scholarly journals Effects of Delta-Like Noncanonical Notch Ligand 1 Expression of Human Fetal Liver Hepatoblasts on Hematopoietic Progenitors

2019 ◽  
Vol 2019 ◽  
pp. 1-8 ◽  
Author(s):  
Jörg C. Gerlach ◽  
Robert L. Thompson ◽  
Bruno Gridelli ◽  
Eva Schmelzer

Although the hepatic and hematopoietic progenitors of the liver are well characterized, the interactions between these two lineages remain mostly elusive. Hepatoblasts express delta-like noncanonical Notch ligand 1 (Dlk1), whose cleaved extracellular domain can become a soluble protein. We assessed the effects of DLK1 gene expression knockdown in cultures of total fetal liver cells. Furthermore, we separated Dlk1+hepatoblasts from the total liver cell fraction and investigated effects of direct cell contact. Dlk1-cells were cultured either without Dlk1+hepatoblasts, in direct contact with hepatoblasts, or separated from hepatoblasts by a porous membrane in inserts to inhibit cell contact but allow free exchange of molecules. Expression of the hepatic and hematopoietic genes, colony forming unit potential of various hematopoietic progenitors, and cell numbers and types were investigated. We found that DLK1 knockdown in total fetal liver cell cultures decreased total cell numbers. The expression of hepatic progenitor genes and mature hematopoietic genes was affected. Hematopoietic BFU-E and CFU-GM colony numbers were reduced significantly. The depletion of Dlk1+hepatoblasts in culture decreased the potential of all hematopoietic progenitors to form colonies of all types and reduced the percentage of mature hematopoietic cells. The addition of hepatoblasts in inserts to Dlk1-cells further decreased the potential to form the CFU-GM and CFU-GEMM colonies and the percentage of mature hematopoietic cells but increased total cell numbers. Conclusively, direct contact of Dlk1 supports hematopoietic progenitor expansion and functionality that cannot be reconstituted in coculture without direct cell contact.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4214-4214
Author(s):  
Feng Ma ◽  
Dan Wang ◽  
Sachiyo Hanada ◽  
Hirohide Kawasaki ◽  
Yuji Zaike ◽  
...  

Abstract Human embryonic stem cells provide a unique tool to study early events occurring in the development of human embryonic hematopoiesis, and their totipotent capability indicates a potent clinical application based on the cellular therapy and the evaluation of drug effects on hematopoietic and blood cells. To achieve efficient production of hematopoietic cells from human embryonic stem cells, we attempted to reproduce the circumstance surrounding embryonic hematopoietic cells in vitro. Since fetal liver is the predominant source of hematopoietic and blood cells in mammalian embryogenesis, we established stromal cells from mouse fetal liver at days 14 to 15 of gestation. In the co-culture of human embryonic stem cells with the established stromal cells, a number of hematopoietic progenitors were generated at around day 14 of co-culture, and this hematopoietic activity was highly enriched in the cobble stone-like cells under the stromal layer. Most of the cobble stone-like cells collected expressed CD34 and contained a variety of hematopoietic colony-forming cells, especially multilineage colony-forming cells, at a high frequency. The multipotential hematopoietic progenitors in the cobble stone-like cells produced all types of mature blood cells, including adult type hemoglobin-synthesizing erythrocytes and tryptase and chymase-bouble positive mast cells in the suspension cultiue with a cytokine cocktail. The developed co-culture system of human embryonic stem cells should offer a novel source for hematopoietic and blood cells applicable to cellular therapies and drug screening.


Blood ◽  
1998 ◽  
Vol 91 (4) ◽  
pp. 1263-1271 ◽  
Author(s):  
Takashi Tsuji ◽  
Iwao Waga ◽  
Katsunari Tezuka ◽  
Masafumi Kamada ◽  
Kimio Yatsunami ◽  
...  

