scholarly journals Nanocapsule-delivered Sleeping Beauty mediates therapeutic Factor VIII expression in liver sinusoidal endothelial cells of hemophilia A mice

Author(s):  
Betsy T. Kren ◽  
Gretchen M. Unger ◽  
Lucas Sjeklocha ◽  
Alycia A. Trossen ◽  
Vicci Korman ◽  
...  
Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2219-2219
Author(s):  
Siyuan Tan ◽  
Kai Chen ◽  
Arjan van der Flier ◽  
Zhan Liu ◽  
David R. Light ◽  
...  

Abstract Abstract 2219 rFVIIIFc is a recombinant fusion protein consisting of human B-domain deleted factor VIII covalently linked to the Fc domain of IgG1. In hemophilia A patients, rFVIIIFc has been shown to display a ∼1.6-fold longer half-life than recombinant full length FVIII (Advate®) (Powell et al., 2012. Blood). This half-life extension can be attributed to a natural pathway mediated by the neonatal Fc receptor (FcRn) that re-circulates IgG molecules into the vascular system, as the long-lasting activity of rFVIIIFc is not observed in FcRn knockout mice. To identify the cell type that takes up and subsequently protects and recycles rFVIIIFc, we have recombinantly replaced the missing B-domain with a Halo tag in rFVIIIFc (rFVIIIFc-Halo) to allow visualization of the protein in the presence of fluorescently labeled Halo-ligand using confocal microscopy. Purified rFVIIIFc-Halo protein displayed similar specific activity and pharmacokinetic properties as rFVIIIFc in hemophilia A (HemA) mice, indicating that the addition of the Halo tag does not alter the functionality and the clearance mechanisms of rFVIIIFc. In quantitative whole body autoradiography studies (QWBA) in HemA mice with radiolabeled rFVIIIFc, we observed that 125I-rFVIIIFc is predominately distributed to the liver. Therefore, we selected primary liver cells isolated from HemA mice to study cellular uptake of rFVIIIFc. A co-culture of hepatocytes and non-parenchymal cells was isolated from HemA mice and prepared at a 1:1 ratio. Liver sinusoidal endothelial cells (LSECs) and Kupffer cells (KCs) in this culture were identified by fluorescently labeled antibodies to CD31 and F4/80 respectively. Both cell types effectively took up the fluorescently labeled AcLDL, confirming that the isolated LSECs and KCs retained the capacity for functional endocytosis in vitro. It was found that LSECs, as opposed to Kupffer cells or hepatocytes, are predominantly responsible for the cellular uptake of rFVIIIFc, as the localization of rFVIIIFc-Halo is apparent only in LSECs within 5 minutes after exposing 10 nM of rFVIIIFc-Halo to primary co-culture freshly isolated from HemA mice. In contrast, even with longer exposure time (up to 1 hour) and higher protein concentration (up to 40 nM), the localization of rFVIIIFc-Halo in Kupffer cells and hepatocytes still remains undetectable. Analysis of recombinant Halo-tagged factor VIII (rFVIII-Halo) yielded similar results, suggesting that the Fc-fusion does not alter the cellular uptake pathway of FVIII, which is consistent with the notion that the interaction of Fc with FcRn occurs at the intracellular level. Therefore, interestingly, both rFVIII-Halo and rFVIIIFc-Halo are internalized by LSEC that are the same cells reported to express FVIII by in situ hybridization studies (Hollestelle et al. 2001 Thromb Haemost). This study, together with recent findings that somatic cells in the liver are primarily responsible for rFVIIIFc recycling (Abstract by van der Flier et al), highlights the critical role of LSECs in the clearance of rFVIIIFc and suggests that rFVIIIFc is primarily recycled by FcRn in LSECs. The impact of VWF on the cellular uptake and recycling of the rFVIIIFc-VWF complex in liver cells may also be assessed utilizing this system. Disclosures: Tan: BiogenIdec: Employment. Chen:BiogenIdec: Employment. van der Flier:BiogenIdec: Employment. Liu:BiogenIdec: Employment. Light:biogenidec: Employment. Jiang:biogenidec: Employment.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1615-1615
Author(s):  
Matthew Y. Suh ◽  
Shirine Dada ◽  
Humberto Azpurua ◽  
Evelyn Flynn ◽  
David M. Briscoe ◽  
...  

