In vitro transdifferentiation of adult bone marrow Sca-1+cKit− cells cocultured with fetal liver cells into hepatic-like cells without fusion

2006 ◽  
Vol 34 (1) ◽  
pp. 97-106 ◽  
Author(s):  
Yasuhiro Yamada ◽  
Eishi Nishimoto ◽  
Hiroaki Mitsuya ◽  
Yuji Yonemura
Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2707-2707
Author(s):  
Ahmed Waraky ◽  
Anders Östlund ◽  
Laleh Arabanian ◽  
Tina Nilsson ◽  
Linda Fogelstrand ◽  
...  

Introduction: Non-random cytogenetic aberrations are often involved in the development of AML in children and several aberrations can serve as diagnostic markers, prognosis predictors and impact choice of therapy. In infant AML, a chromosomal translocation t(7;12)(q36;p13) has been found in up to 20-30 % of the cases, making it the second most common genetic aberration in this age group after KMT2A (MLL) rearrangements. Previous studies indicate that this patient group has a dismal prognosis with virtually no event-free survival. Limiting the chances to improve this is the lack of understanding how the t(7;12)(q36;p13) is involved in leukemia development. The translocation leads to a gene fusion MNX1-ETV6 but also to increased MNX1 gene expression. Although both ETV6 and MNX1 are expressed in normal hematopoietic tissues, the role of the fusion protein MNX1-ETV6in the development of AML is not established. Also unclear is whether the driver of leukemogenesis is the fusion itself or the simultaneous overexpression of MNX1. The aim of this study was to assess the transformation capacity and the molecular mechanism of the MNX1-ETV6 fusion and the overexpressed MNX1in vitro and in vivo using murine transplantation models. Material and methods: In a liquid culture system, we introduced the MNX1-ETV6 fusion, MNX1 overexpression, or empty vector into primary murine (C57BL/6) hematopoietic progenitor cells with retroviral transfection. Cells were isolated from either adult bone marrow after 5-FU stimulation, or from fetal liver at E14.5. After enrichment by fluorescence activated cell sorting based on vector co-expressed green/yellow fluorescence protein, transfected cells were used for in vitro experiments and for transplantation into lethally irradiated immunocompetent C57BL/6 mice or sub-lethally irradiated immunocompromised NSGW41 mice. In vitro, cells were assessed with RNA sequencing for gene expression, gamma H2AX assay for DNA double strand brakes, flow cytometry for lineage marker expression, apoptosis and proliferation, and with colony forming unit assay. Results: Upon transplantation, only fetal liver cells transduced with MNX1 or with MNX1-ETV6 fusion were able to induce leukemia in immunocompromised (NSGW41) mice. When MNX1 or MNX1-ETV6 transduced cells were transplanted into immunocompetent mice (C57BL/6) mice, no leukemia development was seen, when either fetal liver or adult bone marrow cells were used for transduction. However, when immunocompromised mice were transplanted with MNX1 or MNX1-ETV6 fusion expressing cells they typically developed signs of disease after 1-2 months and exhibited leukocytosis and elevated blast cells in blood and bone marrow, severe anemia, and enlarged spleens with infiltration of leukemic cells. The cells showed expression of predominantly myeloid markers. In vitro, cells with overexpression of MNX1 or MNX1-ETV6 fusion expression also showed altered lineage differentiation in favor of myeloid differentiation. In addition, MNX1 overexpressing cells, but not MNX1-ETV6 expressing cells, exhibited increased proliferation and colony formation capacity. Both MNX1 overexpressing and MNX1-ETV6 fusion expressing cells showed increased DNA damage as evident from an increased gamma-phosphorylated H2AX in fetal liver and adult bone marrow transduced cells respectively, accompanied with G1 arrest, compared to cells transduced with empty vectors. Both MNX1 and MNX1-ETV6 also led to increased apoptosis in adult bone marrow (3-fold) and to a lesser extent in fetal liver cells (1.5-fold). Results from transcriptome sequencing showed enrichment for specific pathways in G2/M transition of cell cycle in cells transduced by either MNX1or the MNX1-ETV6 fusion. Further investigations to elucidate the molecular mechanisms and pathways through which MNX1 and/or MNX1-ETV6 induce leukemia is ongoing. Conclusions: MNX1 overexpression and MNX1-ETV6 fusion, both characteristics of infant AML with t(7;12)(q36;p13), induced leukemogenic effects in both fetal liver cells and adult bone marrow cells, but could cause a myeloid leukemia only under immunocompromised conditions. This may be of importance for the exclusive prevalence of this AML subtype in young children, with the highest peak during the first six months of life when the immune system is less developed. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1979 ◽  
Vol 54 (5) ◽  
pp. 1091-1100 ◽  
Author(s):  
JP Blanchet ◽  
J Samarut ◽  
G Mouchiroud

