Skip to main content
Top
Published in: Experimental Hematology & Oncology 1/2017

Open Access 01-12-2017 | Research

Loss of quiescence and self-renewal capacity of hematopoietic stem cell in an in vitro leukemic niche

Authors: Natalia-Del Pilar Vanegas, Jean-Paul Vernot

Published in: Experimental Hematology & Oncology | Issue 1/2017

Login to get access

Abstract

Background

Leukemic and mesenchymal stem cells interact in the leukemic microenvironment and affect each other differently. This interplay has also important implications for the hematopoietic stem cell (HSC) biology and function. This study evaluated human HSC self-renewal potential and quiescence in an in vitro leukemic niche without leukemic cells.

Methods

A leukemic niche was established by co-culturing mesenchymal stem cells with a fresh conditioned medium obtained from a leukemic (REH) cell line. After 3 days, the REH-conditioned medium was removed and freshly isolated CD34+ at a density of up to 100,000 cells/ml were added to the leukemic niche. CD34+ cell evaluations (cell cycle, self-renewal gene expression and migration capacity) were performed after 3 further days of co-culture. Additionally, we preliminary investigated the soluble factors present in the leukemic niche and their effect on the mesenchymal stem cells. Statistical significance was assessed by Student’s t test or the nonparametric test Kolmogorov–Smirnov.

Results

By co-culturing normal mesenchymal stem cells with the REH-conditioned medium we showed that hematopoietic stem cells, normally in a quiescent state, enter cell cycle and proliferate. This loss of quiescence was accompanied by an increased expression of Ki-67 and c-Myc, two well-known cell proliferation-associated markers. Two central regulators of quiescence GATA2 and p53 were also down regulated. Importantly, two genes involved in HSC self-renewal, Klf4 and the histone–lysine N-methyltransferase enzyme Ezh2, were severely affected. On the contrary, c-Kit expression, the stem cell factor receptor, was upregulated in hematopoietic stem cells when compared to the normal niche. Interestingly, mesenchymal stem cells incubated with the REH-conditioned medium stopped growing, showed a flattened morphology with the appearance of small vacuoles, and importantly, became positive for the senescence-associated beta-galactosidase activity. Evaluation of the leukemic-conditioned medium showed increased IL-6 and IL-8, suggesting that these cytokines could be responsible for the observed changes.

