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 -- 979 2023-08-09 21:59:37 |
2 format correction Meta information modification 979 2023-08-10 02:32:34 |

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.
Lucchini, R.; Tieu, K. Genes Documented to Interact with Mn. Encyclopedia. Available online: https://encyclopedia.pub/entry/47857 (accessed on 10 September 2024).
Lucchini R, Tieu K. Genes Documented to Interact with Mn. Encyclopedia. Available at: https://encyclopedia.pub/entry/47857. Accessed September 10, 2024.
Lucchini, Roberto, Kim Tieu. "Genes Documented to Interact with Mn" Encyclopedia, https://encyclopedia.pub/entry/47857 (accessed September 10, 2024).
Lucchini, R., & Tieu, K. (2023, August 09). Genes Documented to Interact with Mn. In Encyclopedia. https://encyclopedia.pub/entry/47857
Lucchini, Roberto and Kim Tieu. "Genes Documented to Interact with Mn." Encyclopedia. Web. 09 August, 2023.
Genes Documented to Interact with Mn
Edit

Manganese (Mn) exposure has evolved from acute, high-level exposure causing manganism to low, chronic lifetime exposure. In this latter scenario, the target areas extend beyond the globus pallidus (as seen with manganism) to the entire basal ganglia, including the substantia nigra pars compacta. This change of exposure paradigm has prompted numerous epidemiological investigations of the occurrence of Parkinson’s disease (PD), or parkinsonism, due to the long-term impact of Mn. In parallel, experimental research has focused on the underlying pathogenic mechanisms of Mn and its interactions with genetic susceptibility.

manganese manganism neurotoxicity parkinsonism Parkinson’s disease

1. SNCA

SNCA encodes α-synuclein, which is predominantly a synaptic protein that is encoded by the SNCA gene. Missense mutations as well as mutations leading to duplications and triplications of SNCA have been identified in autosomal dominant PD [1][2][3][4][5][6]. The discoveries that increasing the gene dosage of SNCA by 2- to 3-fold can cause PD [5] is significant for idiopathic PD, because it indicates that elevated wild-type (WT) α-synuclein alone is sufficient to cause the disease. In addition to familial PD, α-synuclein aggregation is also detected in the Lewy bodies of idiopathic PD [7]. Lewy bodies are intracellular inclusions that consist of aggregated α-synuclein, and more recently, they have also been reported to contain lipids and organelles such as mitochondria [8]. Pathogenic mechanisms associated with α-synuclein mutations range from impairing protein degradation pathways (autophagy and ubiquitin proteasomal system), mitochondrial dysfunction, oxidative stress, synaptic dysfunction, and neuroinflammation [9]. There has also been a significant interest in the spread of α-synuclein pathology from one cell to another in a prion-like fashion [10] and Mn has been demonstrated to enhance this transmission [11].
Mn can bind to α-synuclein via three residues in the C-terminal domain: Asp-121, Asn-122, and Glu-123 [12]. Although this is a low-affinity binding, low concentrations of Mn are sufficient to induce α-synuclein fibril formation [13]. Several independent laboratories have reported the effects of Mn on the aggregation of α-synuclein [14]. The interaction between α-synuclein and Mn has been demonstrated in rat primary midbrain dopaminergic neurons, resulting in a higher intracellular level of Mn in cells with α-synuclein overexpression [15][16]. Although such binding of α-synuclein is protective against Mn neurotoxicity during the early stages of Mn exposure, the accumulation of sequestered Mn by α-synuclein eventually induces protein aggregation and neurotoxicity [17]. In a more recent study, these investigators demonstrated that when combined with α-synuclein, Mn exposure increases α-synuclein transmission intercellularly via exosome release [11]. By spreading the misfolded toxic α-synuclein, these exosomes induce neuroinflammation and degeneration of the nigral dopaminergic neurons [11]. This is the first in vivo demonstration that, although Mn by itself does not cause neurodegeneration, when combined with a PD-linked protein, nigral dopaminergic cell loss occurs. The results of these studies are consistent with a previous report demonstrating that α-synuclein overexpressing neuronal cells released exosomes capable of transferring α-syn protein to other normal neuronal cells [18], forming aggregates and inducing death in the receiving cell [19][20]. In fact, exosome-associated α-synuclein oligomers are more likely to be taken up by recipient cells and are more neurotoxic compared to free α-syn oligomers [21]. To enhance the relevance of their mouse study, Kanthasamy and colleagues isolated exosomes in the serum of welders exposed to Mn and they found increased misfolded α-synuclein in these workers as compared to the non-exposed control subjects [11]. In non-human primates, Mn exposure has also been reported to induce α-synuclein aggregation [22]. In combination, emerging evidence indicates that Mn promotes α-synuclein aggregation and spread, leading to dopaminergic neurodegeneration in experimental models and suggests that such a scenario may also occur in humans [23].

