Transdifferentiation of Bone Marrow-Derived Cells Co-Cultured with Fetal Liver Cells into Hepatic-Like Cells without Fusion.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4176-4176
Author(s):  
Yasuhiro Yamada ◽  
Yuji Yonemura ◽  
Eishi Nishimoto ◽  
Hiroaki Mitsuya

Abstract Several research groups have reported that bone marrow cells (BMCs) transdifferentiate into hepatocytes in rodents. However, it is yet to be studied what factors effectively trigger and sustain the transdifferentiation of BMCs to hepatocytes. In the present study, we investigated whether murine BMCs in the presence of fetal liver cells (FLCs) could differentiate into hepatic-like cells in vitro without fusion. Fractionated BMCs from C57Bl/6-TgN(ACTbEGFP)10bs mice and FLCs from B6.129S7-Gt(ROSA)26Sor mice were co-cultured at 1x105 cells and 1x106 cells in 10% FCS-containing medium supplemented with hepatocyte growth factor on laminin-coated dishes. Hepatocyte-specific markers among BMCs were detected as assessed by immunocytochemistry for albumin and reverse transcription-polymerase chain reaction (RT-PCR) for alpha-fetoprotein (AFP), albumin, and cytokeratin-19 mRNAs. We also found that Sca-1+ BMCs containing both hematopoietic stem cells and AFP-expressing cells could differentiate into hepatic-like cells and such cells were seen adherent to dish together with FLCs in the early phase of culture. Moreover, the AFP-expressing cells were found in a Sca-1+ cKit- cell fraction, which also differentiated into CD45− GFP+ albumin+ cells and proved to be positive for GFP but negative for LacZ as assessed by RT-PCR and immunocytochemistry. These results suggest that albumin+ cells developed through transdifferentiation from BMCs but not through spontaneous cell fusion between BMCs and FLCs.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2525-2525
Author(s):  
Takuo Katsumoto ◽  
Issay Kitabayashi

Abstract Abstract 2525 Poster Board II-502 MOZ (MOnocytic leukemia Zinc finger protein) and MORF (MOz Related Factor), Myst-type histone acetyltransferases, are involved in chromosome translocations associated with FAB-M4/5 subtypes of acute myeloid leukemia. We have reported that MOZ is essential for hematopoietic cell development and self-renewal of hematopoietic stem cells. To explore the possibility MORF also plays important roles in hematopoiesis, we generated Morf-deficient mice with homologous recombination methods. Morf−/− mice were smaller than their wildtype littermates and died within 4 weeks after birth on C57BL/6 background. In MORF−/− fetal liver, Flt3-negative KSL (c-Kit+ Sca-1+ Lineage-) cells containing hematopoietic stem cells were decreased. When fetal liver cells were transplanted into irradiated recipient mice, MORF−/− cells less efficiently reconstituted hematopoiesis than wild-type cells. Additionally, bone marrow cells reconstituted with MORF−/− cells rarely contributed to hematopoiesis in secondary transplants. To reveal relationship between MORF and MOZ in hematopoiesis, we generated double heterozygous (Moz+/− Morf+/−) mouse. Double heterozygous mice were smaller than wild-type littermates and died at least 4 weeks after birth. Numbers of KSL cells, especially Flt3- KSL cells and common myeloid progenitors were decreased in the double heterozygous embryos. The double heterozygous fetal liver cells also displayed less activity to reconstitute hematopoiesis than MOZ+/− or MORF+/− cells. Since MORF−/− mice and MOZ/MORF double heterozygous mice were alive at adult on a mixed C57BL/6/DBA2 genetic background, we investigated adult hematopoiesis in these mice. MORF−/− or MOZ/MORF double heterozygous mice were smaller than their wild-type littermates and had small numbers of thymocytes and splenocytes. However, there were no significant differences in number of bone marrow cells and hematopoietic lineage population in MORF−/− or MOZ/MORF double heterozygous mice. These results suggest that MORF as well as MOZ plays important roles in self-renewal of hematopoietic stem cells. Disclosures: No relevant conflicts of interest to declare.


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.


