Proteomics analysis of mitochondrial dysfunction triggered by complex specific electron transport chain inhibitors reveals common pathways involving protein misfolding in an SH-SY5Y in vitro cell model

Proteomics analysis of mitochondrial dysfunction triggered by complex specific electron transport chain inhibitors reveals common pathways involving protein misfolding in an SH-SY5Y in vitro cell model

Mitochondrial dysfunction has been previously identified in neurodegenerative diseases such as Alzheimer disease, Huntington disease, and Parkinson disease. Chemical inhibition of the mitochondrial electron transport chain (ETC) was shown to trigger symptoms in animal models similar to those observed in human neurodegenerative diseases. In order to understand the effect of mitochondrial dysfunction on the proteome level, LC-MSE based bottom-up, label-free differential proteomics expression analysis was used to monitor protein level changes in SH-SY5Y neuroblastoma cells induced by ETC-specific inhibitors (MPTP, 3-NP, sodium azide, antimycin A, and oligomycin). A total of 379 proteins were identified across the sample set and 75 of them were found to be differentially expressed (>30% fold change). Complex-specific inhibition of the five ETS complexes were expected to result in the aberrant regulation of different molecular pathways, but the bioinformatics analysis of the LC-MSMS data showed that the differentially expressed proteins were mostly involved in similar metabolic processes. The findings suggest that the complex-specific alterations may not be directly linked to neurodegenerative pathways, but could be considered contributors. Moreover, the proteins that showed the highest protein expression difference (>60% fold change) are involved in pathways regarding protein-folding and response to unfolded proteins. The results indicate that protein misfolding pathways might have a central role in the genesis and progression of neurodegenerative diseases and that label-free LC-MSMS proteomics analysis is an invaluable approach for studying of molecular pathways in neurodegeneration.

___

  • Akbar M, Essa MM, Abdelmegeed MA, Choi Y, Mahmood L, Song BJ (2016). Mitochondrial dysfunction and cell death in neurodegenerative diseases through nitroxidative stress. Brain Res 1637: 34-55.
  • Anderson NL, Anderson NG (1998). Proteome and proteomics: new technologies, new concepts, and new words. Electrophoresis 19: 1853-1861.
  • Barrera G, Gentile F, Pizzimenti S, Canuto RA, Daga M, Arcaro A, Centrangolo GP, Lepore A, Ferretti C, Dianzani C et al. (2016). Mitochondrial dysfunction in cancer and neurodegenerative diseases: spotlight on fatty acid oxidation and lipoperoxidation products. Antioxidants 5: 7.
  • Beal MF (2005). Mitochondria take center stage in aging and neurodegeneration. Ann Neurol 58: 495-505.
  • Bennett MC, Mlady GW, Kwon YH, Rose GM (2002). Chronic in vivo sodium azide infusion induces selective and stable inhibition of cytochrome c oxidase. J Neurochem 66: 2606-2611.
  • Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, Tendi EA, Murri L, Rapoport SI, Solaini G (2002). Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer’s disease. Neurobiol Aging 23: 371-376.
  • Bostanci N, Heywood W, Mills K, Parkar M, Nibali L, Donos N (2010). Application of label-free absolute quantitative proteomics in human gingival crevicular fluid by LC/MSE (Gingival Exudatome). J Proteome Res 9: 2191-2199.
  • Bradford MM (1976). A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72: 248-254.
  • Brouillet E, Condé F, Beal MF, Hantraye P (1999). Replicating Huntington’s disease phenotype in experimental animals. Prog Neurobiol 59: 427-468.
  • Brouillet E, Hantraye P, Ferrante RJ, Dolan R, Leroy-Willig A, Kowall NW, Beal MF (1995). Chronic mitochondrial energy impairment produces selective striatal degeneration and abnormal choreiform movements in primates. P Natl Acad Sci-Biol 92: 7105-7109.
  • Brouillet E, Jacquard C, Bizat N, Blum D (2005). 3-Nitropropionic acid: a mitochondrial toxin to uncover physiopathological mechanisms underlying striatal degeneration in Huntington’s disease. J Neurochem 95: 1521-1540.
  • Browne SE, Beal MF (2004). The energetics of Huntington’s disease. Neurochem Res 29: 531-546.
  • Bubber P, Haroutunian V, Fisch G, Blass JP, Gibson GE (2005). Mitochondrial abnormalities in Alzheimer brain: mechanistic implications. Ann Neurol 57: 695-703.
  • Castellani R, Hirai K, Aliev G, Drew KL, Nunomura A, Takeda A, Cash AD, Obrenovich ME, Perry G, Smith MA (2002). Role of mitochondrial dysfunction in Alzheimer’s disease. J Neurosci Res 70: 357-360.