2. Parkin

Mutations in Parkin cause autosomal recessive early-onset PD [24][25], which may present with or without Lewy bodies [25][26]. Parkin mutations are considered as the most prevalent autosomal recessive mutations in PD, accounting for approximately up to 77% of familial early-onset PD and 10–20% of early-onset PD in general [27][28][29]. Parkin is an E3 ubiquitin ligase that mediates protein degradation. Parkin has been reported to regulate Mn transport via the divalent metal transporter 1 (DMT1) [30]. DMT1 is the primary route by which Mn is taken up in the brain. Four DMT1 isoforms have been identified and the 1B isoform is regulated post-translationally and degraded via the proteasomal pathway. Parkin is involved in the ubiquitination and mediate the subsequent proteasomal degradation of this specific isoform of DMT1 [30]. Therefore, a loss of parkin function could facilitate Mn uptake, promoting Mn accumulation in the basal ganglia and accelerating its toxicity.

3. DJ-1

Mutations in DJ-1 cause early-onset autosomal recessive PD [31]. DJ-1 is a multifunctional protein. Under basal conditions, DJ-1 is localized mostly in the cytoplasm and to a lesser extent, in the mitochondria and nucleus [32][33][34]. However, under oxidative stress conditions, cytoplasmic DJ-1 translocates to mitochondria [34], where it confers protection against oxidative stress [34][35]. Mn has been reported to reduce the levels of DJ-1, resulting in effects that resemble the neurotoxic activity of mutant forms of DJ-1 [36]. Given that both Mn and loss of DJ-1 function can individually promote oxidative stress and induce mitochondrial dysfunction, such combination may exacerbate their neurotoxic effects. Indeed, the life-span of C. elegans was reduced after Mn exposure when DJ-1 was deleted [37].

4. ATP13A2

In addition to causing autosomal recessive juvenile-onset Kufor–Rakeb syndrome [38], which is a levodopa-responsive form of pallido-pyramidal neurodegeration, homozygous missense mutations in ATP13A2 cause juvenile parkinsonism and early-onset autosomal recessive PD [39]. ATP13A2 encodes ATPase type 13A, a lysosomal P5B-type transmembrane ATPase which has functional domains like other P-type ATPases that are mainly involved in transporting cations, including Mn. Consistent with these roles of ATP13A2 and its cellular localization, loss of function in this protein has been reported to impair autophagy flux and accumulation of aggregated α-synuclein [40][41][42]. ATP13A2 has been shown to protect cells against Mn toxicity by transporting Mn into lysosomes; however, this protection is lost when mutations in this gene occur [42]. A recent study has shown that ATP13A2 polymorphism could negatively impact the effects of Mn on motor coordination in humans exposed to Mn [43].

