Open Access

Co-culture with podoplanin+ cells protects leukemic blast cells with leukemia-associated antigens in the tumor microenvironment

  • Authors:
    • Ji Yoon Lee
    • A‑Reum Han
    • Sung‑Eun Lee
    • Woo‑Sung Min
    • Hee‑Je Kim
  • View Affiliations

  • Published online on: March 18, 2016     https://doi.org/10.3892/mmr.2016.5009
  • Pages: 3849-3857
  • Copyright: © Lee et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Podoplanin+ cells are indispensable in the tumor microenvironment. Increasing evidence suggests that podoplanin may support the growth and metastasis of solid tumors; however, to the best of our knowledge no studies have determined whether or not podoplanin serves a supportive role in acute myeloid leukemia (AML). The effects of co‑culture with podoplanin+ cells on the cellular activities of the leukemic cells, such as apoptosis and cell proliferation, in addition to the expression of podoplanin in leukemic cells, were investigated. Due to the fact that genetic abnormalities are the primary cause of leukemogenesis, the overexpression of the fibromyalgia‑like tyrosine kinase‑3 gene in colony forming units was also examined following cell sorting. Podoplanin+ cells were found to play a protective role against apoptosis in leukemic cells and to promote cell proliferation. Tumor‑associated antigens, including Wilms' tumor gene 1 and survivin, were increased when leukemic cells were co‑cultured with podoplanin+ cells. In combination, the present results also suggest that podoplanin+ cells can function as stromal cells for blast cell retention in the AML tumor microenvironment.

Introduction

Relapsed acute myeloid leukemia (AML) is considered to be the result of leukemic stem cell (LSC) survival following chemotherapy (1). AML is a heterogeneous clonal disorder, characterized by the accumulation of immature myeloblasts (2). Malignant cell proliferation is maintained by a small fraction of LSCs, and similar to normal hematopoietic stem cells (HSCs), LSCs exhibit certain stem cell properties, including self-renewal, differentiation capacity and expression of cell surface phenotype CD34+CD38 markers (3). LSCs also predominantly produce colony-forming units (CFUs) in vitro, indicating their potential for full differentiation (4). The CFU assay is traditionally used for the detection of hematopoietic progenitor cells (HPCs) in the blood (5). Despite the differences in CFU formation between leukemic and normal progenitor cells, it remains unclear whether colonies are derived from normal HPCs or LSCs/HPCs expressing leukemia-associated genes.

Bone marrow (BM) microenvironments and stem/progenitor cells communicate in order to sustain drug resistance or differentiate into cell lineages; therefore understanding the stromal condition against leukemic cells expressing abnormal genes is required for the development of advanced therapeutic strategies to prevent relapse. Since 1863, when Rudolf Virchow highlighted the importance of the tumor microenviroment for cell growth (6), studies have supported the existence of an association between tumor cell fate and the microenvironment (68).

Podoplanin, a 38 kDa integral membrane mucoprotein, predominantly expressed in the lymphatic capillaries, has been identified to be involved in tumor progression, epithelial-to-mesenchymal transition and lymphatic function (9,10). Its expression has also been observed in intratumoral stromal cells, which can function as normal stromal cells (11,12). Previous studies have been demonstrated that podoplanin is a potent cancer-associated factor in the microenvironments of various tumor types (11,13,14). Podoplanin has been identified to be expressed in osteoblasts and osteocytes in normal bone tissue, and highly expressed in mesenchymal stromal cells, the main component of the BM microenvironment, under conditions of abundant vascular endothelial growth factor C (14,15). Despite the fact that the role of podoplanin in tumor development has been extensively studied (1618), the role of podoplanin+ cells as tumor microenvironmental factors in leukemia remains to be fully elucidated.

The present study examined the role of podoplanin+ cells in leukemia, in addition to investigating its protective role against apoptosis in leukemic blasts, which are enriched by the fibromyalgia-like tyrosine kinase-3 (FLT3) gene. These present study aimed to provide insight into the role of podoplanin as a tumor microenvironmental factor, and contribute to the development of targeted therapies.

Materials and methods

Human primary cells and cell lines

All experiments were approved by the Institutional Review Board of the Human Research at the Catholic University of Korea (Seoul, South Korea). A total of 12 AML blood samples were obtained from patients admitted to the Catholic Blood and Marrow Transplantation Center at Seoul St. Mary's Hospital (Seoul, South Korea). The patients were diagnosed with various subtypes of AML using the World Health Organization (WHO) classification system (19). A total of seven patients had AML not otherwise specified, three had AML with an inversion in chromosome 16, one had AML with myelodysplasia-related change, and one had acute promyelocytic leukemia. BM and peripheral blood (PB) samples were frozen in fetal bovine serum (FBS; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO, USA) and stored in liquid nitrogen. BM- and PB-derived mononuclear cells (MNCs) were fractionated by density gradient centrifugation at 1,220 × g for 30 min at 4°C, using Ficoll-Paque™ (17-1440-03; GE Healthcare Life Sciences, Shanghai, China). The clinical characteristics and laboratory data of the patients with AML enrolled in the present study are listed in Table I. TIB152 human Jurkat cells (American Type Culture Collection, Manassas, VA, USA), were grown in RPMI medium (Thermo Fisher Scientific, Inc.) supplemented with 10% FBS in a humidified atmosphere of 5% CO2 at 37°C. CellTrace™ carboxyfluorescein diacetate succinimidyl ester (CFSE; C34554; Invitrogen, Thermo Fisher Scientific, Inc.) with 5 µM in DMSO was used to stain the Jurkat cells.

Table I

Clinical and laboratory features of patients with AML.

Table I

Clinical and laboratory features of patients with AML.

PatientWHO subtypeCellAge (years)GenderWBC/mm3 at diagnosisMolecular defectsCytogenetic anomalies
1AML NOSPB19M32240NEG46, XY [20]
2AML NOSPB64F127350FLT346, XX [20]
3AML with MRCBM65M260300MRC46, XY, del(5)(q11.2q15)[4]/46, XY[16]
4APLPB41M43010RARA46, XY, t(15;17)(q22;q12)[20]
5AML with inv(16)PB31M154500CBFB46, XY, t(9;22)(q34;q11.2), inv(16)(p13.1q22) [13]/47, idem, +17[15]/48, idem, +8, +17[2]
6AML NOSPB54F227830MLLT346, XX, t(9;11)(p22;q23)[20]
7AML NOSPB41M248521NPM146, XY [20]
8AML with inv(16)PB45M42234CBFB46, XY, inv(16)(p13.1q22)[20]
9AML NOSPB54M195104NPM146, XY [20]
10AML NOSPB36F240640NPM146, XX [20]
11AML with inv(16)PB46M108400CBFB46, XY, inv(16)(p13.1q22)[20]
12AML NOSPB65F114510NEG46, XX [20]

[i] WHO, World Health Organization; WBC, white blood cell; NOS, not otherwise specified; MRC, myelodysplasia-related change; APL, acute promyelocytic leukemia; inv(16), inversion in chromosome 16; PB, peripheral blood; BM, bone marrow; M, male; F, female; NEG, negative; FLT3, receptor-type tyrosine-protein kinase; RARA, retinoic acid receptor alpha; CBFB, core-binding factor subunit beta; MLLT3, myeloid/lymphoid or mixed-lineage leukemia protein translocated to 3; NPM1, nucleophosmin 1.

Magnetic-activated cell sorting and CFU assay

Podoplanin+ cells (BAF3670; R&D Systems, Inc., Minneapolis, MN, USA) were sorted and isolated from AML primary cells using magnetic beads (130-056-701; Miltenyi Biotec, Inc. Cambridge, MA, USA) in order to validate human clonogenic hematopoietic progenitor properties. Anti-biotin microbeads were used to isolate podoplanin (120-000-900; Miltenyi Biotec, Inc.). Sorted cells were cultured in methylcellulose media (H4434; STEMCELL Technologies, Inc., Vancouver, BC, Canada) for 7–10 days and colonies were counted using an inverted microscope (Leica Microsystems, Inc., Buffalo Grove, IL, USA).

Jurkat proliferation assay

CFSE-labeled Jurkat cells (2.5×103) were co-cultured with the sorted podoplanin+ and podoplanin cells (2.5×103) from BM-MNCs in RPMI medium supplemented with 1% FBS. After 24 h, the cells were stained with rabbit anti-human Ki67 antibody (cat no. ab15580; Abcam, Cambridge, UK) and counterstained with 4′,6-diamidino-2-phenylindole (DAPI). Cells positive for green fluorescent protein, CFSE-labeled Jurkat cells, Ki67 positive cells and DAPI-stained cells were counted under the inverted microscope.

Flow cytometry

Fluorescence activated cell sorting (FACS) staining and analysis was performed as previously described (20). Briefly, the cells were resuspended in 100 µl rinsing buffer and incubated with all antibodies at 4°C for 20 min. These included phycoerythrin (PE)-conjugated mouse anti-CD34 (1:20; cat no. 555822; BD Pharmingen, San Diego, CA, USA) and PEcy™ 5-conjugated mouse anti-CD38 (1:20; cat no. 555461; BD Pharmingen) antibodies, which were used to label leukemic stem cells (LSCs), and allophycocyanin (APC)-conjugated anti-human podoplanin polyclonal antibody (1:20; cat no. FAB3670A; R&D Systems, Inc.,), which was used for the detection of podoplanin. Subsequently, the cells were incubated with PE-annexin V (cat no. 556421; BD Pharminogen) for 20 min at room temperature for the detection of apoptosis. Following washing with 1% bovine serum albumin in phosphate-buffered saline (PBS; Thermo Fisher Scientific, Inc.), the cells were analyzed using a FACSCalibur flow cytometer equipped with CellQuest software, version 3.0 (BD Biosciences, San Diego, CA, USA).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA isolation and DNA synthesis were performed as previously described (21). PCR reactions were performed in a 50 µl PCR reaction mixture (Promega Corporation, Madison, WI, USA) containing 100 ng of each primer, 1X Tris-ethylenediaminetetraacetic acid buffer, 100 ng template DNA, 2.5 units HQ Taq polymerase, and 2.5 mM deoxyribonucleotide triphosphate. PCR amplification was performed using a conventional thermocycler (P×2 Thermal Cycler; Thermo Fisher Scientific, Inc.) under the following cycling conditions: 94°C for 4 min; 30–36 cycles at 94°C for 1 min, 53°C for 1 min, and 72°C for 2 min; extension cycle was at 72°C for 7 min. The RT-qPCR products were separated on a 2.0% agarose gel (Sigma-Aldrich) at 12 V/cm using a Tris-acetic acid-ethylenediaminetetraacetic acid buffer, and were subsequently stained with ethidium bromide (Thermo Fisher Scientific, Inc.), and visualized and photographed under an ultra-violet transilluminator (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Information regarding the primer/probe sets (TaqMan; Biosearch Technologies, Inc., Novato, CA, USA) and the primers used in the present study is provided in Table II. The relative mRNA expression of target genes was calculated using the comparative Cq method. All target gene expression was normalized to the expression of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in multiplexed reactions performed in triplicate. Differences in Cq values were calculated for each target mRNA by subtracting the mean value of the GAPDH expression (relative expression = 2−ΔΔCq) (22).

Table II

Primers and probes for reverse transcription-quantitative polymerase chain reaction.

Table II

Primers and probes for reverse transcription-quantitative polymerase chain reaction.

GenePrimers and probes (5′-3′)
Mouse GAPDHF: GGTGGTCTCCTCTGACTTCAACA
R: GTGGTCGTTGAGGGCAATG
P: CCACTCCTCCACCTTTGACGCTGG
Mouse Wt1F: AGCTGTCGGTGGCACAGTTGTCA
R: TGCCTGGGATGCTGGACTGTC
P: ACCCCTCAAAGCGCCAGCTGGAGTTT
Mouse survivinF: TCTGCTTTAAGGAATTGGAAGG
R: CTCTGTCTGTCCAGTTTCAAG
P: ACGGTTAGTTCTTCCATCTGCTTCTTGAC
Human GAPDHF: GGTGGTCTCCTCTGACTTCAACA
R: GTGGTCGTTGAGGGCAATG
Human podoplaninF: CAGGTGCCGAAGATGATGTG
R: TGTTGCCACCAGAGTTGTCA
Human FLT3F: GCATGCCTGGTTCAAGAGAA
R: TGCCAGGGTAAGGATTCACA

[i] GAPDH, glyceraldehyde 3-phosphate dehydrogenase; Wt1, Wilms' tumor gene 1; FLT3, Fms-like tyrosine kinase-3; F, forward; R, reverse; P, probe.

Immunostaining

Immunostaining was conducted as previously described (14). Briefly, using the cytospin method (4,23), cells were spun onto slides and fixed with 2% paraformaldehyde (Sigma-Aldrich) for 10 min at 25°C. Following washing with PBS, the cells were blocked with 5% horse serum (Thermo Fisher Scientific, Inc.) and incubated with the primary antibodies overnight at 4°C, followed by incubation with the secondary antibody for 30 min at room temperature. The primary antibodies used were as follows: Biotinylated anti-podoplanin (cat no. BAF3670; R&D Systems, Inc.), rabbit anti-CD34 (cat no. GWB-BBP214; GenWay Biotech, Inc., San Diego, CA, USA) and rabbit anti-Ki67. The Cy3 affinipure goat anti-IgG (cat no. NC9771594; Jackson Immuno-Research Laboratories, Inc., Inc., West Grove, PA, USA) secondary antibody was used. The cells were incubated with DAPI for 1 min at room temperature to stain the nuclei. Images were captured using the Zeiss LSM 510 META confocal laser scanning microscope and LSM 510 Imaging software, version 3.2 (Carl Zeiss, Inc., Gottingen, Germany).

Statistical analysis

All results are presented as the mean ± standard error. The comparison between groups was performed using the Mann-Whitney U test. GraphPad Prism version 4 software (GraphPad Software, Inc, La Jolla, CA, USA) was used for the statistical analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

High podoplanin expression on CD38+ differentiated cells in leukemia

To investigate the expression of podoplanin in leukemic cells, FACS analysis was performed in AML patient-derived cells. Under normal conditions, podoplanin is expressed in CD45 stromal cells, including osteocytes and osteoblasts; however, this protein is only expressed in CD45+ hematopoietic cells under disease conditions (14,21,24). The results of the present study demonstrated that the expression of podoplanin was markedly higher in mature CD38+ cells in complete remission (CR) than in those cells in the de novo AML state (AML, 53.9%; CR, 95.2%; Fig. 1A). Of note, under normal conditions, podoplanin+ cells were significantly more frequent in mature CD38+ cells (6.9%) than they were in CD34+CD38 HSCs (1.7%) (Fig. 1B). In CD38+ differentiated cells, the expression of podoplanin was significantly and gradually increased during the complete remission (CR) state, compared with the AML and normal states. This suggests that podoplanin-sustaining cells are required for BM reconstruction or blast protection, and that most podoplanin+ cells function as supportive cells rather than as LSCs. Due to the fact that CD38+ cells consist of a number of immune cells such as T, B, and nature killer cells, most CD38+ leukocytes that survive chemotherapy, may serve a role in blast communication in the tumor environment. A low frequency of CD34+ podoplanin+ cells was also detected in flushed cells, whereas, podoplanin single positive cells exhibited a high frequency (Fig. 1C), again suggesting that podoplanin cells can potentially function as supportive cells rather than as LSCs.

Enrichment of FLT3 in podoplanin−, however not podoplanin+ cells and high CFU-colony forming efficiency of podoplanin-cells