Conclusions

Our results showed that quiescence and self-renewal are severely affected in this leukemic niche. This in vitro leukemic niche, established without leukemic cells, will facilitate HSC gene expression evaluation and the development of therapeutic agents aimed to neutralize soluble factors and the cell signaling pathways involved in HSC alterations.
Appendix
Available only for authorised users
Literature
4.
go back to reference Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978;4(1–2):7–25.PubMed Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978;4(1–2):7–25.PubMed
5.
go back to reference Hao S, Chen C, Cheng T. Cell cycle regulation of hematopoietic stem or progenitor cells. Int J Hematol. 2016;103(5):487–97.CrossRefPubMed Hao S, Chen C, Cheng T. Cell cycle regulation of hematopoietic stem or progenitor cells. Int J Hematol. 2016;103(5):487–97.CrossRefPubMed
6.
go back to reference Wang Z, Ema H. Mechanisms of self-renewal in hematopoietic stem cells. Int J Hematol. 2016;103(5):498–509.CrossRefPubMed Wang Z, Ema H. Mechanisms of self-renewal in hematopoietic stem cells. Int J Hematol. 2016;103(5):498–509.CrossRefPubMed
7.
10.
11.
go back to reference Bradford GB, et al. Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp Hematol. 1997;25(5):445–53.PubMed Bradford GB, et al. Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp Hematol. 1997;25(5):445–53.PubMed
12.
go back to reference Passegue E. Hematopoietic stem cells, leukemic stem cells and chronic myelogenous leukemia. Cell Cycle. 2005;4(2):266–8.CrossRefPubMed Passegue E. Hematopoietic stem cells, leukemic stem cells and chronic myelogenous leukemia. Cell Cycle. 2005;4(2):266–8.CrossRefPubMed
13.
go back to reference Yamaguchi M, et al. Different adhesive characteristics and VLA-4 expression of CD34(+) progenitors in G0/G1 versus S+ G2/M phases of the cell cycle. Blood. 1998;92(3):842–8.PubMed Yamaguchi M, et al. Different adhesive characteristics and VLA-4 expression of CD34(+) progenitors in G0/G1 versus S+ G2/M phases of the cell cycle. Blood. 1998;92(3):842–8.PubMed
14.
go back to reference Perdomo-Arciniegas AM, Vernot JP. Co-culture of hematopoietic stem cells with mesenchymal stem cells increases VCAM-1-dependent migration of primitive hematopoietic stem cells. Int J Hematol. 2011;94(6):525–32.CrossRefPubMed Perdomo-Arciniegas AM, Vernot JP. Co-culture of hematopoietic stem cells with mesenchymal stem cells increases VCAM-1-dependent migration of primitive hematopoietic stem cells. Int J Hematol. 2011;94(6):525–32.CrossRefPubMed
15.
go back to reference Cheung TH, Rando TA. Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol. 2013;14(6):329–40.CrossRefPubMed Cheung TH, Rando TA. Molecular regulation of stem cell quiescence. Nat Rev Mol Cell Biol. 2013;14(6):329–40.CrossRefPubMed
16.
go back to reference Arai F, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118(2):149–61.CrossRefPubMed Arai F, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118(2):149–61.CrossRefPubMed
17.
go back to reference Yoshihara H, et al. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell. 2007;1(6):685–97.CrossRefPubMed Yoshihara H, et al. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell. 2007;1(6):685–97.CrossRefPubMed
18.
go back to reference Thoren LA, et al. Kit regulates maintenance of quiescent hematopoietic stem cells. J Immunol. 2008;180(4):2045–53.CrossRefPubMed Thoren LA, et al. Kit regulates maintenance of quiescent hematopoietic stem cells. J Immunol. 2008;180(4):2045–53.CrossRefPubMed
20.
go back to reference Cashman J, et al. Stromal-derived factor 1 inhibits the cycling of very primitive human hematopoietic cells in vitro and in NOD/SCID mice. Blood. 2002;99(3):792–9.CrossRefPubMed Cashman J, et al. Stromal-derived factor 1 inhibits the cycling of very primitive human hematopoietic cells in vitro and in NOD/SCID mice. Blood. 2002;99(3):792–9.CrossRefPubMed
21.
go back to reference Wilson A, Laurenti E, Trumpp A. Balancing dormant and self-renewing hematopoietic stem cells. Curr Opin Genet Dev. 2009;19(5):461–8.CrossRefPubMed Wilson A, Laurenti E, Trumpp A. Balancing dormant and self-renewing hematopoietic stem cells. Curr Opin Genet Dev. 2009;19(5):461–8.CrossRefPubMed
22.
go back to reference Trumpp A, Essers M, Wilson A. Awakening dormant haematopoietic stem cells. Nat Rev Immunol. 2010;10(3):201–9.CrossRefPubMed Trumpp A, Essers M, Wilson A. Awakening dormant haematopoietic stem cells. Nat Rev Immunol. 2010;10(3):201–9.CrossRefPubMed
23.
24.
25.