References

  1. Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; Boyer, R.; et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997, 276, 2045–2047.
  2. Appel-Cresswell, S.; Vilarino-Guell, C.; Encarnacion, M.; Sherman, H.; Yu, I.; Shah, B.; Weir, D.; Thompson, C.; Szu-Tu, C.; Trinh, J.; et al. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. 2013, 28, 811–813.
  3. Kruger, R.; Kuhn, W.; Muller, T.; Woitalla, D.; Graeber, M.; Kosel, S.; Pzuntek, H.; Epplen, J.T.; Schols, L.; Riess, O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat. Genet. 1998, 18, 107–108.
  4. Lesage, S.; Anheim, M.; Letournel, F.; Bousset, L.; Honore, A.; Rozas, N.; Pieri, L.; Madiona, K.; Durr, A.; Melki, R.; et al. G51D alpha-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann. Neurol. 2013, 73, 459–471.
  5. Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841.
  6. Zarranz, J.J.; Alegre, J.; Gomez-Esteban, J.C.; Lezcano, E.; Ros, R.; Ampuero, I.; Vidal, L.; Hoenicka, J.; Rodriguez, O.; Atares, B.; et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 2004, 55, 164–173.
  7. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.Y.; Trojanowski, J.Q.; Roos, J.; Goedert, M. a-Synuclein in Lewy bodies. Nature 1997, 388, 839–840.
  8. Shahmoradian, S.H.; Lewis, A.J.; Genoud, C.; Hench, J.; Moors, T.E.; Navarro, P.P.; Castano-Diez, D.; Schweighauser, G.; Graff-Meyer, A.; Goldie, K.N.; et al. Lewy pathology in Parkinson’s disease consists of crowded organelles and lipid membranes. Nat. Neurosci. 2019, 22, 1099–1109.
  9. Guo, J.L.; Lee, V.M. Cell-to-cell transmission of pathogenic proteins in neurodegenerative diseases. Nat. Med. 2014, 20, 130–138.
  10. Pinnell, J.R.; Cui, M.; Tieu, K. Exosomes in Parkinson disease. J. Neurochem. 2020, 157, 413–428.
  11. Harischandra, D.S.; Rokad, D.; Neal, M.L.; Ghaisas, S.; Manne, S.; Sarkar, S.; Panicker, N.; Zenitsky, G.; Jin, H.; Lewis, M.; et al. Manganese promotes the aggregation and prion-like cell-to-cell exosomal transmission of alpha-synuclein. Sci. Signal. 2019, 12, eaau4543.
  12. Binolfi, A.; Rasia, R.M.; Bertoncini, C.W.; Ceolin, M.; Zweckstetter, M.; Griesinger, C.; Jovin, T.M.; Fernández, C.O. Interaction of alpha-synuclein with divalent metal ions reveals key differences: A link between structure, binding specificity and fibrillation enhancement. J. Am. Chem. Soc. 2006, 128, 9893–9901.
  13. Uversky, V.N.; Li, J.; Fink, A.L. Metal-triggered structural transformations, aggregation, and fibrillation of human alpha-synuclein. A possible molecular NK between Parkinson’s disease and heavy metal exposure. J. Biol. Chem. 2001, 276, 44284–44296.
  14. Peres, T.V.; Parmalee, N.L.; Martinez-Finley, E.J.; Aschner, M. Untangling the Manganese-alpha-Synuclein Web. Front. Neurosci. 2016, 10, 364.
  15. Prabhakaran, K.; Chapman, G.D.; Gunasekar, P.G. Alpha-Synuclein overexpression enhances manganese-induced neurotoxicity through the NF-kB-mediated pathway. Toxicol. Mech. Methods 2011, 21, 435–443.
  16. Ducic, T.; Carboni, E.; Lai, B.; Chen, S.; Michalke, B.; Lázaro, D.F.; Outeiro, T.F.; Bähr, M.; Barski, E.; Lingor, P. Alpha-Synuclein Regulates Neuronal Levels of Manganese and Calcium. ACS Chem. Neurosci. 2015, 6, 1769–1779.
