31.01.2014 Views

Download PDF (all abstracts) - BioMed Central

Download PDF (all abstracts) - BioMed Central

Download PDF (all abstracts) - BioMed Central

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

BMC Proceedings 2013, Volume 7 Suppl 6<br />

http://www.biomedcentral.com/bmcproc/supplements/7/S6<br />

Page 8 of 151<br />

Results: A miniaturized circulation system has been established over a<br />

period of 14 days by fully covering <strong>all</strong> channels and surfaces of the MOC<br />

with human microvascular endothelial cells. By injecting 2 × 10 7 cells ml -1<br />

into the channels, a homogeneous distribution of cells throughout <strong>all</strong><br />

channels was achieved (Figure 1b). During the following static incubation,<br />

cells adhered well to the air plasma treated channel w<strong>all</strong>s. A peristaltic<br />

micro-pump was used to create culture medium circulation. After adaption<br />

to shear stress, HDMEC showed an elongation and alignment par<strong>all</strong>el to<br />

the flow direction. Three-dimensional reconstitutions of image stacks<br />

indicate that cells formed confluent monolayers on <strong>all</strong> w<strong>all</strong>s of the channels<br />

(Figure 1c). During the whole cultivation time they maintained adherence<br />

to the channel w<strong>all</strong>s and were positive for Calcein AM viability staining<br />

(Figure 1b). After 14 days of culture HDMEC forming the microvascular circuit<br />

were positive for ac-LDL uptake (Figure 1d) and expressed the endothelialspecific<br />

marker CD31 and von Willebrand Factor (vWF) (Figure 1e).<br />

Conclusion: A robust procedure applying pulsatile shear stress has been<br />

established to cover <strong>all</strong> fluid contact surfaces of the system with a functional,<br />

tightly closed layer of HDMEC.<br />

Long-term cultivation of elongated and flow-aligned HDMEC inside the chipbased<br />

microcirculation was demonstrated over a period of 14 days. For such<br />

endothelialized microfluidic devices to be useful for substance testing, it is<br />

essential to show long-term viability and function in the presence of<br />

physiological flow rates as shown here. These artificial vessels are an<br />

important approach for systemic substance testing in Multi-Organ-Chips.<br />

The miniaturized circulation system creates the conditions for circulation of<br />

nutrients through the organoid culture chamber, <strong>all</strong>ows for in vivo-like<br />

crosstalk between endothelial cells and tissues and prevents clumping<br />

inside the channels. Compared with conventional cell culture techniques, a<br />

microfluidic-based cell culture may mimic more accurate in vivo-like<br />

extracellular conditions, as the culture of cells and organ models in perfused<br />

microfluidic systems can improve their oxygen and nutrient supply. This<br />

makes it suitable for long-term cultivation and more efficient drug studies.<br />

In future, such endothelialized bioreactors might be used for testing<br />

vasoactive substances. Fin<strong>all</strong>y, the described system can now be used for<br />

the establishment of organ-specific capillary networks. Here, we will adhere<br />

to our recently published roadmap toward vascularized ‘’human-on-a-chip’’<br />

models to generate systemic data fully replacing the animals or human<br />

beings currently used [2].<br />

Acknowledgements: The work has been funded by the German Federal<br />

Ministry for Education and Research, GO-Bio Grant No. 0315569.<br />

References<br />

1. Wu M-H, Huang S-B, Cui Z, Cui Z, Lee G-B: A high throughput perfusionbased<br />

microbioreactor platform integrated with pneumatic micropumps<br />

for three-dimensional cell culture. Biomedical microdevices 2008,<br />

10:309-319.<br />

2. Marx U, W<strong>all</strong>es H, Hoffmann S, Lindner G, Horland R, Sonntag F,<br />

Klotzbach U, Sakharov D, Tonevitsky A, Lauster R: “Human-on-a-chip”<br />

developments: a translational cutting-edge alternative to systemic<br />

safety assessment and efficiency evaluation of substances in laboratory<br />

animals and man? Alternatives to laboratory animals: ATLA 2012,<br />

40:235-257.<br />

O7<br />

Rapid construction of transgene-amplified CHO cell lines by cell cycle<br />

checkpoint engineering<br />

Kyoungho Lee 1 , Kohsuke Honda 1 , Hisao Ohtake 1 , Takeshi Omasa 1,2*<br />

1 Department of Biotechnology, Graduate School of Engineering, Osaka<br />

University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan;<br />

2 Institute of<br />

Technology and Science, The University of Tokushima, 2-1 Minamijosanjimacho,<br />