Abstract Cellular interactions between hematopoietic cells and stromal cells play important roles in the proliferation and differentiation of hematopoietic cells. The proliferation of a human erythroleukemia cell line, HEL cells, which can differentiate into macrophage- and megakaryocyte-like cells, and erythroid precursors was dramatically induced on coculture with a hematopoietic-supportive stromal cell line, HESS-5 cells, which can support long-term hematopoiesis in vitro without fetal bovine serum. HEL cells proliferated when they were cocultured with but not without direct cell contact. Because the coculture supernatants with direct cell contact and cytokines such as interleukins and growth factors did not exhibit growth-stimulating activity toward HEL cells, it was suggested that some molecule that has growth-stimulating activity exists on the surface of the cells. Extracellular matrix components such as fibronectin, laminin, vitronectin, and collagen did not affect the proliferation of HEL cells. An anti-CD18 monoclonal antibody, which recognizes the common β chain of the β2 integrin subfamily, induced dramatic proliferation of HEL cells. Moreover, the proliferation of HEL cells was inhibited by an antisense oligonucleotide of CD18 mRNA. As judged from these observations, the proliferation of HEL cells was mediated by CD18 molecules expressed on HEL cells. On the contrary, the common counter-receptor of the β2 integrin subfamily, intercellular adhesion molecule-1, which is expressed on CHO-K1 cells, did not stimulate the growth of HEL cells. It is known that other counter molecules of the β2 integrin subfamily, such as complement C3bi and fibrinogen, are not produced by stromal cells. These findings suggest that the proliferation of HEL cells may be induced through an interaction between a novel molecule of the β2 integrin subfamily on HEL cells and the counter-receptor on HESS-5 cells. The β2 integrin subfamily may regulate the growth of hematopoietic cells in hematopoiesis in vivo and/or cause the abnormal growth of leukemia cells.


Development ◽  
2002 ◽  
Vol 129 (8) ◽  
pp. 2003-2013 ◽  
Author(s):  
Maria Teresa Mitjavila-Garcia ◽  
Michel Cailleret ◽  
Isabelle Godin ◽  
Maria Manuela Nogueira ◽  
Karine Cohen-Solal ◽  
...  

In this study, we have characterized the early steps of hematopoiesis during embryonic stem cell differentiation. The immunophenotype of hematopoietic progenitor cells derived from murine embryonic stem cells was determined using a panel of monoclonal antibodies specific for hematopoietic differentiation antigens. Surprisingly, the CD41 antigen (αIIb integrin, platelet GPIIb), essentially considered to be restricted to megakaryocytes, was found on a large proportion of cells within embryoid bodies although very few megakaryocytes were detected. In clonogenic assays, more than 80% of all progenitors (megakaryocytic, granulo-macrophagic, erythroid and pluripotent) derived from embryoid bodies expressed the CD41 antigen. CD41 was the most reliable marker of early steps of hematopoiesis. However, CD41 remained a differentiation marker because some CD41– cells from embryoid bodies converted to CD41+ hematopoietic progenitors, whereas the inverse switch was not observed. Immunoprecipitation and western blot analysis confirmed that CD41 was present in cells from embryoid bodies associated with CD61 (β3 integrin, platelet GPIIIa) in a complex. Analysis of CD41 expression during ontogeny revealed that most yolk sac and aorta-gonad-mesonephros hematopoietic progenitor cells were also CD41+, whereas only a minority of bone marrow and fetal liver hematopoietic progenitors expressed this antigen. Differences in CD34 expression were also observed: hematopoietic progenitor cells from embryoid bodies, yolk sac and aorta-gonad-mesonephros displayed variable levels of CD34, whereas more than 90% of fetal liver and bone marrow progenitor cells were CD34+. Thus, these results demonstrate that expression of CD41 is associated with early stages of hematopoiesis and is highly regulated during hematopoietic development. Further studies concerning the adhesive properties of hematopoietic cells are required to assess the biological significance of these developmental changes.


