Mammalian Expression and Biophysical Examination of Human Wild Type Optineurin Protein

  • Hongyu Ying University of Illinois at Chicago Department of Ophthalmology and Visual Sciences
  • Xiang Shen University of Illinois at Chicago Department of Ophthalmology and Visual Sciences
  • Minhua Wang University of Illinois at Chicago Department of Ophthalmology and Visual Sciences
  • Beatrice Y.J.T. Yue University of Illinois at Chicago Department of Ophthalmology & Visual Sciences
Keywords: Human optineurin protein, Mammalian expression, Tet-on inducible cell line, Posttranslational modification sites, Circular dichroism, Bimolecular fluorescence complementation (BiFc), Thioflavin T (ThT) assay.

Abstract

Background: The optineurin gene has been linked to normal tension glaucoma and amyotrophic lateral sclerosis. Optineurin protein, known to interact with a number of proteins including Rab8, myosin VI, and huntingtin, plays a role in vesicle trafficking, Golgi organization, antibacterial and antiviral signaling, autophagic clearance of protein aggregates, and regulation of gene expression. Its basic biophysical properties however have never been explored. The purpose of the study was to obtain sufficient amounts of purified mammalian optineurin protein and to carry out biophysical characterization.

Results: Tetracycline inducible RGC-5 cell line that expresses Halo tagged human wild type optineurin was created. Tag free optineurin was purified and its purity was assessed by SDS-PAGE and Western blotting. The secondary structure of the highly purified optineurin was examined by circular dichroism (CD) spectropolarimeter. The posttranslational modification sites were identified by liquid chromatography-mass spectrometry. Protein-protein interaction was evaluated by bimolecular fluorescence complementation (BiFc) analysis. The aggregation of optineurin was studied by thioflavin T (ThT) assay. The CD spectra indicated that the tag free optineurin protein was folded, containing both ?-helical and ?-sheet secondary structures. One reported phosphorylation site serine (Ser)177, four novel phosphorylation sites (Ser173, 174, 526 and 528), as well as one new acetylation site (Ser2) were identified. Optineurin protein was also shown to be an aggregate prone protein. It interacted with itself as detected by BiFc assay, and formed ?-sheet rich structures or aggregates as evidenced by ThT assay.

Conclusions: The inducible cell lines would allow production of highly purified protein in a scaled-up and cost effective manner. Fundamental biophysical information regarding optineurin including its secondary structure, posttranslational modification sites, and aggregation was attained.

Downloads

Download data is not yet available.

Author Biography

Beatrice Y.J.T. Yue, University of Illinois at Chicago Department of Ophthalmology & Visual Sciences
Emeritus Professor, Department of Ophthalmology and Visual Sciences

References

Li Y, Kang J, Horwitz MS. Interaction of an adenovirus E3 14.7-kilodalton protein with a novel tumor necrosis factor ?-inducible cellular protein containing leucine zipper domains. Mol Cell Biol. 1998;18:1601-1610.

Schwamborn K, Weil R, Courtois G, Whiteside ST, Israel A. Phorbol esters and cytokines regulate the expression of the NEMO-related protein, a molecule involved in a NF-?B-independent pathway. J Biol Chem. 2000;275:22780-22789.

Rezaie T, Child A, Hitchings R, Brice G, Miller L, Coca-Prados M, et al. Adult-onset primary open-angle glaucoma caused by mutations in optineurin. Science. 2002;295:1077-1079.

Maruyama H, Morino H, Ito H, Izumi Y, Kato H, Watanabe Y, et al. Mutations of optineurin in amyotrophic lateral sclerosis. Nature. 2010;465:223-226.

Maruyama H, Kawakami H. Optineurin and amyotrophic lateral sclerosis. Geriatr Gerontol Int. 2013;13:528-532.

Kamada M, Izumi Y, Ayaki T, Nakamura M, Kagawa S, Kudo E, et al. Clinicopathologic features of autosomal recessive amyotrophic lateral sclerosis associated with optineurin mutation. Neuropathology. 2014;34:64-70.

