Your browser doesn't support javascript.
loading
An on-chip model of protein paracellular and transcellular permeability in the microcirculation.
Offeddu, Giovanni S; Haase, Kristina; Gillrie, Mark R; Li, Ran; Morozova, Olga; Hickman, Dean; Knutson, Charles G; Kamm, Roger D.
Afiliação
  • Offeddu GS; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
  • Haase K; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
  • Gillrie MR; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
  • Li R; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
  • Morozova O; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA; Amgen Research, Amgen Inc., 360 Binney Street, Cambridge, MA, USA.
  • Hickman D; Amgen Research, Amgen Inc., 360 Binney Street, Cambridge, MA, USA.
  • Knutson CG; Amgen Research, Amgen Inc., 360 Binney Street, Cambridge, MA, USA. Electronic address: charlie.knutson@amgen.com.
  • Kamm RD; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. Electronic address: rdkamm@mit.edu.
Biomaterials ; 212: 115-125, 2019 08.
Article em En | MEDLINE | ID: mdl-31112823
ABSTRACT
Recent therapeutic success of large-molecule biologics has led to intense interest in assays to measure with precision their transport across the vascular endothelium and into the target tissue. Most current in vitro endothelial models show unrealistically large permeability coefficients due to a non-physiological paracellular transport. Thus, more advanced systems are required to better recapitulate and discern the important contribution of transcellular transport (transcytosis), particularly of pharmaceutically-relevant proteins. Here, a robust platform technology for the measurement of transport through a human endothelium is presented, which utilizes in vitro microvascular networks (MVNs). The self-assembled MVNs recapitulate the morphology and junctional complexity of in vivo capillaries, and express key endothelial vesicular transport proteins. This results in measured permeabilities to large molecules comparable to those observed in vivo, which are orders of magnitude lower than those measured in transwells. The permeability of albumin and immunoglobulin G (IgG), biopharmaceutically-relevant proteins, is shown to occur primarily via transcytosis, with passage of IgG regulated by the receptor FcRn. The physiological relevance of the MVNs make it a valuable tool to assess the distribution of biopharmaceuticals into tissues, and may be used to prioritize candidate molecules from this increasingly important class of therapeutics.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Permeabilidade Capilar / Proteínas / Dispositivos Lab-On-A-Chip / Microcirculação Limite: Humans Idioma: En Revista: Biomaterials Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Permeabilidade Capilar / Proteínas / Dispositivos Lab-On-A-Chip / Microcirculação Limite: Humans Idioma: En Revista: Biomaterials Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Estados Unidos