  17. Harischandra, D.S.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. alpha-Synuclein protects against manganese neurotoxic insult during the early stages of exposure in a dopaminergic cell model of Parkinson’s disease. Toxicol. Sci. 2015, 143, 454–468.
  18. Alvarez-Erviti, L.; Seow, Y.; Schapira, A.H.; Gardiner, C.; Sargent, I.L.; Wood, M.J.; Cooper, J.M. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol. Dis. 2011, 42, 360–367.
  19. Desplats, P.; Lee, H.J.; Bae, E.J.; Patrick, C.; Rockenstein, E.; Crews, L.; Spencer, B.; Masliah, E.; Lee, S.J. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc. Natl. Acad. Sci. USA 2009, 106, 13010–13015.
  20. Hansen, C.; Angot, E.; Bergstrom, A.L.; Steiner, J.A.; Pieri, L.; Paul, G.; Outeiro, T.F.; Melki, R.; Kallunki, P.; Fog, K.; et al. alpha-Synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J. Clin. Invest. 2011, 121, 715–725.
  21. Danzer, K.M.; Kranich, L.R.; Ruf, W.P.; Cagsal-Getkin, O.; Winslow, A.R.; Zhu, L.; Vanderburg, C.R.; McLean, P.J. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 2012, 7, 42–47.
  22. Verina, T.; Schneider, J.S.; Guilarte, T.R.; Guilarte, T.R. Manganese exposure induces alpha-synuclein aggregation in the frontal cortex of non-human primates. Toxicol. Lett. 2013, 13, 177–183.
  23. Schuler, P.; Oyanguren, H.; Maturana, V.; Valenzuela, A.; Cruz, E.; Plaza, V.; Schmidt, E.; Haddad, R. Manganese poisoning; environmental and medical study at a Chilean mine. Ind. Med. Surg. 1957, 26, 167–173.
  24. Matsumine, H.; Saito, M.; Shimoda-Matsubayashi, S.; Tanaka, H.; Ishikawa, A.; Nakagawa-Hattori, Y.; Yokochi, M.; Kobayashi, T.; Igarashi, S.; Takano, H.; et al. Localization of a gene for an autosomal recessive form of juvenile Parkinsonism to chromosome 6q25.2-27. Am. J. Hum. Genet. 1997, 60, 588–596.
  25. Kitada, T.; Asakawa, S.; Hattori, N.; Matsumine, H.; Yamamura, Y.; Minoshima, S.; Yokochi, M.; Mizuno, Y.; Shimizu, N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998, 392, 605–608.
  26. Sasaki, S.; Shirata, A.; Yamane, K.; Iwata, M. Parkin-positive autosomal recessive juvenile Parkinsonism with alpha-synuclein-positive inclusions. Neurology 2004, 63, 678–682.
  27. Klein, C.; Lohmann-Hedrich, K. Impact of recent genetic findings in Parkinson’s disease. Curr. Opin. Neurol. 2007, 20, 453–464.
  28. Klein, C.; Westenberger, A. Genetics of Parkinson’s Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a008888.
  29. Kilarski, L.L.; Pearson, J.P.; Newsway, V.; Majounie, E.; Knipe, M.D.; Misbahuddin, A.; Chinnery, P.F.; Burn, D.J.; Clarke, C.E.; Marion, M.H.; et al. Systematic review and UK-based study of PARK2 (parkin), PINK1, PARK7 (DJ-1) and LRRK2 in early-onset Parkinson’s disease. Mov. Disord. 2012, 27, 1522–1529.
  30. Roth, J.A.; Singleton, S.; Feng, J.; Garrick, M.; Paradkar, P.N. Parkin regulates metal transport via proteasomal degradation of the 1B isoforms of divalent metal transporter 1. J. Neurochem. 2010, 113, 454–464.
  31. Bonifati, V.; Rizzu, P.; Van Baren, M.J.; Schaap, O.; Breedveld, G.J.; Krieger, E.; Dekker, M.C.; Squitieri, F.; Ibanez, P.; Joosse, M.; et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003, 299, 256–259.
