Alpha-synuclein and neurodegenerative diseases. Nat. Rev. Neurosci. 2001, 2, 492–501.
  4. Araki, K.; Yagi, N.; Nakatani, R.; Sekiguchi, H.; So, M.; Yagi, H.; Ohta, N.; Nagai, Y.; Goto, Y.; Mochizuki, H. A small-angle X-ray scattering study of alpha-synuclein from human red blood cells. Sci. Rep. 2016, 6, 30473.
  5. Wu, K.P.; Kim, S.; Fela, D.A.; Baum, J. Characterization of conformational and dynamic properties of natively unfolded human and mouse alpha-synuclein ensembles by NMR: Implication for aggregation. J. Mol. Biol. 2008, 378, 1104–1115.
  6. Pirc, K.; Ulrih, N.P. alpha-Synuclein interactions with phospholipid model membranes: Key roles for electrostatic interactions and lipid-bilayer structure. Biochim. Biophys. Acta 2015, 1848, 2002–2012.
  7. Hasegawa, T.; Konno, M.; Baba, T.; Sugeno, N.; Kikuchi, A.; Kobayashi, M.; Miura, E.; Tanaka, N.; Tamai, K.; Furukawa, K.; et al. The AAA-ATPase VPS4 regulates extracellular secretion and lysosomal targeting of alpha-synuclein. PLoS ONE 2011, 6, e29460.
  8. Tokuda, T.; Salem, S.A.; Allsop, D.; Mizuno, T.; Nakagawa, M.; Qureshi, M.M.; Locascio, J.J.; Schlossmacher, M.G.; El-Agnaf, O.M. Decreased alpha-synuclein in cerebrospinal fluid of aged individuals and subjects with Parkinson’s disease. Biochem. Biophys. Res. Commun. 2006, 349, 162–166.
  9. El-Agnaf, O.M.; Salem, S.A.; Paleologou, K.E.; Curran, M.D.; Gibson, M.J.; Court, J.A.; Schlossmacher, M.G.; Allsop, D. Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006, 20, 419–425.
  10. Eller, M.; Williams, D.R. Biological fluid biomarkers in neurodegenerative parkinsonism. Nat. Rev. Neurol. 2009, 5, 561–570.
  11. Jensen, P.H.; Hager, H.; Nielsen, M.S.; Hojrup, P.; Gliemann, J.; Jakes, R. alpha-synuclein binds to Tau and stimulates the protein kinase A-catalyzed tau phosphorylation of serine residues 262 and 356. J. Biol. Chem. 1999, 274, 25481–25489.
  12. Giasson, B.I.; Forman, M.S.; Higuchi, M.; Golbe, L.I.; Graves, C.L.; Kotzbauer, P.T.; Trojanowski, J.Q.; Lee, V.M. Initiation and synergistic fibrillization of tau and alpha-synuclein. Science 2003, 300, 636–640.
  13. Clinton, L.K.; Blurton-Jones, M.; Myczek, K.; Trojanowski, J.Q.; LaFerla, F.M. Synergistic Interactions between Abeta, tau, and alpha-synuclein: Acceleration of neuropathology and cognitive decline. J. Neurosci. 2010, 30, 7281–7289.
  14. Shrivastava, A.N.; Redeker, V.; Fritz, N.; Pieri, L.; Almeida, L.G.; Spolidoro, M.; Liebmann, T.; Bousset, L.; Renner, M.; Lena, C.; et al. alpha-synuclein assemblies sequester neuronal alpha3-Na+/K+-ATPase and impair Na+ gradient. EMBO J. 2015, 34, 2408–2423.
  15. Rodriguez, L.; Marano, M.M.; Tandon, A. Import and Export of Misfolded alpha-Synuclein. Front. Neurosci. 2018, 12, 344.
  16. Mao, X.; Ou, M.T.; Karuppagounder, S.S.; Kam, T.I.; Yin, X.; Xiong, Y.; Ge, P.; Umanah, G.E.; Brahmachari, S.; Shin, J.H.; et al. Pathological alpha-synuclein transmission initiated by binding lymphocyte-activation gene 3. Science 2016, 353, aah3374.
