Enhanced transient expression of an anti-CD52 monoclonal antibody in CHO cells through utilization of miRNA sponge technology

Morvarid Sadat Pairawan , Azam Bolhassani, Azam Rahimpour

Abstract


Chinese hamster ovary (CHO) cells are the dominant mammalian host system for the production of recombinant therapeutic proteins. Improving the viable cell density during culture of recombinant CHO cells can greatly affect the production yield. MicroRNAs (miRs) -15a and 16-1 are known as negative regulators of multiple genes involved in cell cycle progression and apoptotic inhibition. miR sponges, which act  as decoy targets, are transcripts which contain complementary binding sites to the seed region of  related miRs. Stably expressed miR sponges are known as efficient tools for miR loss of function studies.In this study, stable CHO cell pools and clones expressing miRs-15a and 16-1 specific decoy transcript downstream of an enhanced green fluorescent protein reporter gene was developed. Analysis of cell growth during 12 days of batch culture indicated improved maximum viable cell density of CHO cells and clones expressing the decoy transcript. In addition, transient expression of a recombinant anti-CD52 monoclonal antibody was significantly improved in a decoy harboring CHO cell clone, representing a 3.37-fold increase in yield after 4 days of culture. Our results indicated that miR sponge technology can be successfully applied for the improvement of cell viability and transient monoclonal antibody expression in CHO host cells.


Keywords


Chinese hamster ovary cells; MicroRNA sponge; Decoy; miR-15a; miR-16-1; Monoclonal antibody.

Full Text:

PDF

References


Lalonde ME, Durocher Y. Therapeutic glycoprotein production in mammalian cells. J Biotechnol. 2017;251:128-140.

Bayat H, Hossienzadeh S, Pourmaleki E, Ahani R, Rahimpour A. Evaluation of different vector design strategies for the expression of recombinant monoclonal antibody in CHO cells. Prep Biochem Biotechnol. 2018;48(2):160-164.

Wurm FM. Production of recombinant protein therapeutics in cultivated mammalian cells. Nat Biotechnol. 2004;22(11):1393-1398.

Han S, Rhee WJ. Inhibition of apoptosis using exosomes in Chinese hamster ovary cell culture. Biotechnol Bioeng. 2018;115(5):1331-1339.

Rangan S, Kamal S, Konorov SO, Schulze HG, Blades MW, Turner RFB, et al. Types of cell death and apoptotic stages in Chinese Hamster Ovary cells distinguished by Raman spectroscopy. Biotechnol Bioeng. 2018;115(2):401-412.

Kim JY, Kim Y-G, Lee GM. CHO cells in biotechnology for production of recombinant proteins: current state and further potential. Appl Microbiol Biotechnol. 2012;93(3):917-930.

Majors BS, Betenbaugh MJ, Pederson NE, Chiang GG. Mcl‐1 overexpression leads to higher viabilities and increased production of humanized monoclonal antibody in Chinese hamster ovary cells. Biotechnol Prog. 2009;25(4):1161-1168.

Zustiak MP, Jose L, Xie Y, Zhu J, Betenbaugh MJ. Enhanced transient recombinant protein production in CHO cells through the co-transfection of the product gene with Bcl-xL. Biotechnol J. 2014;9(9):1164-1174.

Jeon MK, Yu DY, Lee GM. Combinatorial engineering of ldh-a and bcl-2 for reducing lactate production and improving cell growth in dihydrofolate reductase-deficient Chinese hamster ovary cells. Appl Microbiol Biotechnol. 2011;92(4):779-790.

Meents H, Enenkel B, Eppenberger HM, Werner RG, Fussenegger M. Impact of coexpression and coamplification of sICAM and antiapoptosis determinants bcl-2/bcl-x(L) on productivity, cell survival, and mitochondria number in CHO-DG44 grown in suspension and serum-free media. Biotechnol Bioeng. 2002;80(6):706-716.

Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215-233.

Barron N, Sanchez N, Kelly P, Clynes M. MicroRNAs: tiny targets for engineering CHO cell phenotypes? Biotechnol Lett. 2011;33(1):11-21.

Jadhav V, Hackl M, Druz A, Shridhar S, Chung CY, Heffner KM, et al. CHO microRNA engineering is growing up: recent successes and future challenges. Biotechnol Adv. 2013;31(8):1501-1513.