1984 ◽  
Vol 160 (4) ◽  
pp. 1087-1101 ◽  
Author(s):  
K A Denis ◽  
L J Treiman ◽  
J I St Claire ◽  
O N Witte

Long-term cultures of murine fetal liver have been successfully established using a modification of our in vitro bone marrow culture system (14, 15). Fetal liver cells from midgestation BALB/c embryos were plated onto BAB-14 bone marrow stromal cell-adherent layers. After a 3-5 wk period, cell growth began to increase and these cells were expanded in number on fresh feeder layers. The cultured fetal liver cells were lymphoid in morphology, 5-20% cytoplasmic Ig-positive, but less than 1% surface Ig-positive. Southern blot analysis of the cultured fetal liver cells, as well as cultured bone marrow-derived B cells, demonstrated a population with germline Ig heavy chain loci, possibly representing very early B cell precursors. Abelson murine leukemia virus (A-MuLV) clonal transformants of such cultured fetal liver cells had a phenotypic distribution similar to that seen with fresh fetal liver transformants but distinct from those obtained with the transformation of either cultured or fresh bone marrow. All A-MuLV transformants isolated had rearrangements at the mu heavy chain locus of both chromosomes, irrespective of Ig production. In addition, most mu heavy chain producers had at least one rearranged kappa gene locus. These long-term fetal liver cultures provide large numbers of cells for studying events early in the B lymphocyte lineage. The cultured fetal liver cells retained phenotypic traits similar to fresh fetal liver B cells and distinctive from bone marrow cells cultured under similar conditions.


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 ◽  
2000 ◽  
Vol 95 (7) ◽  
pp. 2284-2288 ◽  
Author(s):  
Hideo Ema ◽  
Hiromitsu Nakauchi

Abstract The activity of hematopoietic stem cells in the developing liver of a C57BL/6 mouse embryo was quantified by a competitive repopulation assay. Different doses of fetal liver cells at days 11 to 18 of gestation were transplanted into irradiated mice together with 2 × 105 adult bone marrow cells. A long-term repopulation in myeloid-, B-cell, and T-cell lineage by fetal liver cells was evaluated at 20 weeks after transplantation. At day 12 of gestation multilineage repopulating activity was first detected in the liver as 50 repopulating units (RU) per liver. The number of RU per liver increased 10-fold and 33-fold by day 14 and day 16 of gestation, and decreased thereafter, suggesting a single wave of stem cell development in the fetal liver. A limiting dilution analysis revealed that the frequency of competitive repopulating units (CRU) in fetal liver cells at day 12 of gestation was similar to that at day 16 of gestation. Because of an increase of total fetal liver cell number, the absolute number of CRU per liver from days 12 to 16 of gestation increased 38-fold. Hence, the mean activity of stem cells (MAS) that is given by RU per CRU remained constant from days 12 to 16 of gestation. From these data we conclude that hematopoietic stem cells expand in the fetal liver maintaining their level of repopulating potential.


1991 ◽  
Vol 174 (5) ◽  
pp. 1283-1286 ◽  
Author(s):  
B Péault ◽  
I L Weissman ◽  
C Baum ◽  
J M McCune ◽  
A Tsukamoto

The search for human hematopoietic stem cells has been hampered by the lack of appropriate assay systems. Demonstration of the ability of precursor cell candidates to give rise to T cells is of significant difficulty since dissociated in vitro cultured thymus stroma cells lose their ability to sustain thymocyte maturation. To define further the differentiative capacities of the rare human fetal liver and bone marrow cells that express the CD34 surface antigen and exhibit in vitro myeloid and pre-B cell activities, we have microinjected them into HLA-mismatched fetal thymus fragments, partially depleted of hematopoietic cells by low temperature culture. In vitro colonized thymuses have then been allowed to develop upon engraftment into immunodeficient SCID mice. Using this modification of the SCID-hu system, we show that low numbers of fetal CD34+ progenitor cells can repopulate the lymphoid compartment in the human thymus.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1269-1269 ◽  
Author(s):  
Lynn S. White ◽  
Deepti Soodgupta ◽  
Rachel L. Johnston ◽  
Jeffrey A. Magee ◽  
Jeffrey J. Bednarski