Blood ◽  
2000 ◽  
Vol 96 (3) ◽  
pp. 950-957 ◽  
Author(s):  
Schickwann Tsai ◽  
Jutta Fero ◽  
Steve Bartelmez

To study the regulation of the early stages of hematopoiesis, cDNA representational difference analysis was used to isolate genes that were differentially expressed in primitive hematopoietic progenitors. The reasoning was that such genes were more likely to provide functions important to hematopoietic stem cells and progenitors. One of the genes identified through this approach encodes mouse Jagged2(mJagged2). Using quantitative reverse transcription–polymerase chain reaction, it was shown that mJagged2 was differentially expressed in c-kit+ hematopoietic progenitors, including those with the phenotypes of Lin− c-kit+Rhlo Holo and Lin−c-kit+ Rhhi Holo, and that they have been shown to be highly enriched for long-term and short-term repopulating hematopoietic stem cells, respectively. Western blot analyses showed that endothelial cells also expressed high levels of Jagged2, but stromal fibroblasts did not. Using a coculture system we found that exogenous, full-length mJagged2 promoted the survival and proliferation of hematopoietic progenitors, including the high-proliferative potential colony-forming cells. Direct cell-to-cell contact was required for this effect. Taken together, these findings indicate that both c-kit+ hematopoietic progenitors and endothelial cells express Jagged2 and that exogenous, full-length Jagged2 promotes the survival and proliferation of hematopoietic progenitors.


Blood ◽  
1998 ◽  
Vol 92 (3) ◽  
pp. 834-841 ◽  
Author(s):  
Huijie Jiang ◽  
Kenkichi Sugimoto ◽  
Hitoshi Sawada ◽  
Emi Takashita ◽  
Maki Tohma ◽  
...  

Abstract A stroma-dependent cell line (HB-1) was established from myelogenous leukemic cells of CBA/N mouse. Characterization of the cells showed that HB-1 proliferated on hematopoietic supportive stromal cells (MS-10), but did not survive or proliferate on hematopoietic nonsupportive cells (MS-K). Direct contact between HB-1 and MS-10 appears to be necessary for HB-1 to proliferate on MS-10. We found that interleukin-1α (IL-1α) produced by MS-10 plays a major role in the survival and proliferation of HB-1. IL-11 did not support the proliferation of HB-1 cells by itself, but enhanced the proliferation of HB-1 cells in the presence of IL-1α. The expression of IL-1α and IL-11 was induced in MS-10 by the direct contact with HB-1 cells, and the expression of IL-1 receptor type I (IL-1RI) and interleukin-11 receptor (IL-11R) was induced in HB-1 cells by the attachment of the cells to MS-10. These findings show the existence of two-way interactions between HB-1 and MS-10. © 1998 by The American Society of Hematology.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1559-1559
Author(s):  
Akanksha Chhabra ◽  
Andrew J Lechner ◽  
Asha Acharya ◽  
Masaya Ueno ◽  
Ben Van Handel ◽  
...  

