4.6 Article

Enabling Large-Scale Ex Vivo Production of Megakaryocytes from CD34+ Cells Using Gas-Permeable Surfaces

期刊

STEM CELLS TRANSLATIONAL MEDICINE
卷 8, 期 7, 页码 658-670

出版社

WILEY
DOI: 10.1002/sctm.18-0160

关键词

Megakaryocyte; CD34(+); Thrombopoiesis; Cell culture; Cellular therapy

资金

  1. NIH [1R01HL130760-01, T32 GM008449]
  2. NSF [ECCS-1542205, DMR-1720139]
  3. State of Illinois
  4. Northwestern University
  5. NCI [CA060553]
  6. Biological Imaging Facility at Northwestern University

向作者/读者索取更多资源

Patients suffering from acute or sustained thrombocytopenia require platelet transfusions, which are entirely donor-based and limited by challenges related to storage and fluctuating supply. Developing cell-culture technologies will enable ex vivo and donor-independent platelet production. However, critical advancements are needed to improve scalability and increase megakaryocyte (Mk) culture productivity. To address these needs, we evaluated Mk production from mobilized peripheral blood CD34(+) cells cultured on a commercially available gas-permeable silicone rubber membrane, which provides efficient gas exchange, and investigated the use of fed-batch media dilution schemes. Starting with a cell-surface density of 40 x 10(3) CD34(+) cells per cm(2) (G40D), culturing cells on the membrane for the first 5 days and employing media dilutions yielded 39 +/- 19 CD41(+)CD42b(+) Mks per input CD34(+) cell by day 11-a 2.2-fold increase compared with using standard culture surfaces and full media exchanges. By day 7, G40D conditions generated 1.5-fold more CD34(+) cells and nearly doubled the numbers of Mk progenitors. The increased number of Mk progenitors coupled with media dilutions, potentially due to the retention of interleukin (IL)-3, increased Mk production in G40D. Compared with controls, G40D had higher viability, yielded threefold more Mks per milliliter of media used and exhibited lower mean ploidy, but had higher numbers of high-ploidy Mks. Finally, G40D-Mks produced proplatelets and platelet-like-particles that activate and aggregate upon stimulation. These results highlight distinct improvements in Mk cell-culture and demonstrate how new technologies and techniques are needed to enable clinically relevant production of Mks for platelet generation and cell-based therapies.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据