go back to reference Shah N, Oseth L, LeBien TW. Development of a model for evaluating the interaction between human pre-B acute lymphoblastic leukemic cells and the bone marrow stromal cell microenvironment. Blood. 1998;92(10):3817–28.PubMed Shah N, Oseth L, LeBien TW. Development of a model for evaluating the interaction between human pre-B acute lymphoblastic leukemic cells and the bone marrow stromal cell microenvironment. Blood. 1998;92(10):3817–28.PubMed
26.
go back to reference Douer D, et al. T-cell acute lymphoblastic leukemia with severe leukopenia: evidence for suppression of myeloid progenitor cells by leukemic blasts. Acta Haematol. 1988;80(4):185–9.CrossRefPubMed Douer D, et al. T-cell acute lymphoblastic leukemia with severe leukopenia: evidence for suppression of myeloid progenitor cells by leukemic blasts. Acta Haematol. 1988;80(4):185–9.CrossRefPubMed
27.
go back to reference Duan CW, et al. Leukemia propagating cells rebuild an evolving niche in response to therapy. Cancer Cell. 2014;25(6):778–93.CrossRefPubMed Duan CW, et al. Leukemia propagating cells rebuild an evolving niche in response to therapy. Cancer Cell. 2014;25(6):778–93.CrossRefPubMed
28.
go back to reference Colmone A, et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science. 2008;322(5909):1861–5.CrossRefPubMed Colmone A, et al. Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells. Science. 2008;322(5909):1861–5.CrossRefPubMed
29.
go back to reference Khalid S, et al. Retrospective review of pediatric patients with acute lymphoblastic leukemia: a single center experience. Indian J Pathol Microbiol. 2010;53(4):704–10.CrossRefPubMed Khalid S, et al. Retrospective review of pediatric patients with acute lymphoblastic leukemia: a single center experience. Indian J Pathol Microbiol. 2010;53(4):704–10.CrossRefPubMed
30.
go back to reference Dominici M, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–7.CrossRefPubMed Dominici M, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315–7.CrossRefPubMed
31.
go back to reference Rodriguez-Pardo VM, Vernot JP. Mesenchymal stem cells promote a primitive phenotype CD34+ c-kit+ in human cord blood-derived hematopoietic stem cells during ex vivo expansion. Cell Mol Biol Lett. 2013;18(1):11–33.CrossRefPubMed Rodriguez-Pardo VM, Vernot JP. Mesenchymal stem cells promote a primitive phenotype CD34+ c-kit+ in human cord blood-derived hematopoietic stem cells during ex vivo expansion. Cell Mol Biol Lett. 2013;18(1):11–33.CrossRefPubMed
32.
go back to reference Ehninger A, et al. Posttranscriptional regulation of c-Myc expression in adult murine HSCs during homeostasis and interferon-alpha-induced stress response. Blood. 2014;123(25):3909–13.CrossRefPubMed Ehninger A, et al. Posttranscriptional regulation of c-Myc expression in adult murine HSCs during homeostasis and interferon-alpha-induced stress response. Blood. 2014;123(25):3909–13.CrossRefPubMed
33.
go back to reference Tsai FY, Orkin SH. Transcription factor GATA-2 is required for proliferation/survival of early hematopoietic cells and mast cell formation, but not for erythroid and myeloid terminal differentiation. Blood. 1997;89(10):3636–43.PubMed Tsai FY, Orkin SH. Transcription factor GATA-2 is required for proliferation/survival of early hematopoietic cells and mast cell formation, but not for erythroid and myeloid terminal differentiation. Blood. 1997;89(10):3636–43.PubMed
34.
go back to reference Zon LI. Intrinsic and extrinsic control of haematopoietic stem-cell self-renewal. Nature. 2008;453(7193):306–13.CrossRefPubMed Zon LI. Intrinsic and extrinsic control of haematopoietic stem-cell self-renewal. Nature. 2008;453(7193):306–13.CrossRefPubMed
35.
go back to reference Miyamoto K, et al. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell. 2007;1(1):101–12.CrossRefPubMed Miyamoto K, et al. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell. 2007;1(1):101–12.CrossRefPubMed
36.
go back to reference Argiropoulos B, Humphries RK. Hox genes in hematopoiesis and leukemogenesis. Oncogene. 2007;26(47):6766–76.CrossRefPubMed Argiropoulos B, Humphries RK. Hox genes in hematopoiesis and leukemogenesis. Oncogene. 2007;26(47):6766–76.CrossRefPubMed
38.
go back to reference Imperato MR, et al. The RUNX1-PU.1 axis in the control of hematopoiesis. Int J Hematol. 2015;101(4):319–29.CrossRefPubMed Imperato MR, et al. The RUNX1-PU.1 axis in the control of hematopoiesis. Int J Hematol. 2015;101(4):319–29.CrossRefPubMed
39.
go back to reference Lataillade JJ, et al. Chemokine SDF-1 enhances circulating CD34(+) cell proliferation in synergy with cytokines: possible role in progenitor survival. Blood. 2000;95(3):756–68.PubMed Lataillade JJ, et al. Chemokine SDF-1 enhances circulating CD34(+) cell proliferation in synergy with cytokines: possible role in progenitor survival. Blood. 2000;95(3):756–68.PubMed