  17. Kobayashi, J.; Hasegawa, T.; Sugeno, N.; Yoshida, S.; Akiyama, T.; Fujimori, K.; Hatakeyama, H.; Miki, Y.; Tomiyama, A.; Kawata, Y.; et al. Extracellular alpha-synuclein enters dopaminergic cells by modulating flotillin-1-assisted dopamine transporter endocytosis. FASEB J. 2019, 33, 10240–10256.
  18. Kawahata, I.; Bousset, L.; Melki, R.; Fukunaga, K. Fatty Acid-Binding Protein 3 is Critical for alpha-Synuclein Uptake and MPP(+)-Induced Mitochondrial Dysfunction in Cultured Dopaminergic Neurons. Int. J. Mol. Sci. 2019, 20, 5358.
  19. Fukui, N.; Yamamoto, H.; Miyabe, M.; Aoyama, Y.; Hongo, K.; Mizobata, T.; Kawahata, I.; Yabuki, Y.; Shinoda, Y.; Fukunaga, K.; et al. An alpha-synuclein decoy peptide prevents cytotoxic alpha-synuclein aggregation caused by fatty acid binding protein 3. J. Biol. Chem. 2021, 296, 100663.
  20. Kawahata, I.; Sekimori, T.; Wang, H.; Wang, Y.; Sasaoka, T.; Bousset, L.; Melki, R.; Mizobata, T.; Kawata, Y.; Fukunaga, K. Dopamine D2 Long Receptors Are Critical for Caveolae-Mediated alpha-Synuclein Uptake in Cultured Dopaminergic Neurons. Biomedicines 2021, 9, 49.
  21. Kawahata, I.; Fukunaga, K. Impact of fatty acid-binding proteins and dopamine receptors on alpha-synucleinopathy. J. Pharmacol. Sci. 2022, 148, 248–254.
  22. Paik, S.R.; Shin, H.J.; Lee, J.H.; Chang, C.S.; Kim, J. Copper(II)-induced self-oligomerization of alpha-synuclein. Biochem. J. 1999, 340 Pt 3, 821–828.
  23. Brown, D.R. Interactions between metals and alpha-synuclein—Function or artefact? FEBS J. 2007, 274, 3766–3774.
  24. Sugeno, N.; Jäckel, S.; Voigt, A.; Wassouf, Z.; Schulze-Hentrich, J.; Kahle, P.J. α-Synuclein enhances histone H3 lysine-9 dimethylation and H3K9me2-dependent transcriptional responses. Sci. Rep. 2016, 6, 36328.
  25. Goers, J.; Manning-Bog, A.B.; McCormack, A.L.; Millett, I.S.; Doniach, S.; Di Monte, D.A.; Uversky, V.N.; Fink, A.L. Nuclear localization of alpha-synuclein and its interaction with histones. Biochemistry 2003, 42, 8465–8471.
  26. Jiang, K.; Rocha, S.; Westling, A.; Kesarimangalam, S.; Dorfman, K.D.; Wittung-Stafshede, P.; Westerlund, F. Alpha-Synuclein Modulates the Physical Properties of DNA. Chemistry 2018, 24, 15685–15690.
  27. Tenreiro, S.; Eckermann, K.; Outeiro, T.F. Protein phosphorylation in neurodegeneration: Friend or foe? Front. Mol. Neurosci. 2014, 7, 42.
  28. Paleologou, K.E.; El-Agnaf, O.M. α-Synuclein aggregation and modulating factors. Subcell. Biochem. 2012, 65, 109–164.
  29. Oueslati, A.; Fournier, M.; Lashuel, H.A. Role of post-translational modifications in modulating the structure, function and toxicity of alpha-synuclein: Implications for Parkinson’s disease pathogenesis and therapies. Prog. Brain Res. 2010, 183, 115–145.
  30. Hasegawa, M.; Fujiwara, H.; Nonaka, T.; Wakabayashi, K.; Takahashi, H.; Lee, V.M.; Trojanowski, J.Q.; Mann, D.; Iwatsubo, T. Phosphorylated alpha-synuclein is ubiquitinated in alpha-synucleinopathy lesions. J. Biol. Chem. 2002, 277, 49071–49076.
  31. Fujiwara, H.; Hasegawa, M.; Dohmae, N.; Kawashima, A.; Masliah, E.; Goldberg, M.S.; Shen, J.; Takio, K.; Iwatsubo, T. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat. Cell Biol. 2002, 4, 160–164.
  32. Muntane, G.; Ferrer, I.; Martinez-Vicente, M. alpha-synuclein phosphorylation and truncation are normal events in the adult human brain. Neuroscience 2012, 200, 106–119.
  33. Okochi, M.; Walter, J.; Koyama, A.; Nakajo, S.; Baba, M.; Iwatsubo, T.; Meijer, L.; Kahle, P.J.; Haass, C. Constitutive phosphorylation of the Parkinson’s disease associated alpha-synuclein. J. Biol. Chem. 2000, 275, 390–397.
  34. Kahle, P.J.; Neumann, M.; Ozmen, L.; Haass, C. Physiology and pathophysiology of alpha-synuclein. Cell culture and transgenic animal models based on a Parkinson’s disease-associated protein. Ann. N. Y. Acad. Sci. 2000, 920, 33–41.
  35. Anderson, J.P.; Walker, D.E.; Goldstein, J.M.; de Laat, R.; Banducci, K.; Caccavello, R.J.; Barbour, R.; Huang, J.; Kling, K.; Lee, M.; et al. Phosphorylation of Ser-129 is the dominant pathological modification of alpha-synuclein in familial and sporadic Lewy body disease. J. Biol. Chem. 2006, 281, 29739–29752.
  36. Neumann, M.; Kahle, P.J.; Giasson, B.I.; Ozmen, L.; Borroni, E.; Spooren, W.; Muller, V.; Odoy, S.; Fujiwara, H.; Hasegawa, M.; et al. Misfolded proteinase K-resistant hyperphosphorylated alpha-synuclein in aged transgenic mice with locomotor deterioration and in human alpha-synucleinopathies. J. Clin. Investig. 2002, 110, 1429–1439.
  37. Takahashi, M.; Kanuka, H.; Fujiwara, H.; Koyama, A.; Hasegawa, M.; Miura, M.; Iwatsubo, T. Phosphorylation of alpha-synuclein characteristic of synucleinopathy lesions is recapitulated in alpha-synuclein transgenic Drosophila. Neurosci. Lett. 2003, 336, 155–158.
  38. Yamada, M.; Iwatsubo, T.; Mizuno, Y.; Mochizuki, H. Overexpression of alpha-synuclein in rat substantia nigra results in loss of dopaminergic neurons, phosphorylation of alpha-synuclein and activation of caspase-9: Resemblance to pathogenetic changes in Parkinson’s disease. J. Neurochem. 2004, 91, 451–461.
  39. Chen, L.; Feany, M.B. Alpha-synuclein phosphorylation controls neurotoxicity and inclusion formation in a Drosophila model of Parkinson disease. Nat. Neurosci. 2005, 8, 657–663.
  40. Xu, J.; Kawahata, I.; Izumi, H.; Fukunaga, K. T-Type Ca(2+) Enhancer SAK3 Activates CaMKII and Proteasome Activities in Lewy Body Dementia Mice Model. Int. J. Mol. Sci. 2021, 22, 6185.
  41. Bertini, I.; Gupta, Y.K.; Luchinat, C.; Parigi, G.; Peana, M.; Sgheri, L.; Yuan, J. Paramagnetism-based NMR restraints provide maximum allowed probabilities for the different conformations of partially independent protein domains. J. Am. Chem. Soc. 2007, 129, 12786–12794.
  42. Eliezer, D.; Kutluay, E.; Bussell, R., Jr.; Browne, G. Conformational properties of alpha-synuclein in its free and lipid-associated states. J. Mol. Biol. 2001, 307, 1061–1073.
  43. Del Mar, C.; Greenbaum, E.A.; Mayne, L.; Englander, S.W.; Woods, V.L., Jr. Structure and properties of alpha-synuclein and other amyloids determined at the amino acid level. Proc. Natl. Acad. Sci. USA 2005, 102, 15477–15482.
  44. Pronin, A.N.; Morris, A.J.; Surguchov, A.; Benovic, J.L. Synucleins are a novel class of substrates for G protein-coupled receptor kinases. J. Biol. Chem. 2000, 275, 26515–26522.
  45. Arawaka, S.; Wada, M.; Goto, S.; Karube, H.; Sakamoto, M.; Ren, C.H.; Koyama, S.; Nagasawa, H.; Kimura, H.; Kawanami, T.; et al. The role of G-protein-coupled receptor kinase 5 in pathogenesis of sporadic Parkinson’s disease. J. Neurosci. 2006, 26, 9227–9238.
  46. Wu, W.; Sung, C.C.; Yu, P.; Li, J.; Chung, K.K.K. S-Nitrosylation of G protein-coupled receptor kinase 6 and Casein kinase 2 alpha modulates their kinase activity toward alpha-synuclein phosphorylation in an animal model of Parkinson’s disease. PLoS ONE 2020, 15, e0232019.
  47. Ishii, A.; Nonaka, T.; Taniguchi, S.; Saito, T.; Arai, T.; Mann, D.; Iwatsubo, T.; Hisanaga, S.; Goedert, M.; Hasegawa, M. Casein kinase 2 is the major enzyme in brain that phosphorylates Ser129 of human alpha-synuclein: Implication for alpha-synucleinopathies. FEBS Lett. 2007, 581, 4711–4717.
  48. Sano, K.; Iwasaki, Y.; Yamashita, Y.; Irie, K.; Hosokawa, M.; Satoh, K.; Mishima, K. Tyrosine 136 phosphorylation of α-synuclein aggregates in the Lewy body dementia brain: Involvement of serine 129 phosphorylation by casein kinase 2. Acta Neuropathol. Commun. 2021, 9, 182.
  49. Inglis, K.J.; Chereau, D.; Brigham, E.F.; Chiou, S.S.; Schöbel, S.; Frigon, N.L.; Yu, M.; Caccavello, R.J.; Nelson, S.; Motter, R.; et al. Polo-like kinase 2 (PLK2) phosphorylates alpha-synuclein at serine 129 in central nervous system. J. Biol. Chem. 2009, 284, 2598–2602.
  50. Mbefo, M.K.; Paleologou, K.E.; Boucharaba, A.; Oueslati, A.; Schell, H.; Fournier, M.; Olschewski, D.; Yin, G.; Zweckstetter, M.; Masliah, E.; et al. Phosphorylation of synucleins by members of the Polo-like kinase family. J. Biol. Chem. 2010, 285, 2807–2822.
  51. Bergeron, M.; Motter, R.; Tanaka, P.; Fauss, D.; Babcock, M.; Chiou, S.S.; Nelson, S.; San Pablo, F.; Anderson, J.P. In Vivo modulation of polo-like kinases supports a key role for PLK2 in Ser129 α-synuclein phosphorylation in mouse brain. Neuroscience 2014, 256, 72–82.
  52. Qing, H.; Wong, W.; McGeer, E.G.; McGeer, P.L. Lrrk2 phosphorylates alpha synuclein at serine 129: Parkinson disease implications. Biochem. Biophys. Res. Commun. 2009, 387, 149–152.
  53. Qing, H.; Zhang, Y.; Deng, Y.; McGeer, E.G.; McGeer, P.L. Lrrk2 interaction with alpha-synuclein in diffuse Lewy body disease. Biochem. Biophys. Res. Commun. 2009, 390, 1229–1234.
  54. Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912.
  55. Nübling, G.S.; Levin, J.; Bader, B.; Lorenzl, S.; Hillmer, A.; Högen, T.; Kamp, F.; Giese, A. Modelling Ser129 phosphorylation inhibits membrane binding of pore-forming alpha-synuclein oligomers. PLoS ONE 2014, 9, e98906.