Abstract The production of erythrocytes bearing an “immature” antigen (Im+ cells) and a “fetal” antigen (Ft+ cells) has been studied in irradiated adult mice grafted either with fetal liver or adult bone marrow cells. The Im+ cells reach a peak 8–11 days after grafting. Ft+ cells are detected only after graft of fetal liver cells; the younger the liver, the greater the number. Since Ft+ cells are rapidly and briefly produced, they could be the progeny of erythroid-committed precursors, which are particularly numerous among fetal liver cells. Environmental factors directing the erythropoietic differentiation towards Ft+ erythrocytes in fetuses or Ft- erythrocytes in adults are proposed.


Blood ◽  
1979 ◽  
Vol 54 (5) ◽  
pp. 1091-1100
Author(s):  
JP Blanchet ◽  
J Samarut ◽  
G Mouchiroud

The production of erythrocytes bearing an “immature” antigen (Im+ cells) and a “fetal” antigen (Ft+ cells) has been studied in irradiated adult mice grafted either with fetal liver or adult bone marrow cells. The Im+ cells reach a peak 8–11 days after grafting. Ft+ cells are detected only after graft of fetal liver cells; the younger the liver, the greater the number. Since Ft+ cells are rapidly and briefly produced, they could be the progeny of erythroid-committed precursors, which are particularly numerous among fetal liver cells. Environmental factors directing the erythropoietic differentiation towards Ft+ erythrocytes in fetuses or Ft- erythrocytes in adults are proposed.


Blood ◽  
2002 ◽  
Vol 99 (5) ◽  
pp. 1870-1872 ◽  
Author(s):  
Patricia A. Taylor ◽  
Ronald T. McElmurry ◽  
Christopher J. Lees ◽  
David E. Harrison ◽  
Bruce R. Blazar

In utero transplantation (IUT) is becoming a viable option for the treatment of various immune and metabolic disorders diagnosed early in gestation. In this study, donor fetal liver cells had a 10-fold competitive engraftment advantage relative to adult bone marrow in allogeneic fetal severe combined immunodeficient (SCID) recipients compared with adult recipients. In contrast, adult bone marrow cells engrafted slightly better than fetal liver cells in allogeneic adult SCID transplant recipients. By using different ratios of fetal and adult cell mixtures, fetal liver cells repopulated 8.2 times better than adult bone marrow cells in fetal recipients, but only 0.8 times as well in adult recipients. Fetal SCID recipients were more permissive to an allogeneic donor graft than adult recipients. These data indicate that the recipient microenvironment may regulate the engraftment efficiency of a given stem cell source and suggest that the use of cord blood should be tested in clinical IUT.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Yi Yu ◽  
Alejandra Vargas Valderrama ◽  
Zhongchao Han ◽  
Georges Uzan ◽  
Sina Naserian ◽  
...  

Abstract Background Mesenchymal stem cells (MSCs) exhibit active abilities to suppress or modulate deleterious immune responses by various molecular mechanisms. These cells are the subject of major translational efforts as cellular therapies for immune-related diseases and transplantations. Plenty of preclinical studies and clinical trials employing MSCs have shown promising safety and efficacy outcomes and also shed light on the modifications in the frequency and function of regulatory T cells (T regs). Nevertheless, the mechanisms underlying these observations are not well known. Direct cell contact, soluble factor production, and turning antigen-presenting cells into tolerogenic phenotypes, have been proposed to be among possible mechanisms by which MSCs produce an immunomodulatory environment for T reg expansion and activity. We and others demonstrated that adult bone marrow (BM)-MSCs suppress adaptive immune responses directly by inhibiting the proliferation of CD4+ helper and CD8+ cytotoxic T cells but also indirectly through the induction of T regs. In parallel, we demonstrated that fetal liver (FL)-MSCs demonstrates much longer-lasting immunomodulatory properties compared to BM-MSCs, by inhibiting directly the proliferation and activation of CD4+ and CD8+ T cells. Therefore, we investigated if FL-MSCs exert their strong immunosuppressive effect also indirectly through induction of T regs. Methods MSCs were obtained from FL and adult BM and characterized according to their surface antigen expression, their multilineage differentiation, and their proliferation potential. Using different in vitro combinations, we performed co-cultures of FL- or BM-MSCs and murine CD3+CD25−T cells to investigate immunosuppressive effects of MSCs on T cells and to quantify their capacity to induce functional T regs. Results We demonstrated that although both types of MSC display similar cell surface phenotypic profile and differentiation capacity, FL-MSCs have significantly higher proliferative capacity and ability to suppress both CD4+ and CD8+ murine T cell proliferation and to modulate them towards less active phenotypes than adult BM-MSCs. Moreover, their substantial suppressive effect was associated with an outstanding increase of functional CD4+CD25+Foxp3+ T regs compared to BM-MSCs. Conclusions These results highlight the immunosuppressive activity of FL-MSCs on T cells and show for the first time that one of the main immunoregulatory mechanisms of FL-MSCs passes through active and functional T reg induction.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2635-2635
Author(s):  
Marie Terpager ◽  
Hiroshi Kataoka ◽  
Ivo Cornelissen ◽  
Shaun R. Coughlin