Abstract Abstract 1559 The goals of hematopoiesis during embryogenesis are two-fold, to rapidly produce red blood cells to support the survival and development of the embryo, and to establish a pool of undifferentiated hematopoietic stem cells (HSC) for postnatal life. These goals are achieved by segregation of fetal hematopoiesis in multiple waves that are executed in distinct anatomical sites and microenvironmental niches. However, the microenvironmental cues that promote “stemness” vs. differentiation remain poorly understood. The placenta is a recently discovered hematopoietic organ that supports HSC generation and expansion without promoting their differentiation. The placental HSC pool is thought to reside in the placental labyrinth, which is comprised of an intricate vascular network surrounded by trophoblasts. So far, it has been unknown how a disruption of the integrity of the placental vascular labyrinth affects fetal hematopoiesis. The structure of the placental labyrinth is compromised in embryos that lack components of PDGF-B signaling. PDGF-B-/- and PDGF-Rβ-/- (receptor for PDGF-B) embryos display dilation of fetal blood vessels and reduction of trophoblast cells in the placental labyrinth. Our studies revealed that loss of PDGF-B signaling alters the unique placental hematopoietic microenvironment, resulting in active erythropoiesis in the placenta. The unexpected erythropoiesis in the placenta exhibited the same hallmarks of normal definitive erythropoiesis observed in the fetal liver as confirmed by flow cytometry for erythroid markers, morphological analysis and association of erythroblasts with macrophages. Interestingly, deletion of PDGF-Rβ in hematopoietic cells by using a Tie2-Cre strain did not induce the differentiation of placental hematopoietic progenitors into erythroid cells, implying that the ectopic definitive erythropoiesis results from the lack of PDGF-B signaling in the placental microenvironment rather than in the hematopoietic cells themselves. Our studies revealed that the erythroid differentiation in placentas of PDGF-B-/- embryos was induced by marked upregulation of Epo in placental trophoblasts. Strikingly, lentiviral overexpression of Epo specifically in the placental trophoblasts was sufficient to convert the placenta into an ectopic erythropoietic organ. These data reveal a critical function of PDGF-B signaling in protecting the integrity of the placental microenvironment that is required for preventing hematopoietic progenitors from differentiation during their residence in the placenta. Furthermore, these studies highlight the placenta as a versatile hematopoietic organ that supports HSC development during normal pregnancy but can be recruited as a site for definitive erythropoiesis during pathological conditions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2062-2062
Author(s):  
Juraj Kokavec ◽  
Romit Majumdar ◽  
Martina Kapalova ◽  
Nikola Curik ◽  
Filipp Savvulidi ◽  
...  

Abstract Abstract 2062 Chromatin remodeling plays an important role during normal and malignant hematopoiesis. The chromatin remodeling associated Imitation Switch (ISWI class) ATPase Smarca5 is abundantly expressed in mammalian hematopoietic cells. Smarca5 levels are downregulated during erythroid terminal differentiation (Stopka 2000) and they are significantly increased in human acute myelogenous leukemia blasts (Stopka 2000). In addition, Smarca5 has been reported to interact with transcription factor GATA-1 (Rodriguez, 2005). Our previous studies showed that homozygous inactivation of Smarca5 leads to early embryonic lethality (Stopka 2003). To study the role of Smarca5 in hematopoiesis, we generated a conditional allele (cKO) of Smarca5 by introducing loxP sites around exon 5 in mouse ES cells and transmitted this allele to the mouse germline. Deletion of exon 5 mediated by Cre-recombinase leads to complete loss of production of Smarca5 protein. Deletion of Smarca5 in definitive hematopoietic progenitors using the Vav1-iCre transgene resulted in fetal anemia and death by E18. The block in erythropoiesis is characterized by accumulation of hematopoietic progenitors (Sca-1+) and progressive decrease of terminally differentiated Ter-119+ and Ter-119+ Cd71+ cells in mutant fetal livers. Colony forming assays indicate growth and differentiation defects in mutant cells compared to wild type littermates. Gene expression analyses of mutant fetal liver-derived hematopoietic cells indicate loss of expression of Gata1 and several of its gene targets including adult globins. Gata2 expression was unchanged. In turn, embryonic globins Ey and Zeta were significantly upregulated (~15 fold) in mutant fetal liver-derived hematopoietic cells, indicating a defect in hemoglobin switching. To summarize, our results show that Smarca5 plays important roles in erythroid differentiation and globin gene regulation. (Grant # IGA 10310-3, MSMT 2B06077, 0021620806, LC06044, SVV-2010-254260507, NIH R01CA154239). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 92 (3) ◽  
pp. 834-841 ◽  
Author(s):  
Huijie Jiang ◽  
Kenkichi Sugimoto ◽  
Hitoshi Sawada ◽  
Emi Takashita ◽  
Maki Tohma ◽  
...  