  56. Fiske, M.; Valtierra, S.; Solvang, K.; Zorniak, M.; White, M.; Herrera, S.; Konnikova, A.; Brezinsky, R.; Debburman, S. Contribution of Alanine-76 and Serine Phosphorylation in α-Synuclein Membrane Association and Aggregation in Yeasts. Parkinsons Dis. 2011, 2011, 392180.
  57. Kuwahara, T.; Tonegawa, R.; Ito, G.; Mitani, S.; Iwatsubo, T. Phosphorylation of α-synuclein protein at Ser-129 reduces neuronal dysfunction by lowering its membrane binding property in Caenorhabditis elegans. J. Biol. Chem. 2012, 287, 7098–7109.
  58. Wang, X.; Becker, K.; Levine, N.; Zhang, M.; Lieberman, A.P.; Moore, D.J.; Ma, J. Pathogenic alpha-synuclein aggregates preferentially bind to mitochondria and affect cellular respiration. Acta Neuropathol. Commun. 2019, 7, 41.
  59. Di Maio, R.; Barrett, P.J.; Hoffman, E.K.; Barrett, C.W.; Zharikov, A.; Borah, A.; Hu, X.; McCoy, J.; Chu, C.T.; Burton, E.A.; et al. alpha-Synuclein binds to TOM20 and inhibits mitochondrial protein import in Parkinson’s disease. Sci. Transl. Med. 2016, 8, 342ra378.
  60. Ryu, M.Y.; Kim, D.W.; Arima, K.; Mouradian, M.M.; Kim, S.U.; Lee, G. Localization of CKII beta subunits in Lewy bodies of Parkinson’s disease. J. Neurol. Sci. 2008, 266, 9–12.
  61. De Carcer, G.; Manning, G.; Malumbres, M. From Plk1 to Plk5: Functional evolution of polo-like kinases. Cell Cycle 2011, 10, 2255–2262.
  62. Ahmad, K.A.; Wang, G.; Unger, G.; Slaton, J.; Ahmed, K. Protein kinase CK2—A key suppressor of apoptosis. Adv. Enzym. Regul. 2008, 48, 179–187.
  63. Perez, R.G.; Waymire, J.C.; Lin, E.; Liu, J.J.; Guo, F.; Zigmond, M.J. A role for alpha-synuclein in the regulation of dopamine biosynthesis. J. Neurosci. 2002, 22, 3090–3099.
  64. Lou, H.; Montoya, S.E.; Alerte, T.N.; Wang, J.; Wu, J.; Peng, X.; Hong, C.S.; Friedrich, E.E.; Mader, S.A.; Pedersen, C.J.; et al. Serine 129 phosphorylation reduces the ability of alpha-synuclein to regulate tyrosine hydroxylase and protein phosphatase 2A in vitro and in vivo. J. Biol. Chem. 2010, 285, 17648–17661.
  65. Kawahata, I.; Tokuoka, H.; Parvez, H.; Ichinose, H. Accumulation of phosphorylated tyrosine hydroxylase into insoluble protein aggregates by inhibition of an ubiquitin-proteasome system in PC12D cells. J. Neural Transm. 2009, 116, 1571–1578.
  66. Kawahata, I.; Yagishita, S.; Hasegawa, K.; Nagatsu, I.; Nagatsu, T.; Ichinose, H. Immunohistochemical analyses of the postmortem human brains from patients with Parkinson’s disease with anti-tyrosine hydroxylase antibodies. Biog. Amines 2009, 23, 1–7.
  67. Kawahata, I.; Ohtaku, S.; Tomioka, Y.; Ichinose, H.; Yamakuni, T. Dopamine or biopterin deficiency potentiates phosphorylation at (40)Ser and ubiquitination of tyrosine hydroxylase to be degraded by the ubiquitin proteasome system. Biochem. Biophys. Res. Commun. 2015, 465, 53–58.
