4.6 Article

Functional Maintenance of Differentiated Embryoid Bodies in Microfluidic Systems: A Platform for Personalized Medicine

Journal

STEM CELLS TRANSLATIONAL MEDICINE
Volume 4, Issue 3, Pages 261-268

Publisher

WILEY
DOI: 10.5966/sctm.2014-0119

Keywords

Stem cells; Cryopreservation; Microenvironment; Microphysiological systems; Hormone secretion; Reproductive medicine

Funding

  1. National Science Foundation under NSF CAREER [1150733, R01 EB015776]
  2. Michael Cassidy and Caroline Wang, the Department of OB GYN, BWH
  3. MSys Inc., India
  4. NIH [P510D011106]
  5. Research Facilities Improvement Program [RR15459-01, RR020141-01]
  6. Harvard NeuroDiscovery Center
  7. Enhanced Neuroimaging Core Facility
  8. Directorate For Engineering
  9. Div Of Chem, Bioeng, Env, & Transp Sys [1461602] Funding Source: National Science Foundation

Ask authors/readers for more resources

Hormone replacement therapies have become important for treating diseases such as premature ovarian failure or menopausal complications. The clinical use of bioidentical hormones might significantly reduce some of the potential risks reportedly associated with the use of synthetic hormones. In the present study, we demonstrate the utility and advantage of a microfluidic chip culture system to enhance the development of personalized, on-demand, treatment modules using embryoid bodies (EBs). Functional EBs cultured on microfluidic chips represent a platform for personalized, patient-specific treatment cassettes that can be cryopreserved until required for treatment. We assessed the viability, differentiation, and functionality of EBs cultured and cryopreserved in this system. During extended microfluidic culture, estradiol, progesterone, testosterone, and anti-mullerian hormone levels were measured, and the expression of differentiated steroidogenic cells was confirmed by immunocytochemistry assay for the ovarian tissue markers anti-mullerian hormone receptor type II, follicle-stimulating hormone receptor, and inhibin p-A and the estrogen biosynthesis enzyme aromatase. Our studies showed that under microfluidic conditions, differentiated steroidogenic EBs continued to secrete estradiol and progesterone at physiologically relevant concentrations (30-120 pg/ml and 150-450 pg/ml, respectively) for up to 21 days. Collectively, we have demonstrated for the first time the feasibility of using a microfluidic chip system with continuous flow for the differentiation and extended culture of functional steroidogenic stem cell-derived EBs, the differentiation of EBs into cells expressing ovarian antigens in a microfluidic system, and the ability to cryopreserve this system with restoration of growth and functionality on thawing. These results present a platform for the development of a new therapeutic system for personalized medicine.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available