40.
go back to reference Lasry A, Ben-Neriah Y. Senescence-associated inflammatory responses: aging and cancer perspectives. Trends Immunol. 2015;36(4):217–28.CrossRefPubMed Lasry A, Ben-Neriah Y. Senescence-associated inflammatory responses: aging and cancer perspectives. Trends Immunol. 2015;36(4):217–28.CrossRefPubMed
42.
go back to reference Tipping AJ, et al. High GATA-2 expression inhibits human hematopoietic stem and progenitor cell function by effects on cell cycle. Blood. 2009;113(12):2661–72.CrossRefPubMed Tipping AJ, et al. High GATA-2 expression inhibits human hematopoietic stem and progenitor cell function by effects on cell cycle. Blood. 2009;113(12):2661–72.CrossRefPubMed
44.
go back to reference Foudi A, et al. Analysis of histone 2B-GFP retention reveals slowly cycling hematopoietic stem cells. Nat Biotechnol. 2009;27(1):84–90.CrossRefPubMed Foudi A, et al. Analysis of histone 2B-GFP retention reveals slowly cycling hematopoietic stem cells. Nat Biotechnol. 2009;27(1):84–90.CrossRefPubMed
45.
go back to reference Wilson A, et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 2008;135(6):1118–29.CrossRefPubMed Wilson A, et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 2008;135(6):1118–29.CrossRefPubMed
47.
go back to reference Su IH, et al. Polycomb group protein ezh2 controls actin polymerization and cell signaling. Cell. 2005;121(3):425–36.CrossRefPubMed Su IH, et al. Polycomb group protein ezh2 controls actin polymerization and cell signaling. Cell. 2005;121(3):425–36.CrossRefPubMed
48.
go back to reference Grossmann V, et al. EZH2 mutations and their association with PICALM-MLLT10 positive acute leukaemia. Br J Haematol. 2012;157(3):387–90.CrossRefPubMed Grossmann V, et al. EZH2 mutations and their association with PICALM-MLLT10 positive acute leukaemia. Br J Haematol. 2012;157(3):387–90.CrossRefPubMed
49.
go back to reference Morris VA, et al. Deregulated KLF4 expression in myeloid leukemias alters cell proliferation and differentiation through microRNA and gene targets. Mol Cell Biol. 2015;36(4):559–73.CrossRefPubMed Morris VA, et al. Deregulated KLF4 expression in myeloid leukemias alters cell proliferation and differentiation through microRNA and gene targets. Mol Cell Biol. 2015;36(4):559–73.CrossRefPubMed
50.
go back to reference Keller JR, Ortiz M, Ruscetti FW. Steel factor (c-kit ligand) promotes the survival of hematopoietic stem/progenitor cells in the absence of cell division. Blood. 1995;86(5):1757–64.PubMed Keller JR, Ortiz M, Ruscetti FW. Steel factor (c-kit ligand) promotes the survival of hematopoietic stem/progenitor cells in the absence of cell division. Blood. 1995;86(5):1757–64.PubMed
51.
go back to reference Kimura Y, et al. c-Kit-mediated functional positioning of stem cells to their niches is essential for maintenance and regeneration of adult hematopoiesis. PLoS ONE. 2011;6(10):e26918.CrossRefPubMedPubMedCentral Kimura Y, et al. c-Kit-mediated functional positioning of stem cells to their niches is essential for maintenance and regeneration of adult hematopoiesis. PLoS ONE. 2011;6(10):e26918.CrossRefPubMedPubMedCentral
52.
go back to reference Yoon JY, et al. Association of interleukin-6 and interleukin-8 with poor prognosis in elderly patients with chronic lymphocytic leukemia. Leuk Lymphoma. 2012;53(9):1735–42.CrossRefPubMed Yoon JY, et al. Association of interleukin-6 and interleukin-8 with poor prognosis in elderly patients with chronic lymphocytic leukemia. Leuk Lymphoma. 2012;53(9):1735–42.CrossRefPubMed
53.
go back to reference Lu K, et al. The STAT3 inhibitor WP1066 reverses the resistance of chronic lymphocytic leukemia cells to histone deacetylase inhibitors induced by interleukin-6. Cancer Lett. 2015;359(2):250–8.CrossRefPubMed Lu K, et al. The STAT3 inhibitor WP1066 reverses the resistance of chronic lymphocytic leukemia cells to histone deacetylase inhibitors induced by interleukin-6. Cancer Lett. 2015;359(2):250–8.CrossRefPubMed
Metadata
Title
Loss of quiescence and self-renewal capacity of hematopoietic stem cell in an in vitro leukemic niche
Authors
Natalia-Del Pilar Vanegas
Jean-Paul Vernot
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Experimental Hematology & Oncology / Issue 1/2017
Electronic ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-016-0062-1

Other articles of this Issue 1/2017

Experimental Hematology & Oncology 1/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine