scholarly journals Efficient generation of human hepatocytes by the intrahepatic delivery of clonal human mesenchymal stem cells in fetal sheep

Hepatology ◽  
2007 ◽  
Vol 46 (6) ◽  
pp. 1935-1945 ◽  
Author(s):  
Jason Chamberlain ◽  
Takashi Yamagami ◽  
Evan Colletti ◽  
Neil D. Theise ◽  
Jyoti Desai ◽  
...  
Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1195-1195 ◽  
Author(s):  
Takashi Yamagami ◽  
Chad Sanada ◽  
Heinz Wiendl ◽  
Esmail D. Zanjani ◽  
Christopher D. Porada ◽  
...  

Abstract Although mesenchymal stem cells (MSC) have been shown to be fairly non-immunogenic, recent studies demonstrated that MSC are capable of activating and becoming targets of NK-mediated lysis. Furthermore, the ability of IFN-γ to induce MSC expression of HLA-II molecules shows that MSC are not completely immuno-inert. Expression of HLA-G has been associated with the maintenance of fetomaternal tolerance during pregnancy through its inhibition of the cytolytic functions of NK and cytotoxic T cells and of dendritic cell maturation. Here we investigated whether transduction of human MSC with a retroviral vector encoding HLA-G1 (MSC-G1) or G5 (MSC-G5) would render these cells and their differentiated progeny undetectable by the recipient’s immune system, and thereby allow us to efficiently transplant these cells into immuno-competent xenogeneic recipients. First, we performed mixed lymphocyte reactions using unmodified MSC (unMSC), MSC-G1 or MSC-G5 as stimulators, and lymphocytes from allogeneic human or sheep donors as responders. While neither unMSC nor MSC-G1 or -G5 elicited a significant proliferative response from human lymphocytes, sheep lymphocytes proliferated 2-6 fold more when cultured with unMSC than with MSC-G1 or MSC-G5. Next, we tested whether HLA-G1 and HLA-G5 expression would enable the engraftment of human MSC in fetal sheep later in gestation when donor cells are normally rejected due to the presence of a competent immune system. To this end, 105 unMSC, MSC-G1, or MSC-G5 were transplanted (Tx) into fetal sheep recipients during the pre-immune period (55 days; n=9), or after immunocompetence was achieved, at 82 days (n=10) or 104 days (n=8) of gestation. Evaluation of the recipients’ hematopoietic system at 42 days post-transplant for the presence of human cell engraftment, using a panel of antibodies specific to human blood cells, revealed that Tx at 55 days resulted in similar levels of engraftment for all cell types (MSC:4±0.9; MSC-G1:6±0.3; MSC-G5:5±0.3%). In contrast, while unMSC engrafted at very low levels at 82d and 104d, Tx of MSC-G1 and-G5 at these later time points not only overcame the immune barriers, but resulted in levels of engraftment that were considerably higher than those achieved during the pre-immune period (82d:7±1;18±2.% and 104d:12±2;16±2%). A similar outcome was seen with liver engraftment and hepatic differentiation with MSG-1 and -G5 giving rise to 3–5 times more hepatocytes than unMSC at later time points. However, even at the early Tx time point of 55days, MSC-G1 and -G5 gave rise to, respectively, 5 times and 2 times more donor-derived hepatocytes than their unMSC counterpart. Our studies demonstrate that the forced expression of HLA-G1 or G5 enables MSC to evade a competent recipient immune system and engraft at significant levels at times in gestation when donor cells are normally rejected. These studies may allow the broadening of the use of MSC to diseases in which an underlying MSC defect precludes the use of the patient’s own MSC.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 436-436 ◽  
Author(s):  
Evan J. Colletti ◽  
Judith A. Airey ◽  
Esmail D. Zanjani ◽  
Christopher D. Porada ◽  
Graça Almeida-Porada

Abstract Despite the exciting reports regarding the ability of human mesenchymal stem cells (MSC) to differentiate into different cells of different organs, the mechanism by which this process occurs remains controversial. Several possible explanations have been put forth as an alternative to the existence of a true differentiation mechanism. We previously showed that MSC, at a single cell level, are able to differentiate into cells of different germ cell layers. In the present study, we investigated whether transfer of mitochondria or membrane-derived vesicles between cells and/or cell fusion participate in the events that lead to the change of phenotype of MSC upon transplantation (Tx). To this end, 54 sheep fetuses (55–60 gestational days) were Tx intra-peritoneally with Stro-1+,CD45−, Gly-A- MSC labeled prior to Tx with either CFSE, that irreversibly couples to both intracellular and cell-surface proteins, or DiD that efficiently labels all cell membranes and intracellular organelles, such as mitochondria. Evaluation of the recipients’ different organs started at 20h post-Tx and continued at 25,30,40,60 and 120h. MSC reached the liver at 25h post-Tx (0.033%±0.0) with maximal engraftment at 40h (0.13%±0.02). MSC were first detected in the lung (0.028%±0.0) and brain (0.034%±0.0) at 30h and 40h respectively. In the brain, engraftment peaked at 60 hours post-Tx (0.08%±0.0) and in the lung at 120h (0.09%±0.01). Normalization of the number of engrafted cells per tissue mass and number of Tx cells revealed that 26% of the Tx MSC reached the lung; 2% the liver; and 3% the brain. Since the decreasing number of CFSE+ and DiD+ cells detected after 120h could be due to cell division, Ki67 staining was performed and revealed that 85–95% of the engrafted cells proliferated upon lodging in the organs, and divided throughout the evaluation period. To determine MSC differentiative timeline, confocal microscopy was performed to assess whether CFSE+ or DiD+ cells expressed tissue-specific markers (MSC were negative for these markers prior to transplant) within the engrafted organs. In the liver at 25h post-Tx, all CFSE+ or DiD+ cells co-expressed alpha-fetoprotein, demonstrating the rapid switch from an MSC to a fetal hepatocyte-like phenotype. In the lung, co-localization of pro-surfactant protein and CFSE/DiD was first detected at 30h post-Tx, but cells remained negative for Caveolin1; a phenotype that is consistent with differentiation to a type II epithelial cell, but not to a more mature type I. In the brain, MSC expressed Tau promptly, but synaptophysin expression was not detected until 120h. In situ hybridization on serial sections using either a human- or sheep-specific probe, with simultaneous visualization of CFSE+ or DiD+ cells allowed us to show that no membrane or mitochondrial transfer had occurred, since none of the sheep cells contained CFSE or DiD, and all of the dye+ cells hybridized only to the human probe. Furthermore, this combined methodology enabled us to determine that differentiation to all of the different cell types had occurred in the absence of cell fusion. In conclusion, MSC engraft multiple tissues rapidly, undergo proliferation, and give rise to tissue-specific cell types in the absence of cellular fusion or the transfer of mitochondria or membrane vesicles.


Circulation ◽  
2004 ◽  
Vol 109 (11) ◽  
pp. 1401-1407 ◽  
Author(s):  
Judith A. Airey ◽  
Graça Almeida-Porada ◽  
Evan J. Colletti ◽  
Christopher D. Porada ◽  
Jason Chamberlain ◽  
...  

2000 ◽  
Vol 191 (4) ◽  
pp. S96 ◽  
Author(s):  
Kenneth W Liechty ◽  
Tippi C Saydam ◽  
Aimen Shaaban ◽  
Ross Milner ◽  
AnneMarie B Moseley ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 676-676
Author(s):  
Jason Chamberlain ◽  
Christopher D. Porada ◽  
Ana Frias ◽  
Takashi Yamagami ◽  
Esmail D. Zanjani ◽  
...  

Abstract The ability of bone marrow-derived Hematopoietic and Mesenchymal Stem Cells (MSC) to generate hepatocytes in a large non-injury sheep model of human stem cell transplantation has been demonstrated. Although in our sheep model the liver is still a hematopoietic organ at the time of transplantation, we hypothesized that the human BM-derived MSCs migrated preferentially to the BM, thus leaving few MSCs available to lodge in the liver and give rise to hepatocytes. Thus, in the present studies, we compared the ability and efficiency of clonally-derived BM MSC populations to give rise to liver cells upon either intra-hepatic (IH) or intra-peritoneal (IP) transplantation into fetal sheep. Five MSC clones were established from human adult BM by single cell deposition of Stro-1+ Gly-A-CD45- cells and expanded until sufficient cells were obtained. Phenotypic characterization demonstrated that all clones were positive for CD90, CD13, and CD29, and negative for hematopoietic markers such as CD34, CD133, and CD45. Five different clonal populations were used and each clone was transplanted either IP (n=7) or IH (n=6) into 55-60 days old fetal sheep recipients at 5x105cells/fetus. Evaluation of the recipients at 56-70 days post-transplant showed that clonally-derived MSC were not only able to generate hepatocytes, but they were also able to generate hematopoietic cells detectable both in the BM and in the PB. IP transplanted animals had a significantly higher level of human CD45+ in their BM (CD45=2.7±0.4%) compared to the IH transplanted animals (CD45=0.25±0.05%), demonstrating that IP injection favors the homing of these cells to the BM while upon IH injection, the human blood cells generated remain in circulation (2.6% CD45+). When we evaluated the presence of hematopoietic cells in the livers of these animals, the IP transplanted animals contained almost 2% CD45+ cells within their livers, while IH transplanted animals had levels of only 0.5-0.65%. When we looked at the generation of liver cells in both groups of animals by immunohistochemistry using monoclonal antibodies specific for human hepatocytes and albumin, we observed that IH injection was much more efficient at generating human hepatocytes than IP injection. The levels of human hepatocytes in most of the IH transplanted animals (10-15% positive cells) were roughly 5X higher than their IP counterparts (2.5-3%). Furthermore, human hepatocytes produced following IH injection were widely distributed throughout the liver parenchyma, while IP injection resulted in a periportal pattern of human hepatocytic activity. We also confirmed the functionality of the generated hepatocytes by detecting human albumin in circulation in these animals by ELISA. In conclusion, these studies show that in a non-injury model the number of available donor cells reaching the liver can be controlled by altering the route of administration, and that this availability determines both the levels of donor-derived hepatocytes generated and the pattern of distribution of these hepatocytes throughout the recipient liver.


PLoS ONE ◽  
2013 ◽  
Vol 8 (3) ◽  
pp. e57759 ◽  
Author(s):  
Maximilian Y. Emmert ◽  
Benedikt Weber ◽  
Petra Wolint ◽  
Thomas Frauenfelder ◽  
Steffen M. Zeisberger ◽  
...  

2010 ◽  
Vol 30 (6) ◽  
pp. 455-455 ◽  
Author(s):  
Dongyan Shi ◽  
Dan Ma ◽  
Feiqing Dong ◽  
Chen Zong ◽  
Liyue Liu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document