  32. Canet-Aviles, R.M.; Wilson, M.A.; Miller, D.W.; Ahmad, R.; McLendon, C.; Bandyopadhyay, S.; Baptista, M.J.; Ringe, D.; Petsko, G.A.; Cookson, M.R. The Parkinson’s disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization. Proc. Natl. Acad. Sci. USA 2004, 101, 9103–9108.
  33. Li, H.M.; Niki, T.; Taira, T.; Iguchi-Ariga, S.M.; Ariga, H. Association of DJ-1 with chaperones and enhanced association and colocalization with mitochondrial Hsp70 by oxidative stress. Free Radic. Res. 2005, 39, 1091–1099.
  34. Junn, E.; Jang, W.H.; Zhao, X.; Jeong, B.S.; Mouradian, M.M. Mitochondrial localization of DJ-1 leads to enhanced neuroprotection. J. Neurosci. Res. 2009, 87, 123–129.
  35. Taira, T.; Saito, Y.; Niki, T.; Iguchi-Ariga, S.M.; Takahashi, K.; Ariga, H. DJ-1 has a role in antioxidative stress to prevent cell death. EMBO Rep. 2004, 5, 213–218.
  36. Lee, E.; Yin, Z.; Sidoryk-Węgrzynowicz, M.; Jiang, H.; Aschner, M. 15-Deoxy-delta12,14-prostaglandin J2‚‚ modulates manganese-induced activation of the NF-kB, Nrf2, and PI3K pathways in astrocytes. Free Radic. Biol. Med. 2012, 52, 1067–1074.
  37. Chen, P.; DeWitt, M.R.; Bornhorst, J.; Soares, F.A.; Mukhopadhyay, S.; Bowman, A.B.; Aschner, M. Age- and manganese-dependent modulation of dopaminergic phenotypes in a C. elegans DJ-1 genetic model of Parkinson’s disease. Metallomics 2015, 7, 289–298.
  38. Ramirez, A.; Heimbach, A.; Gründemann, J.; Stiller, B.; Hampshire, D.; Cid, L.P.; Goebel, I.; Mubaidin, A.F.; Wriekat, A.L.; Roeper, J.; et al. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat. Genet. 2006, 38, 1184–1191.
  39. Di Fonzo, A.; Chien, H.F.; Socal, M.; Giraudo, S.; Tassorelli, C.; Iliceto, G.; Fabbrini, G.; Marconi, R.; Fincati, E.; Abbruzzese, G.; et al. ATP13A2 missense mutations in juvenile parkinsonism and young onset Parkinson disease. Neurology 2007, 68, 1557–1562.
  40. Usenovic, M.; Tresse, E.; Mazzulli, J.R.; Taylor, J.P.; Krainc, D. Deficiency of ATP13A2 leads to lysosomal dysfunction, alpha-synuclein accumulation, and neurotoxicity. J. Neurosci. 2012, 32, 4240–4246.
  41. Dehay, B.; Ramirez, A.; Martinez-Vicente, M.; Perier, C.; Canron, M.H.; Doudnikoff, E.; Vital, A.; Vila, M.; Klein, C.; Bezard, E. Loss of P-type ATPase ATP13A2/PARK9 function induces general lysosomal deficiency and leads to Parkinson disease neurodegeneration. Proc. Natl. Acad. Sci. USA 2012, 109, 9611–9616.
  42. Gitler, A.D.; Chesi, A.; Geddie, M.L.; Strathearn, K.E.; Hamamichi, S.; Hill, K.J.; Caldwell, K.A.; Caldwell, G.A.; Cooper, A.A.; Rochet, J.C.; et al. Alpha-synuclein is part of a diverse and highly conserved interaction network that includes PARK9 and manganese toxicity. Nat. Genet. 2009, 41, 308–315.
  43. Rentschler, G.; Covolo, L.; Haddad, A.A.; Lucchini, R.G.; Zoni, S.; Broberg, K. ATP13A2 (PARK9) polymorphisms influence the neurotoxic effects of manganese. Neurotoxicology 2012, 20, 697–702.
More
Information
Subjects: Toxicology
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: 260
Revisions: 2 times (View History)
Update Date: 10 Aug 2023
1000/1000
ScholarVision Creations