Ebert MS, Sharp PA. MicroRNA sponges: progress and possibilities. RNA. 2010;16(11):2043-2050.

Liu Q, Fu H, Sun F, Zhang H, Tie Y, Zhu J, et al. miR-16 family induces cell cycle arrest by regulating multiple cell cycle genes. Nucleic Acids Res. 2008;36(16):5391-5404.

Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L, et al. The miR-15a-–miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med. 2008;14(11):1271-1277.

Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A. 2005;102(39):13944-13949.

Pekarsky Y, Croce CM. Role of miR-15/16 in CLL. Cell Death Differ. 2015;22(1):6-11.

Lian YF, Huang YL, Wang JL, Deng MH, Xia TL, Zeng MS, et al. Anillin is required for tumor growth and regulated by miR-15a/miR-16-1 in HBV-related hepatocellular carcinoma. Aging (Albany NY). 2018;10(8):1884-1901.

Havrdova E, Horakova D, Kovarova I. Alemtuzumab in the treatment of multiple sclerosis: key clinical trial results and considerations for use. Ther Adv Neurol Disord. 2015;8(1):31-45.

Rommer PS, Dudesek A, Stüve O, Zettl UK. Monoclonal antibodies in treatment of multiple sclerosis. Clin Exp Immunol. 2014;175(3):373-384.

Fischer S, Handrick R, Otte K. The art of CHO cell engineering: A comprehensive retrospect and future perspectives. Biotechnol Adv. 2015;33(8): 1878-1896.

Wells E, Robinson AS. Cellular engineering for therapeutic protein production: product quality, host modification, and process improvement. Biotechnol J. 2017;12(1).: DOI: 10.1002/biot.201600105. 1600105.

Grav LM, Lee JS, Gerling S, Kallehauge TB, Hansen AH, Kol S, et al. One-step generation of triple knockout CHO cell lines using CRISPR/Cas9 and fluorescent enrichment. Biotechnol J. 2015;10(9):1446-1456.

Druz A, Chu C, Majors B, Santuary R, Betenbaugh M, Shiloach J. A novel microRNA mmu‐miR‐466h affects apoptosis regulation in mammalian cells. Biotechnol Bioeng. 2011;108(7):1651-1661.

Fischer S, Buck T, Wagner A, Ehrhart C, Giancaterino J, Mang S, et al. A functional high-content miRNA screen identifies miR-30 family to boost recombinant protein production in CHO cells. Biotechnol J. 2014;9(10):1279-1292.

Druz A, Son YJ, Betenbaugh M, Shiloach J. Stable inhibition of mmu-miR-466h-5p improves apoptosis resistance and protein production in CHO cells. Metab eng. 2013;16:87-94.

Diniz MG, Gomes CC, de Castro WH, Guimarães ALS, De Paula AMB, Amm H, et al. miR-15a/16-1 influences BCL2 expression in keratocystic odontogenic tumors. Cell Oncol (Dordr). 2012;35(4):285-291.

Tey BT, Singh RP, Piredda L, Piacentini M, Al-Rubeai M. Influence of bcl-2 on cell death during the cultivation of a Chinese hamster ovary cell line expressing a chimeric antibody. Biotechnol Bioeng. 2000;68(1):31-43.

Sanchez N, Kelly P, Gallagher C, Lao NT, Clarke C, Clynes M, et al. CHO cell culture longevity and recombinant protein yield are enhanced by depletion of miR-7 activity via sponge decoy vectors. Biotechnol J. 2014;9(3):396-404.

Costello A, Coleman O, Lao NT, Henry M, Meleady P, Barron N, et al. Depletion of endogenous miRNA-378-3p increases peak cell density of CHO DP12 cells and is correlated with elevated levels of ubiquitin carboxyl-terminal hydrolase 14. J Biotechnol. 2018;288:30-40.

Kelly PS, Breen L, Gallagher C, Kelly S, Henry M, Lao NT, et al. Re-programming CHO cell metabolism using miR-23 tips the balance towards a highly productive phenotype. Biotechnol J. 2015;10(7): 1029-1040.


Refbacks

  • There are currently no refbacks.


Creative Commons LicenseThis work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License which allows users to read, copy, distribute and make derivative works for non-commercial purposes from the material, as long as the author of the original work is cited properly.