Tokushima 770-8506, Japan<br />

E-mail: omasa@bio.tokushima-u.ac.jp<br />

BMC Proceedings 2013, 7(Suppl 6):O7<br />

Introduction: Dihydrofolate reductase (DHFR)-mediated gene amplification<br />

has been widely used to establish high-producing mammalian cell lines<br />

[1-3]. However, since gene amplification is an infrequent event, in that many<br />

rounds of methotrexate (MTX) selection to amplify the transgene and<br />

screening of over several hundred individual clones are required to obtain<br />

cells with high gene copy numbers [4]. Consequently, the process for DHFRmediated<br />

gene amplification is a time-consuming and laborious step for cell<br />

line construction. Here, we present a novel concept to accelerate gene<br />

amplification through cell cycle checkpoint engineering. In our knowledge,<br />

there is no previous report which focused on controlling cell cycle<br />

checkpoint to enhance the efficiency of DHFR gene amplification system.<br />

Materials and methods: A sm<strong>all</strong> interfering RNA (siRNA) expression<br />

vector against Ataxia-Telangiectasia and Rad3-Related (ATR), a cell cycle<br />

checkpoint kinase, was transfected into Chinese hamster ovary (CHO) cells.<br />

The effects of ATR down-regulation on gene amplification and productivity<br />

in CHO cells producing green fluorescent protein (GFP) and monoclonal<br />

antibody (mAb) were investigated.<br />

Results and discussion: Analysis of GFP expression level during gene<br />

amplification process: The ratio of GFP-expressing cells was evaluated<br />

by flow cytometry analysis during the gene amplification process at 100-,<br />

250-, and 500-nM MTX concentrations. In the process of gene amplification<br />

at <strong>all</strong> MTX concentrations, the pools of ATR-downregulated cells showed a<br />

much higher percentage of GFP-positive cells as compared with the pools<br />

of mock cells. At 100-nM MTX concentration, the percentage of GFPpositive<br />

cells in the CHO-siATR cell pool was 18.7% of total cells, which<br />

was approximately twice of the 8.4% in the mock cells. At 250- and<br />

500-nM MTX concentrations, CHO-siATR cell pools had 28.6 and 39.2%<br />

GFP-positive cells, respectively, which were up to six times higher than the<br />

4.6 and 6.8% of the pools of mock cells.<br />

Comparison of IgG productivity: IgG-producing cell lines were generated<br />

to confirm the previous results obtained in GFP-producing cell lines. The<br />

ATR-downregulated cells showed a significant increase in specific production<br />

rate of an average of 0.08 pg cell −1 day −1 , which was approximately four<br />

times higher than the average of 0.02 pg cell −1 day −1 in the mock cells.<br />

The volumetric productivity of each cell line was also investigated to<br />

evaluate the influence of ATR downregulation. The volumetric productivity<br />

of ATR knockdown cells was an average of 0.035 mg L −1 day −1 , which was<br />

approximately three times higher than the average of 0.013 mg L −1 day −1<br />

of the mock cells, suggesting that ATR knockdown generated the pool of<br />

higher-producing cells during the gene amplification process.<br />

Estimation of amplified transgene copy number: Quantitative real-time<br />

PCR was used to estimate the amplified transgene copy number of GFPproducing<br />

cell lines during the gene amplification process. The average<br />

copy number of ATR-downregulated cells was 15.4 ± 0.8, 27.6 ± 0.3,<br />

and 62.0 ± 2.9 at 100-, 250-, and 500-nM MTX concentrations, respectively.<br />

These numbers were up to 24 times higher than 3.98 ± 0.09, 2.20 ± 0.03,<br />

and 2.59 ± 0.07 of the mock cells. Interestingly, the amplified transgene<br />

copy numbers in the pools of ATR-downregulated cells were increased<br />

proportion<strong>all</strong>y with the MTX concentration. The amplified transgene copy<br />

numbers in the IgG-producing cells were also investigated during the gene<br />

amplification process at 100-nM MTX concentration. The amplified light- and<br />

heavy-chain copy numbers of the pool of ATR knockdown cells were 13.2 ±<br />

3.8 and 11.8 ± 1.8, respectively, which were up to seven times higher than<br />

6.95 ± 0.07 and 1.68 ± 0.04 of the mock cells. The results from both the<br />

GFP- and IgG-producing cells showed that the pools of ATR-downregulated<br />

cells had much higher amplified transgene copy numbers as compared with<br />

the pools of mock cells during the gene amplification process.<br />

Conclusions: In conclusion, we have demonstrated that gene amplification<br />

can be accelerated by the downregulation of a cell cycle checkpoint kinase,<br />

ATR, and a pool of high-producing cells can be rapidly derived in a short<br />

time after MTX treatment. This novel method focuses on generating more<br />

high-producing cells in a heterogeneous pool as compared with the<br />

conventional method and would thus contribute to reducing the time and<br />

labor required for cell line establishment by increasing the possibility of<br />

selecting high-producing clones.<br />

Acknowledgements: This work is parti<strong>all</strong>y supported by grants from the<br />

Program for the Promotion of Fundamental Studies in Health Sciences of<br />

NIBIO and a Grant-in-Aid for Scientific Research of JSPS. We thank Prof.<br />

Yoshikazu Kurosawa at Fujita Health University for kindly providing heavyand<br />

light-chain genes of humanized IgG.<br />

References<br />

1. Gandor C, Leist C, Fiechter A, Asselbergs FA: Amplification and expression<br />

of recombinant genes in serum-independent Chinese hamster ovary<br />

cells. FEBS Lett 1995, 377:290-294.<br />

2. Kim JY, Kim YG, Lee GM: CHO cells in biotechnology for production of<br />

recombinant proteins: current state and further potential. Appl Microbiol<br />

Biotechnol 2012, 93:917-930.<br />

3. Wurm FM: Production of recombinant protein therapeutics in cultivated<br />

mammalian cells. Nat Biotechnol 2004, 22:1393-1398.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!