  68. Kawahata, I.; Fukunaga, K. Degradation of Tyrosine Hydroxylase by the Ubiquitin-Proteasome System in the Pathogenesis of Parkinson’s Disease and Dopa-Responsive Dystonia. Int. J. Mol. Sci. 2020, 21, 3779.
  69. Walker, D.G.; Lue, L.F.; Adler, C.H.; Shill, H.A.; Caviness, J.N.; Sabbagh, M.N.; Akiyama, H.; Serrano, G.E.; Sue, L.I.; Beach, T.G.; et al. Changes in properties of serine 129 phosphorylated alpha-synuclein with progression of Lewy-type histopathology in human brains. Exp. Neurol. 2013, 240, 190–204.
  70. Beach, T.G.; Adler, C.H.; Lue, L.; Sue, L.I.; Bachalakuri, J.; Henry-Watson, J.; Sasse, J.; Boyer, S.; Shirohi, S.; Brooks, R.; et al. Unified staging system for Lewy body disorders: Correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. 2009, 117, 613–634.
  71. Zhou, J.; Broe, M.; Huang, Y.; Anderson, J.P.; Gai, W.P.; Milward, E.A.; Porritt, M.; Howells, D.; Hughes, A.J.; Wang, X.; et al. Changes in the solubility and phosphorylation of alpha-synuclein over the course of Parkinson’s disease. Acta Neuropathol. 2011, 121, 695–704.
  72. Chau, K.-Y.; Cooper, J.M.; Schapira, A.H.V. Pramipexole reduces phosphorylation of α-synuclein at serine-129. J. Mol. Neurosci. 2013, 51, 573–580.
  73. Chia, R.; Haddock, S.; Beilina, A.; Rudenko, I.N.; Mamais, A.; Kaganovich, A.; Li, Y.; Kumaran, R.; Nalls, M.A.; Cookson, M.R. Phosphorylation of LRRK2 by casein kinase 1alpha regulates trans-Golgi clustering via differential interaction with ARHGEF7. Nat. Commun. 2014, 5, 5827.
  74. Li, J.; Ma, W.; Wang, P.Y.; Hurley, P.J.; Bunz, F.; Hwang, P.M. Polo-like kinase 2 activates an antioxidant pathway to promote the survival of cells with mitochondrial dysfunction. Free Radic. Biol. Med. 2014, 73, 270–277.
  75. Weston, L.J.; Stackhouse, T.L.; Spinelli, K.J.; Boutros, S.W.; Rose, E.P.; Osterberg, V.R.; Luk, K.C.; Raber, J.; Weissman, T.A.; Unni, V.K. Genetic deletion of Polo-like kinase 2 reduces alpha-synuclein serine-129 phosphorylation in presynaptic terminals but not Lewy bodies. J. Biol. Chem. 2021, 296, 100273.
  76. Bowers, S.; Truong, A.P.; Ye, M.; Aubele, D.L.; Sealy, J.M.; Neitz, R.J.; Hom, R.K.; Chan, W.; Dappen, M.S.; Galemmo, R.A., Jr.; et al. Design and synthesis of highly selective, orally active Polo-like kinase-2 (Plk-2) inhibitors. Bioorganic Med. Chem. Lett. 2013, 23, 2743–2749.
  77. Aubele, D.L.; Hom, R.K.; Adler, M.; Galemmo, R.A., Jr.; Bowers, S.; Truong, A.P.; Pan, H.; Beroza, P.; Neitz, R.J.; Yao, N.; et al. Selective and brain-permeable polo-like kinase-2 (Plk-2) inhibitors that reduce alpha-synuclein phosphorylation in rat brain. Chem. Med. Chem. 2013, 8, 1295–1313.
  78. Elfarrash, S.; Jensen, N.M.; Ferreira, N.; Schmidt, S.I.; Gregersen, E.; Vestergaard, M.V.; Nabavi, S.; Meyer, M.; Jensen, P.H. Polo-like kinase 2 inhibition reduces serine-129 phosphorylation of physiological nuclear alpha-synuclein but not of the aggregated alpha-synuclein. PLoS ONE 2021, 16, e0252635.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 428
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 08 Jun 2022
1000/1000