Abstract Hematopoietic stem cells (HSC) persist throughout life by undergoing extensive self-renewal divisions while maintaining an undifferentiated state. The mechanisms that support HSC self-renewal change throughout the course of development as temporal changes in transcriptional regulators coordinate distinct genetic programs in fetal, post-natal and adult HSCs. These self-renewal programs are often ectopically activated in leukemia cells to drive neoplastic proliferation and high expression of HSC-associated genes predicts a poor prognosis in acute myelogenous leukemia (AML). In this regard, it was recently shown that expression of the transcriptional regulator BCLAF1 (Bcl2 associated transcription factor 1) is increased in AML blasts relative to normal precursor populations and suppression of BCLAF1 causes reduced proliferation and induction of differentiation to a dendritic cell fate. These findings raise the question of whether BCLAF1 may regulate normal as well as neoplastic self-renewal programs. We find that Bclaf1 is highly expressed in HSCs versus committed bone marrow populations consistent with a potential role for this gene in HSC functions. To test whether BCLAF1 regulates HSC development and hematopoiesis, we used germline loss of function mice. Bclaf1-/- mice succumb to pulmonary failure shortly after birth due to poor lung development, so we assessed prenatal hematopoiesis. Bclaf1-deficient mice had significantly reduced HSC and hematopoietic progenitor cell (HPC) frequencies and numbers despite normal fetal liver cellularity. To determine if Bclaf1 is required for HSC function during fetal development, we performed competitive reconstitution assays. Fetal liver cells from Bclaf1+/+or Bclaf1-/-mice were transplanted along with wild-type competitor bone marrow cells into lethally irradiated recipient mice. Compared to recipients of Bclaf1+/+fetal liver cells, recipients of Bclaf1-/-cells had a significantly lower percentage of donor-derived leukocytes at all time points after transplantation as well as reduced percentage of donor HSCs at 16 weeks post-transplant. Notably, all leukocyte populations (B cells, T cells, granulocytes and macrophages) from Bclaf1-/-donors were reduced consistent with an abnormality in HSC repopulating activity rather than a defect in a specific differentiation pathway. Consistent with these findings, Bclaf-deficient cells did not engraft in competitive transplants with limiting numbers of sorted fetal liver HSCs whereas sorted wild-type Bclaf1+/+cells effectively reconstituted hematopoiesis in recipient mice. In addition, Vav-cre:Bclaf1flox/floxmice, which have selective deletion of Bclaf1 in hematopoietic cells, have reduced frequencies and numbers of fetal liver HSCs identical to the findings observed in germline Bclaf1-/-mice. These results show that loss of Bclaf1 leads to defective development and repopulating activity of fetal HSCs. Interestingly, when adult mice are successfully engrafted with Bclaf1-deficient HSCs, the donor HSCs suffer no additional functional impairment. Furthermore, in secondary transplant experiments Bclaf1-deficient HSCs maintain long-term repopulating activity. Thus, Bclaf1 may have distinct functions in fetal versus adult HSC self-renewal. Collectively, our findings reveal Bclaf1 is a novel regulator of fetal HSC function and suggest that it may have distinct functions in different developmental contexts. 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 ◽  
1994 ◽  
Vol 84 (2) ◽  
pp. 433-439 ◽  
Author(s):  
C Richardson ◽  
M Ward ◽  
S Podda ◽  
A Bank

Abstract We have been transducing mouse hematopoietic cells with the human MDR1 (MDR) gene in retroviral vectors to determine the optimal conditions for retroviral gene transfer as a model system for potential human gene therapy. In these studies, we have demonstrated transduction and expression of the human MDR gene using ecotropic and amphotropic MDR- retroviral producer lines. To obtain more mouse hematopoietic cells for detailed study, mouse fetal liver cells (FLC) have been used for MDR transduction and expression, and to reconstitute the ablated marrows of live adult mice. FLC contain hematopoietic cells that have a reconstituting capacity comparable to that of adult mouse bone marrow cells. However, to our surprise, FLC can only be transduced with ecotropic retrovirus and not with amphotropic virus. This restriction of transduction of FLC cannot be overcome by higher titer virus. The resistance to amphotropic transduction by FLC may be part of a changing developmental program that results in a different antigen repertoire on FLC as compared with adult bone marrow cells.


Sign in / Sign up

Export Citation Format

Share Document