Abstract Abstract 2635 G protein-coupled receptors (GPCRs) can regulate cell migration, survival, proliferation and differentiation –– key processes in hematopoiesis. The Gi-coupled receptor CXCR4 plays a key role in hematopoiesis, suggesting that related receptors might also contribute. Because all Gi family members except Gz are inhibited by pertussis toxin (PTX), we utilized a ROSA26-CreOnPTX mouse line that expresses PTX in a Cre-dependent manner to broadly probe the role of Gi signaling in hematopoiesis. Mice hemizygous for the hematopoietic lineage-specific Cre transgene Vav-iCre were crossed with ROSA26-CreOnPTX/CreOnPTX mice to generate offspring expressing PTX in hematopoietic lineages (Vav-PTX) and Cre-negative controls in which the PTX allele remained silent. Vav-PTX mice were born at the expected Mendelian rate, and except for a smaller thymus, were grossly normal, but all died with pneumonia between days 2 and 14. Bone marrow in 3 day-old Vav-PTX mice was hypocellular with significant underrepresentation of granulocytic and lymphocytic lineages as well as hematopoietic stem and progenitor cells (lin-, c-kit+, Sca1+). In bone marrow reconstitution studies, cells from Vav-PTX fetal livers (E14.5) showed impaired short-term and no long-term repopulating activity. Additionally, Vav-PTX fetal liver cells were significantly impaired in their ability to form granulocyte/macrophage and erythroid colonies in vitro. Interestingly, when wild-type E14.5 fetal liver cells were grown in vitro in presence of exogenous PTX, only erythroid colony formation was impaired, and flow cytometric analysis of the progenitor populations of Vav-PTX fetal liver revealed a significant decrease in granulocyte-macrophage progenitors (GMPs) as well as in common myeloid progenitors (CMPs) but not in megakaryocyte/erythroid progenitors (MEPs). Thus, reduced progenitor populations may account for reduced granulocyte/macrophage colony-forming activity in fetal liver cell cultures but does not account for reduced erythroid colony-forming activity. Indeed, normal MEP numbers in Vav-PTX livers and the ability of exogenous PTX to inhibit formation of erythroid colonies in wild-type fetal liver cultures suggests that Gi signaling in MEPs or their progeny may contribute to erythropoiesis in fetal liver. Several of the necessary roles of Gi signaling identified above are not accounted for by the function of CXCR4, and, taken together, our data suggest that Gi-coupled GPCRs likely contribute to hematopoiesis at multiple levels and in different lineages. An effort to identify GPCRs that contribute to erythropoiesis is underway. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 448-448 ◽  
Author(s):  
Baobing Zhao ◽  
Matthew John Schipma ◽  
Yang Mei ◽  
Ganesan Keerthivansan ◽  
Jing Yang ◽  
...  

