PMID- 32493346 OWN - NLM STAT- MEDLINE DCOM- 20210503 LR - 20210503 IS - 2045-8118 (Electronic) IS - 2045-8118 (Linking) VI - 17 IP - 1 DP - 2020 Jun 3 TI - Advances in blood-brain barrier modeling in microphysiological systems highlight critical differences in opioid transport due to cortisol exposure. PG - 38 LID - 10.1186/s12987-020-00200-9 [doi] LID - 38 AB - BACKGROUND: The United States faces a national crisis involving opioid medications, where currently more than 130 people die every day. To combat this epidemic, a better understanding is needed of how opioids penetrate into the central nervous system (CNS) to facilitate pain relief and, potentially, result in addiction and/or misuse. Animal models, however, are a poor predictor of blood-brain barrier (BBB) transport and CNS drug penetration in humans, and many traditional 2D cell culture models of the BBB and neurovascular unit have inadequate barrier function and weak or inappropriate efflux transporter expression. Here, we sought to better understand opioid transport mechanisms using a simplified microfluidic neurovascular unit (NVU) model consisting of human brain microvascular endothelial cells (BMECs) co-cultured with astrocytes. METHODS: Human primary and induced pluripotent stem cell (iPSC)-derived BMECs were incorporated into a microfluidic NVU model with several technical improvements over our previous design. Passive barrier function was assessed by permeability of fluorescent dextrans with varying sizes, and P-glycoprotein function was assessed by rhodamine permeability in the presence or absence of inhibitors; quantification was performed with a fluorescent plate reader. Loperamide, morphine, and oxycodone permeability was assessed in the presence or absence of P-glycoprotein inhibitors and cortisol; quantification was performed with mass spectrometry. RESULTS: We first report technical and methodological optimizations to our previously described microfluidic model using primary human BMECs, which results in accelerated barrier formation, decreased variability, and reduced passive permeability relative to Transwell models. We then demonstrate proper transport and efflux of loperamide, morphine, and oxycodone in the microfluidic NVU containing BMECs derived from human iPSCs. We further demonstrate that cortisol can alter permeability of loperamide and morphine in a divergent manner. CONCLUSIONS: We reveal a novel role for the stress hormone cortisol in modulating the transport of opioids across the BBB, which could contribute to their abuse or overdose. Our updated BBB model represents a powerful tool available to researchers, clinicians, and drug manufacturers for understanding the mechanisms by which opioids access the CNS. FAU - Brown, Jacquelyn A AU - Brown JA AD - Vanderbilt Institute for Integrated Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA. FAU - Faley, Shannon L AU - Faley SL AD - Vanderbilt Institute for Integrated Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA. AD - Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA. FAU - Shi, Yajuan AU - Shi Y AD - Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA. FAU - Hillgren, Kathleen M AU - Hillgren KM AD - Departments of ADME and Toxicology, Eli Lilly and Company, Indianapolis, IN, USA. FAU - Sawada, Geri A AU - Sawada GA AD - Departments of ADME and Toxicology, Eli Lilly and Company, Indianapolis, IN, USA. FAU - Baker, Thomas K AU - Baker TK AD - Departments of ADME and Toxicology, Eli Lilly and Company, Indianapolis, IN, USA. FAU - Wikswo, John P AU - Wikswo JP AD - Vanderbilt Institute for Integrated Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA. john.p.wikswo@vanderbilt.edu. AD - Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA. john.p.wikswo@vanderbilt.edu. AD - Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA. john.p.wikswo@vanderbilt.edu. AD - Department of Physics, Vanderbilt University, Nashville, TN, USA. john.p.wikswo@vanderbilt.edu. FAU - Lippmann, Ethan S AU - Lippmann ES AUID- ORCID: 0000-0001-5703-5747 AD - Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA. ethan.s.lippmann@vanderbilt.edu. AD - Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA. ethan.s.lippmann@vanderbilt.edu. AD - Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA. ethan.s.lippmann@vanderbilt.edu. LA - eng GR - UH3 TR002097/TR/NCATS NIH HHS/United States GR - 5UG3TR002097-02/NH/NIH HHS/United States GR - 5UG3TR002097-02S1/NH/NIH HHS/United States PT - Journal Article DEP - 20200603 PL - England TA - Fluids Barriers CNS JT - Fluids and barriers of the CNS JID - 101553157 RN - 0 (Analgesics, Opioid) RN - WI4X0X7BPJ (Hydrocortisone) SB - IM MH - Analgesics, Opioid/*pharmacokinetics MH - Astrocytes/drug effects/*physiology MH - Blood-Brain Barrier/*drug effects/*physiology MH - Cells, Cultured MH - Coculture Techniques MH - Endothelial Cells/drug effects/*physiology MH - Humans MH - Hydrocortisone/*metabolism MH - Induced Pluripotent Stem Cells/drug effects/*physiology MH - Microvessels/cytology MH - *Models, Neurological PMC - PMC7269003 COIS- Elements of the work contained in this manuscript were funded by a collaborative agreement between Eli Lilly and Company and Vanderbilt University. Eli Lilly and Company did not specify results or outcomes for the work contained in this manuscript. EDAT- 2020/06/05 06:00 MHDA- 2021/05/04 06:00 PMCR- 2020/06/03 CRDT- 2020/06/05 06:00 PHST- 2019/12/20 00:00 [received] PHST- 2020/05/27 00:00 [accepted] PHST- 2020/06/05 06:00 [entrez] PHST- 2020/06/05 06:00 [pubmed] PHST- 2021/05/04 06:00 [medline] PHST- 2020/06/03 00:00 [pmc-release] AID - 10.1186/s12987-020-00200-9 [pii] AID - 200 [pii] AID - 10.1186/s12987-020-00200-9 [doi] PST - epublish SO - Fluids Barriers CNS. 2020 Jun 3;17(1):38. doi: 10.1186/s12987-020-00200-9.