Rezaie T, Waitzman DM, Seeman JL, Kaufman PL, Sarfarazi M. Molecular cloning and expression profiling of optineurin in the rhesus monkey. Invest Ophthalmol Vis Sci. 2005;46:2404-2410.

Wild P, Farhan H, McEwan DG, Wagner S, Rogov VV, Brady NR, et al. Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth. Science. 2011;333:228-233.

Hattula K, Peranen J. FIP-2, a coiled-coil protein, links Huntingtin to Rab8 and modulates cellular morphogenesis. Curr Biol. 2000;10:1603-1606.

Stroissnigg H, Repitz M, Miloloza A, Linhartova I, Beug H, Wiche G, Propst F. FIP-2, an I?B-kinase-?-related protein, is associated with the Golgi apparatus and translocates to the marginal band during chicken erythroblast differentiation. Exp Cell Res. 2002;278:133-145.

Ying H, Shen X,Yue BYJT. Establishment of inducible wild type and mutant myocilin-GFP-expressing RGC-5 cell lines. PLoS One. 2012;7:e47307.

Ying H, Shen X, Park B,Yue BYJT. Posttranslational modifications, localization, and protein interactions of optineurin, the product of a glaucoma gene. PLoS One. 2010;5:e9168.

Gao J, Ohtsubo M, Hotta Y, Minoshima S. Oligomerization of optineurin and its oxidative stress- or E50K mutation-driven covalent cross-linking: possible relationship with glaucoma pathology. PLoS One. 2014;9:e101206.

De Marco N, Buono M, Troise F, Diez-Roux G. Optineurin increases cell survival and translocates to the nucleus in a Rab8-dependent manner upon an apoptotic stimulus. J Biol Chem. 2006;281:16147-16156.

Chibalina MV, Roberts RC, Arden SD, Kendrick-Jones J, Buss F. Rab8-optineurin-myosin VI: analysis of interactions and functions in the secretory pathway. Methods Enzymol. 2008;438:11-24.

del Toro D, Alberch J, Lazaro-Dieguez F, Martin-Ibanez R, Xifro X, Egea G, et al. Mutant huntingtin impairs post-Golgi trafficking to lysosomes by delocalizing optineurin/Rab8 complex from the Golgi apparatus. Mol Biol Cell. 2009;20:1478-1492.

Sahlender DA, Roberts RC, Arden SD, Spudich G, Taylor MJ, Luzio JP, et al. Optineurin links myosin VI to the Golgi complex and is involved in Golgi organization and exocytosis. J Cell Biol. 2005;169:285-295.

Park B, Ying H, Shen X, Park JS, Qiu Y, Shyam R,Yue BYJT. Impairment of protein trafficking upon overexpression and mutation of optineurin. PLoS One. 2010;5:e11547.

Nagabhushana A, Chalasani ML, Jain N, Radha V, Rangaraj N, Balasubramanian D, et al. Regulation of endocytic trafficking of transferrin receptor by optineurin and its impairment by a glaucoma-associated mutant. BMC Cell Biol. 2010;11:4.

Anborgh PH, Godin C, Pampillo M, Dhami GK, Dale LB, Cregan SP, et al. Inhibition of metabotropic glutamate receptor signaling by the huntingtin-binding protein optineurin. J Biol Chem. 2005;280:34840-34848.

Kachaner D, Filipe J, Laplantine E, Bauch A, Bennett KL, Superti-Furga G, et al. Plk1-dependent phosphorylation of optineurin provides a negative feedback mechanism for mitotic progression. Mol Cell. 2012;45:553-566.

Morton S, Hesson L, Peggie M, Cohen P. Enhanced binding of TBK1 by an optineurin mutant that causes a familial form of primary open angle glaucoma. FEBS Lett. 2008;582:997-1002.

Bond LM, Peden AA, Kendrick-Jones J, Sellers JR, Buss F. Myosin VI and its binding partner optineurin are involved in secretory vesicle fusion at the plasma membrane. Mol Biol Cell. 2011;22:54-65.