Abstract Synthesis of factor VIII by liver sinusoidal endothelial cells has been documented, and liver transplantation for end stage liver disease has been shown to cure concomitant hemophilia A. Given the irreversible nature of liver transplantation, the spleen is an alternate organ of interest in transplant therapy to cure hemophilia A. Factor VIII gene expression in the spleen has been documented by RT-PCR (Hollestelle, M. J. et al, 2001, Thromb Haemost86:855–61), and a recent report of living related splenic transplantation for hemophilia A has shown symptom-free survival up to 4 years (Jiang, H. C. et al, 2006, Transplant Proc38:1483). However, it is unclear which cell populations in the spleen synthesize factor VIII. Immunohistochemistry of murine spleen showed factor VIII staining of the sinusoidal endothelial cells as expected. Surprisingly, clusters of large mononuclear cells in the red pulp also intensely stained for factor VIII. CD31 staining revealed same pattern of sinusoidal endothelial cells and large mononuclear cells in the red pulp. Single cell suspensions of murine spleen analyzed by flow cytometry corroborated significant factor VIII expression (47–70% of all splenic cells). At least 60% of splenic cells were CD31+ CD45+, and these CD31+ CD45+ cells showed significant factor VIII expression (38–48%). When CD31+ cells were depleted by magnetic cell sorting, there was still a subgroup of cells expressing factor VIII (approximately 50%). In summary, we have shown that a significant proportion of splenic cells synthesize factor VIII by flow cytometry and immunohistochemistry. In addition to the sinusoidal endothelial cells, CD31+ CD45+ cells (possibly the large mononuclear cells seen on immunohistochemistry) contribute to splenic synthesis of factor VIII. Based on CD31 depletion study, there is still an unidentified group of cells (CD31-) that contributes to factor VIII synthesis. Further characterization of these cells and an in vivo study in a murine hemophilia A model have been initiated. We conclude that the spleen is a viable source of factor VIII and may be useful in cell-based and/or organ transplant therapy of hemophilia A.


1999 ◽  
Vol 274 (28) ◽  
pp. 19587-19592 ◽  
Author(s):  
Hung Do ◽  
John F. Healey ◽  
Edmund K. Waller ◽  
Pete Lollar

2020 ◽  
Vol 40 (S 01) ◽  
pp. S26-S31
Author(s):  
Osman El-Maarri ◽  
Muhammad Ahmer Jamil ◽  
Johannes Oldenburg

AbstractHuman factor VIII (FVIII), which deficiency leads to hemophilia A, is largely synthesized and secreted by the liver sinusoidal endothelial cells (LSECs). However, the characteristics of these cells that secrete FVIII are not well known. We have previously reported that based on genome-wide expression and CpG methylation profiling, LSECs have a distinct molecular profile that distinguishes them from other endothelial cells. Hepatocytes are targeted by gene therapy protocols to treat hemophilia A. However, the hepatocyte is not the natural site for FVIII synthesis and current gene therapy protocols are eliciting immune responses that require immune suppression with corticosteroid therapy in a fairly high proportion of patients over a significant period of time. Cellular stress because of ectopic FVIII expression and codon optimization are discussed as potential underlying mechanisms. Here, we highlight the molecular differences between LSECs and hepatocytes.


2014 ◽  
Vol 12 (1) ◽  
pp. 36-42 ◽  
Author(s):  
T. Shahani ◽  
K. Covens ◽  
R. Lavend'homme ◽  
N. Jazouli ◽  
E. Sokal ◽  
...  

PLoS ONE ◽  
2013 ◽  
Vol 8 (10) ◽  
pp. e77255 ◽  
Author(s):  
Marina E. Fomin ◽  
Yanchen Zhou ◽  
Ashley I. Beyer ◽  
Jean Publicover ◽  
Jody L. Baron ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document