A stroma-dependent cell line (HB-1) was established from myelogenous leukemic cells of CBA/N mouse. Characterization of the cells showed that HB-1 proliferated on hematopoietic supportive stromal cells (MS-10), but did not survive or proliferate on hematopoietic nonsupportive cells (MS-K). Direct contact between HB-1 and MS-10 appears to be necessary for HB-1 to proliferate on MS-10. We found that interleukin-1α (IL-1α) produced by MS-10 plays a major role in the survival and proliferation of HB-1. IL-11 did not support the proliferation of HB-1 cells by itself, but enhanced the proliferation of HB-1 cells in the presence of IL-1α. The expression of IL-1α and IL-11 was induced in MS-10 by the direct contact with HB-1 cells, and the expression of IL-1 receptor type I (IL-1RI) and interleukin-11 receptor (IL-11R) was induced in HB-1 cells by the attachment of the cells to MS-10. These findings show the existence of two-way interactions between HB-1 and MS-10. © 1998 by The American Society of Hematology.


1982 ◽  
Vol 55 (1) ◽  
pp. 211-231
Author(s):  
I. Olsen ◽  
M.F. Dean ◽  
H. Muir ◽  
G. Harris

Fibroblasts deficient in beta-glucuronidase acquired high levels of this enzyme when they were co-cultured with concanavalin A-stimulated lymphocytes. Acquired enzyme activity, determined using a single-cell cytochemical assay, was directly proportional to the number of lymphocytes added and persisted for several days in fibroblasts maintained at high density. Lymphocytes did not secret significant levels of beta-glucuronidase into their culture medium, and did not release other substances able to induce synthesis of the enzyme by the deficient fibroblasts. Nor did beta-glucuronidase acquisition result from concanavalin A-mediated uptake of enzyme, since alpha-methylmannoside did not reduce acquired activity. Moreover, lymphocytes from various sources, whether unstimulated or activated by a different mitogen, bacterial lipopolysaccharide, were equally effective in promoting the appearance of beta-glucuronidase. Deficient fibroblasts did not acquire beta-glucuronidase by active endocytosis when co-cultured with lymphocytes, since enzyme extracted from lymphocytes was not itself effective in this respect. Furthermore, mannose 6-phosphate, which did inhibit, endocytosis by deficient fibroblasts of exogenous beta-glucuronidase prepared from 3T3 cells, had no effect on enzyme acquisition by fibroblasts during their co-culture with lymphocytes. Conversely, inhibitors of protein synthesis and energy metabolism, which did not interfere with endocytosis of exogenous enzyme, abolished the acquisition of beta-glucuronidase during co-culture. Deficient fibroblasts did not acquire beta-glucuronidase when they were cultured together with lymphocytes but separated from them by Millipore membranes permeable to exogenous enzyme. Thus, although the mechanism of acquisition is still unclear, the present results suggest that beta-glucuronidase is transferred from lymphocytes to deficient fibroblasts by a process in which direct cell-to-cell contact is obligatory.


2001 ◽  
Vol 280 (4) ◽  
pp. G720-G728 ◽  
Author(s):  
Kasper H. N. Hoebe ◽  
Renger F. Witkamp ◽  
Johanna Fink-Gremmels ◽  
Adelbert S. J. P. A. M. Van Miert ◽  
Mario Monshouwer

This study focuses on the importance of direct contact between Kupffer cells (KCs) and hepatocytes (HCs) during the hepatic inflammatory response using an in vitro approach. The lipopolysaccharide (LPS)-induced inflammatory response in monocultures of porcine HCs and KCs were compared with cocultures prepared either with direct contact between KCs and HCs (DC cocultures) or without direct contact using cell culture membrane inserts. Our data show that DC cocultures exhibited the highest production of tumor necrosis factor (TNF)-α, interleukin-6, and nitric oxide (NO) compared with the other cultures. Immunohistochemical studies revealed that TNF-α was exclusively produced by KCs, whereas HCs were responsible for NO production after LPS stimulation. Biotransformation capacity, as determined by cytochrome P-450 and UDP glucuronosyl transferase enzyme activities, was most significantly decreased in DC cocultures. These results provide evidence that direct contact between KCs and HCs favors the extensive TNF-α production by KCs but in turn affects HC functionality and viability. These findings suggest that direct contact between KCs and HCs plays a key role in the development of a fulminating hepatic inflammatory response.


Sign in / Sign up

Export Citation Format

Share Document