Park BC, Shen X, Samaraweera M, Yue BYJT. Studies of optineurin, a glaucoma gene: Golgi fragmentation and cell death from overexpression of wild-type and mutant optineurin in two ocular cell types. Am J Pathol. 2006;169:1976-1989.

Sippl C, Zeilbeck LF, Fuchshofer R, Tamm ER. Optineurin associates with the podocyte Golgi complex to maintain its structure. Cell Tissue Res. 2014;358:567-583.

Zhu G, Wu CJ, Zhao Y, Ashwell JD. Optineurin negatively regulates TNF?- induced NF-?B activation by competing with NEMO for ubiquitinated RIP. Curr Biol. 2007;17:1438-1443.

Sudhakar C, Nagabhushana A, Jain N, Swarup G. NF-?B mediates tumor necrosis factor ?-induced expression of optineurin, a negative regulator of NF-?B. PLoS One. 2009;4:e5114.

Mankouri J, Fragkoudis R, Richards KH, Wetherill LF, Harris M, Kohl A, et al. Optineurin negatively regulates the induction of IFN? in response to RNA virus infection. PLoS Pathog. 2010;6:e1000778.

Nagabhushana A, Bansal M, Swarup G. Optineurin is required for CYLD-dependent inhibition of TNF?-induced NF-?B activation. PLoS One. 2011;6:e17477.

Akizuki M, Yamashita H, Uemura K, Maruyama H, Kawakami H, Ito H, et al. Optineurin suppression causes neuronal cell death via NF-?B pathway. J Neurochem. 2013;126:699-704.

Shen X, Ying H, Qiu Y, Park JS, Shyam R, Chi ZL, et al. Processing

of optineurin in neuronal cells. J Biol Chem. 2011;286:3618-3629.

Chalasani ML, Kumari A, Radha V, Swarup G. E50K-OPTN-induced retinal cell death involves the Rab GTPase-activating protein, TBC1D17 mediated block in autophagy. PLoS One. 2014;9:e95758.

Ying H, Yue BYJT. Optineurin: The autophagy connection. Exp Eye Res. 2015;epub ahead of print.

Rogov VV, Suzuki H, Fiskin E, Wild P, Kniss A, Rozenknop A, et al. Structural basis for phosphorylation-triggered autophagic clearance of Salmonella. Biochem J. 2013;454:459-466.

Kerppola TK. Bimolecular fluorescence complementation (BiFC) analysis of protein interactions in live cells. Cold Spring Harb Protoc. 2013:727-731.

Outeiro TF, Putcha P, Tetzlaff JE, Spoelgen R, Koker M, et al. Formation of toxic oligomeric ?-synuclein species in living cells. PLoS One. 2008;3:e1867.

Ying H, Yue BYJT. Cellular and molecular biology of optineurin. Int Rev Cell Mol Biol. 2012;294:223-258.

Hu CD, Chinenov Y, Kerppola TK. Visualization of interactions among bZIP and Rel family proteins in living cells using bimolecular fluorescence complementation. Mol Cell. 2002;9:789-798.

Whitmore L, Wallace BA. DICHROWEB, an online server for protein secondary structure analyses from circular dichroism spectroscopic data. Nucleic Acids Res. 2004;2:W668-W673.

Whitmore L, Wallace BA. Protein secondary structure analyses from circular dichroism spectroscopy: Methods and reference databases. Biopolymers. 2008;89:392-400.

Osawa T, Mizuno Y, Fujita Y, Takatama M, Nakazato Y, Okamoto K. Optineurin in neurodegenerative diseases. Neuropathology. 2011;31:569-574.

Schwab C, Yu S, McGeer EG, McGeer PL. Optineurin in Huntington's disease intranuclear inclusions. Neurosci Lett. 2012;506:149-154.

Fuse N, Takahashi K, Akiyama H, Nakazawa T, Seimiya M, Kuwahara S, et al. Molecular genetic analysis of optineurin gene for primary open-angle and normal tension glaucoma in the Japanese population. J Glaucoma. 2004;13:299-303.