Abstract Mammalian erythropoiesis is a dynamic process in which erythroid progenitors proliferate and differentiate into mature enucleated red blood cells. In the late stage of terminal erythropoiesis, erythroblasts undergo cell-cycle exit, chromatin condensation, and extrusion of the pycnotic nucleus via an asymmetric cell division. Recent genetic and biochemical studies illustrated that various signaling pathways, including histone deacetylation and other chromatin modifications, are involved in chromatin condensation and enucleation. However, it is unclear the global dynamic changes of the nucleosome and how different histones are regulated during chromatin condensation. We proposed to directly characterize the expression levels and localization of different histones and their variants during erythropoiesis. Using a mouse fetal liver erythroblast in vitro culture system and immunofluorescence analysis, we found an unexpected, gradually enlarged nuclear opening through which most histones, except H2AZ, were released out of the nucleus. The same phenotype was observed in freshly purified fetal liver and bone marrow erythroblasts (Figure 1). These openings lack nuclear lamina, nuclear pore complexes, and nuclear membrane but are distinct from nuclear envelope changes during apoptosis and mitosis. Western blot analysis also demonstrated the nuclear release of a fraction of the major histones, however, many well-known nuclear proteins remained in the nucleus in this process. We also demonstrated that the histone release was cell cycle regulated and independent of nuclear exportin. Using micrococcal nuclease digestion of chromatin followed by next generation sequencing (MNase-seq) technique, we demonstrated that histone release from the nuclear opening is associated with dynamic nucleosomal changes during mouse terminal erythropoiesis. Mechanistically, caspase-3 is involved in the regulation of nuclear openings during erythropoiesis. Inhibition or knockdown of caspase-3 completely blocked nuclear opening formation and histone release, which led to inhibition of chromatin condensation, cell differentiation, and ultimate cell death. In summary, our study revealed a caspase-3 mediated nuclear opening formation with histone release in mouse erythroblasts that is unique in mammalian cells. The dynamic nuclear opening formation is required for fast release of major histones into cytoplasm to facilitate chromatin condensation throughout erythropoiesis. This “prokaryotic phase” of erythroblast is also associated with genome wide nucleosome localization changes that correlate with the size of the nuclear opening and histone release, which may provide clues for the pathogenesis of erythroid related diseases with unknown etiology. Figure 1 Lamin B opening with H2 release in mouse erythropoiesis. (A) E13.5 TER119 negative mouse fetal liver erythroblasts were purified and cultured in vitro in erythropoietin containing medium. Immunofluorescence stains for lamin B, H2A and DNA (DAPI) from erythroblasts cultured on different days were performed. Arrows indicate lamin B openings. Scale bar: 5mm. (B) Same as A except the cells were from fresh total fetal liver cells (left) and bone marrow erythroblasts. Scale bar: 5mm. Figure 1. Lamin B opening with H2 release in mouse erythropoiesis. (A) E13.5 TER119 negative mouse fetal liver erythroblasts were purified and cultured in vitro in erythropoietin containing medium. Immunofluorescence stains for lamin B, H2A and DNA (DAPI) from erythroblasts cultured on different days were performed. Arrows indicate lamin B openings. Scale bar: 5mm. (B) Same as A except the cells were from fresh total fetal liver cells (left) and bone marrow erythroblasts. Scale bar: 5mm. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1999 ◽  
Vol 93 (3) ◽  
pp. 826-837 ◽  
Author(s):  
Fernando Cortés ◽  
Frédéric Deschaseaux ◽  
Nobuko Uchida ◽  
Marie-Claude Labastie ◽  
Annabelle M. Friera ◽  
...  

We have previously shown that the HCA/ALCAM (CD166) glycoprotein, a member of the immunoglobulin family that mediates both homophilic and heterophilic cell-cell adhesion, via the CD6 ligand, is expressed at the surface of all of the most primitive CD38−/lo, Thy-1+, rho123lo, CD34+hematopoietic cells in human fetal liver and fetal and adult bone marrow. In the present report we show that HCA is also expressed by subsets of stromal cells in the primary hematopoietic sites that sequentially develop in the human embryo and fetus, ie, the paraaortic mesoderm, liver, thymus, and bone marrow. Adult bone marrow stromal cells established in vitro, including those derived from Stro-1+ progenitors and cells from immortalized cell lines, express HCA. In contrast, no HCA expression could be detected in peripheral lymphoid tissues, fetal spleen, and lymph nodes. HCA membrane molecules purified from marrow stromal cells interact with intact marrow stromal cells, CD34+ CD38−hematopoietic precursors, and CD3+ CD6+peripheral blood lymphocytes. Finally, low but significant levels of CD6 are here for the first time detected at the surface of CD34+ rho123med/lo progenitors in the bone marrow and in mobilized blood from healthy individuals. Altogether, these results indicate that the HCA/ALCAM surface molecule is involved in homophilic or heterophilic (with CD6) adhesive interactions between early hematopoietic progenitors and associated stromal cells in primary blood-forming organs.


Sign in / Sign up

Export Citation Format

Share Document