  • Cheng F, Blackburn K, Lin Y, Goshe MB, Williamson JD (2009). Absolute protein quantification by LC/MS(E) for global analysis of salicylic acid-induced plant protein secretion responses. J Proteome Res 8: 82-93.
  • Corrado M, Scorrano L, Campello S (2012). Mitochondrial dynamics in cancer and neurodegenerative and neuroinflammatory diseases. Int J Biochem Cell Biol 2012: 729290.
  • D’Aguanno S, D’Alessandro A, Pieroni L, Roveri A, Zaccarin M, Marzano V, De Canio M, Bernardini S, Federici G, Urbani A (2011). New insights into neuroblastoma cisplatin resistance: a comparative proteomic and meta-mining investigation. J Proteome Res 10: 416-428.
  • Fernandez-Medarde A, Porteros A, de las Rivas J, Nunex A, Fuster JJ, Santos E (2007). Laser microdissection and microarray analysis of the hippocampus of Ras-GFR1 knockout mice reveals gene expression changes affecting signal transduction pathways related to memory and listening. Neuroscience 146: 272-285.
  • Fribley A, Zhang K, Kaufman RJ (2009). Regulation of apoptosis by the unfolded protein response. Method Mol Cell Biol 559: 191- 204.
  • Galante YM, Wong SY, Hatefi Y (1979). Composition of complex V of the mitochondrial oxidative phosphorylation system. J Biol Chem 254: 12372-12378.
  • Gibson GE, Sheu KFR, Blass JP (1998). Abnormalities of mitochondrial enzymes in Alzheimer disease. J Neural Transm 105: 855-870.
  • Haçarız O, Sayers G, Baykal AT (2012). A proteomic approach to investigate the distribution and abundance of surface and internal Fasciola hepatica proteins during the chronic stage of natural liver fluke infection in cattle. J Proteome Res. 11: 3592- 604.
  • He F, Zhang S, Qian F, Zhang C (1995). Delayed dystonia with striatal CT lucencies induced by a mycotoxin (3-nitropropionic acid). Neurology 45: 2178-2183.
  • Honda S, Hirose S (2003). Stage-specific enhanced expression of mitochondrial fusion and fission factors during spermatogenesis in rat testis. Biochem Bioph Res Co 311: 424- 432.
  • Hosp F, Vossfeldt H, Heinig M, Vasiljevic D, Arumughan A, Wyler E, Genetic and Environmental Risk for Alzheimer’s Disease GERAD1 Consortium; Landthaler M, Hubner N, Wanker EE et al. (2015). Quantitative interaction proteomics of neurodegenerative disease proteins. Cell Rep 11: 1134-1146.
  • Jiang Y, Wang X (2012). Comparative mitochondrial proteomics: perspective in human diseases. J Hematol Oncol 5: 11.
  • Kish SJ, Bergeron C, Rajput A, Dozic S, Mastrogiacomo F, Chang LJ, Wilson JM, DiStefano LM, Nobrega JN (1992). Brain cytochrome oxidase in Alzheimer’s disease. J Neurochem 59: 776-779.
  • Lin MT, Beal MF (2006). Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443: 787-795. Ludolph AC, He F, Spencer PS, Hammerstad J, Sabri M (1991). 3-Nitropropionic acid-exogenous animal neurotoxin and possible human striatal toxin. Canadian J Neurol Sci 18: 492- 498.
  • Ma X, Jin M, Cai Y, Xia H, Long K, Liu J, Yu Q, Yuan J (2011). Mitochondrial electron transport chain complex III is required for antimycin A to inhibit autophagy. Chem Biol 18: 1474- 1481.
  • Maeda S, Sasaki K, Halder SK, Fujita W, Ueda H (2016). Neuroprotective DAMPs member prothymosin alpha has additional beneficial actions against cerebral ischemia-induced vascular damages. J Pharmacol Sci 132: 100-104.
  • Manczak M, Park BS, Jung Y, Reddy PH (2004). Differential expression of oxidative phosphorylation genes in patients with Alzheimer’s disease: implications for early mitochondrial dysfunction and oxidative damage. NeuroMolecular Med 5: 147-162.
  • McDonald TG, Van Eyk JE (2003). Mitochondrial proteomics. Undercover in the lipid bilayer. Basic Res Cardiol 98: 219-227.
  • Morais VA, De Strooper B (2010). Mitochondria dysfunction and neurodegenerative disorders: cause or consequence. J Alzheimers Dis 2: 255-263.
  • Morais VA, Haddad D, Craessaerts K, De Bock PJ, Swerts J, Vilain S, Aerts L, Overbergh L, Grünewald A, Seibler P et al. (2014). PINK1 loss-of-function mutations affects mitochondrial complex I activity via NdufA10 ubiquinone uncoupling. Science 344: 203-207.