Liu YH, Tian T. Hypothesis of optineurin as a new common risk factor in normal-tension glaucoma and Alzheimer's disease. Med Hypotheses. 2011;77:591-592.

Ito H, Nakamura M, Komure O, Ayaki T, Wate R, Maruyama H, et al. Clinicopathologic study on an ALS family with a heterozygous E478G optineurin mutation. Acta Neuropathol. 2011;122:223-229.

Ito H, Fujita K, Nakamura M, Wate R, Kaneko S, Sasaki S, et al. Optineurin is co-localized with FUS in basophilic inclusions of ALS with FUS mutation and in basophilic inclusion body disease. Acta Neuropathol. 2011;121:555-557.

Van Bergen NJ, Wood JP, Chidlow G, Trounce IA, Casson RJ, Ju WK, et al. Recharacterization of the RGC-5 retinal ganglion cell line. Invest Ophthalmol Vis Sci. 2009;50:4267-4272.

Krishnamoorthy RR, Clark AF, Daudt D, Vishwanatha JK, Yorio T. A forensic path to RGC-5 cell line identification: lessons learned. Invest Ophthalmol Vis Sci. 2013;54:5712-5719.

Sippl C, Tamm ER. What is the nature of the RGC-5 cell line? Adv Exp Med Biol. 2014;801:145-154.

Ohana RF, Hurst R, Vidugiriene J, Slater MR, Wood KV, Urh M. HaloTag-based purification of functional human kinases from mammalian cells. Protein Expr Purif. 2011;76:154-164.

Archambault V, Glover DM. Polo-like kinases: conservation and divergence in their functions and regulation. Nat Rev Mol Cell Biol. 2009;10:265-275.

Helgason E, Phung QT, Dueber EC. Recent insights into the complexity of Tank-binding kinase 1 signaling networks: the emerging role of cellular localization in the activation and substrate specificity of TBK1. FEBS Lett. 2013;587:1230-1237.

Korac J, Schaeffer V, Kovacevic I, Clement AM, Jungblut B, Behl C, et al. Ubiquitin-independent function of optineurin in autophagic clearance of protein aggregates. J Cell Sci. 2013;126:580-592.

Starheim KK, Gevaert K, Arnesen T. Protein N-terminal acetyltransferases: when the start matters. Trends Biochem Sci. 2012;37:152-161.

LeVine H 3rd. Quantification of ?-sheet amyloid fibril structures with thioflavin T. Methods Enzymol. 1999;309:274-284.

Chhangani D, Mishra A. Protein quality control system in neurodegeneration: a healing company hard to beat but failure is fatal. Mol Neurobiol. 2013;48:141-156.

Johnston JA, Illing ME, Kopito RR. Cytoplasmic dynein/dynactin mediates the assembly of aggresomes. Cell Motil Cytoskeleton. 2002;53:26-38.

Koga T, Shen X, Park JS, Qiu Y, Park BC, Shyam R, et al. Differential effects of myocilin and optineurin, two glaucoma genes, on neurite outgrowth. Am J Pathol. 2010;176:343-352.

Ying H, Turturro S, Nguyen T, Shen X, Zelkha R, Johnson EC, et al. Induction of autophagy in rats upon overexpression of wild-type and mutant optineurin gene. BMC Cell Biol. 2015;16:14.

Aung T, Rezaie T, Okada K, Viswanathan AC, Child AH, Brice G, et al. Clinical features and course of patients with glaucoma with the E50K mutation in the optineurin gene. Invest Ophthalmol Vis Sci. 2005;46:2816-2822.

Published
2015-10-25
How to Cite
Ying, H., Shen, X., Wang, M., & Yue, B. (2015). Mammalian Expression and Biophysical Examination of Human Wild Type Optineurin Protein. Journal of Progressive Research in Biology, 2(2), 66-77. Retrieved from http://scitecresearch.com/journals/index.php/jprb/article/view/426
Section
Articles