  • Neilson KA, Ali NA, Muralidharan S, Mirzaei M, Mariani M, Assadourian G, Lee A, van Sluyter SC, Haynes PA (2011). Less label, more free: approaches in label-free quantitative mass spectrometry. Proteomics 11: 535-553.
  • Oakes SA, Korsmeyer SJ (2004). Untangling the web: mitochondrial fission and apoptosis. Developmental Cell 7: 460-462.
  • Parlato R, Bierhoff H (2015). Role of nucleolar dysfunction in neurodegenerative disorders: a game of genes? AIMS Mol Sci 2: 211-224.
  • Parone PA, Cruz SD, Han JS, McAlois-Downes M, Vetto AP, Lee SK, Tseng E, Cleveland DW (2013). Enhancing mitochondrial calcium buffering capacity reduces aggregation of misfolded sod1 and motor neuron cell death without extending survival in mouse models of inherited amyotrophic lateral sclerosis. J Neurosci 11: 4657-4671.
  • Pfister JA, D’Mello SR (2016). Regulation of neuronal survival by nucleophosmin 1 (NPM1) is dependent on its expression level, subcellular localization and oligomerization status. J Biol Chem 291: 2078-2097.
  • Polyzos AA, McMurray CT (2017). The chicken or the egg: mitochondrial dysfunction as a cause or consequence of toxicity in Huntington’s disease. Mech Ageing Dev 161: 181- 197.
  • Ramsay RR, Salach JI, Singer TP (1986). Uptake of the neurotoxin 1-methyl-4-phenylpyridine (MPP+) by mitochondria and its relation to the inhibition of the mitochondrial oxidation of NAD+-linked substrates by MPP+. Biochem Biophys Res Commun 134: 743-748.
  • Reddy PH, Reddy TP, Manczak M, Calkins MJ, Shirendeb U, Mao P (2011). Dynamin-related protein 1 and mitochondrial fragmentation in neurodegenerative diseases. Brain Res Rev 67: 103-118.
  • Ross C, Poirier M (2004). Protein aggregation and neurodegenerative disease. Nat Med 10 Suppl, S10-7.
  • Schapira AH, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD (1990). Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem 54: 823-827.
  • Schon EA, Manfredi G (2003). Neuronal degeneration and mitochondrial dysfunction. J Clin Invest 111: 303-312.
  • Shaw JM, Nunnari J (2002). Mitochondrial dynamics and division in budding yeast. Trends Cell Biol 12: 178-184.
  • Silva JC (2005). Absolute quantification of proteins by LCMSE: a virtue of parallel ms acquisition. Mol Cell Proteomics 5: 144- 156.
  • Smalley JL, Breda C, Mason RP, Kooner G, Luthi-Carter R, Gant TW, Giorgini F (2016). Connectivity mapping uncovers small molecules that modulate neurodegeneration in Huntington’s disease models. J Mol Med 94: 235-245.
  • Swerdlow RH, Burns JM, Khan SM (2010). The Alzheimer’s disease mitochondrial cascade hypothesis. J Alzheimers Dis 2: 265- 279.
  • Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP et al. (2015). STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43: 447-452.
  • Wang X, Su B, Liu W, He X, Gao Y, Castellani RJ, Perry G, Smith MA, Zhu X (2011). DLP1-dependent mitochondrial fragmentation mediates 1-methyl-4-phenylpyridinium toxicity in neurons: implications for Parkinson’s disease. Aging Cell 10: 807-823.
  • Witte ME, Geurts JJG, de Vries HE, van der Valk P, van Horssen J (2010). Mitochondrial dysfunction: a potential link between neuroinflammation and neurodegeneration? Mitochondrion 10: 411-418.
  • Xie H, Hu L, Li G (2010). SH-SY5Y human neuroblastoma cell line: in vitro cell model of dopaminergic neurons in Parkinson’s disease. The Chinese Medical Journal (Engl) 123: 1086-1092.
  • Yin F, Boveris A, Cadenas E (2012). Mitochondrial energy metabolism and redox signaling in brain aging and neurodegeneration. Antioxid Redox Signal 20: 353-371.
  • Yoshino H, Nakagawa-Hattori Y, Kondo T, Mizuno Y (1992). Mitochondrial complex I and II activities of lymphocytes and platelets in Parkinson’s disease. J Neural Transm Park Dis Dement Sect 4: 27-34.
  • Zhang J, Keene CD, Pan C, Montine KS, Montine TJ (2008). Proteomics of human neurodegenerative diseases. J Neuropathol Exp Neurol 67: 923-932.
  • Zhang L, Li L, Ban L, An W, Liu S, Li X, Xue B, Xu Y (2000). Effect of sodium azide on mitochondrial membrane potential in SHSY5Y human neuroblastoma cells. Acta Academiae Medicinae Sinicae 22: 436-439.