To further examine CFU potency, sorted cells were cultured in Matrigel gel supplemented with cytokines, and CFUs were observed after 10 days. Common myeloid progenitors were identified to be able to differentiate into two cell lineages: i) Granulocyte, erythrocyte, monocyte, megakaryocyte (GEMM), which includes megakaryocytes and erythrocytes, and ii) granulocyte-macrophage (GM) cells, which represent myeloblasts. Fig. 2 presents the colonies formed, including GEMM, G, GM and M from podoplanin+ or podoplanin cells. The number of CFU-GM colonies detected in CD34+ podoplanin cells was significantly higher than that of other colonies (Fig. 2). Colonies produced from normal HSCs were characterized and enumerated by their distinct cell morphology. Similarly, leukemic-derived colonies were also rapidly formed by a progenitor population; however, leukemic-derived colonies with atypical morphologies in CD34+ podoplanin cells overwhelmingly produced abnormal HSCs. The majority of formed colonies were small and condensed (<0.4 mm), which is consistent with previous studies (4,25,26), suggesting a putative leukemic stem/progenitor cell function of podoplanin cells. To examine whether these CFUs expressed leukemia-associated genes, and had a differential potency based on podoplanin expression, CD34+ podoplanin+ or CD34+ podoplanin cells were isolated using a microbead system. Sorted cells were immediately subjected to RT-qPCR to confirm the purity using podoplanin-specific primers, and the cells were then measured for FLT3, which is known to be overexpressed in patients with leukemia (27,28). The RT-qPCR data demonstrated that the podoplanin gene was exclusively expressed by the sorted podo-planin+ cells, and that the FLT3 gene was markedly increased in podoplanin cells, however not in podoplanin+ cells; however, the expression of these genes was similar in both podoplanin+ and podoplanin cells during differentiation (Fig. 3A). Sorted cells exhibited changeable expression of FLT3 and podoplanin at the time of differentiation, implying that there is some flexibility in the expression of AML genes.

These results suggested that leukemic properties are enriched by podoplanin rather than podoplanin+ cells. FLT3 acts as a molecular marker, and so it reflects a leukemic state (29,30); however, podoplanin+ cells may not be directly representative of leukemic cells. It has been reported that translocation of the chromosome containing the core-binding factor subunit beta 1 (CBFB1) gene results in AML (31). The expression of CBFB1 was restricted in podoplanin cells regardless of further differentiation, suggesting that podoplanin+ cells may function as stromal cells to podoplanin cells (data not shown), which contain leukemic stem cells expressing FLT3. At a protein level, podoplanin is primarily sustained in differentiated CFUs, and simultaneously detected in podoplanin cells (Fig. 3B), further suggesting its necessity in the maintenance of leukemic cells.

Leukemic cells can promote proliferative and anti-apoptotic effects under co-culture with podoplanin+ cells

To investigate the function of podoplanin+ cells as stromal cells, CFSE-stained Jurkat cells were cultured with podoplanin+ or podoplanin cells. After 24 h the Jurkat/podoplanin+ co-cultured cells exhibited a lower number of annexin-V+ cells (2.29-fold), compared with the Jurkat/podoplanin co-cultured cells (Fig. 4A), thus suggesting that podoplanin+ cells can protect leukemic cells from apoptosis. Additionally, Jurkat cells proliferated rapidly during co-culture with podoplanin+ cells. There was a significantly increased number of Ki67+ green fluorescent protein+ Jurkat cells during co-culture with podoplanin+ cells (1.47-fold), compared with the results of co-culture with podoplanin cells (Fig. 4B), suggesting the supportive role of podoplanin+ cells in leukemic cell activity. These results raised the question of whether primary blasts are able to upregulate their leukemic-associated genes in podoplanin+ stromal cell. Wilms' tumor gene 1 (WT1) and survivin, an apoptosis inhibitor encoded by survivin and expressed primarily in human blast cells, were selected for co-culture with podoplanin+ or podoplanin cells. Both genes are commonly regarded as leukemic-specific antigens and have been suggested to be upregulated under leukemic conditions (32). It was identified that the expression of WT1 and survivin was significantly increased (27.4-fold and 6.2-fold, respectively) in the cells co-cultured with podoplanin+ in vitro (Fig. 5), which supports a role of podoplanin+ cells in the maintenance of leukemic cells.

Discussion

Podoplanin was originally known as a protein marker for lymphatic endothelium (10). Previous studies have suggested a potential role of podoplanin in sustaining tumor cells in the tumor microenvironment (33,34). In addtition, podoplanin+ cells may function as stem/progenitor cells under lymphan-giogenic or lymphavasculogenic conditions in BM-derived cells (21) and regulate tumor metastasis (35), suggesting a multifactorial role of podoplanin in solid tumors. The role of podoplanin in leukemia, however, remains unclear. Previous studies reported that lymphangiogenic cytokines and markers, including podoplanin, are involved in leukemia, and in the BM microenvironment in particular (36,37).

Leukemic stem cells require stromal cells to survive chemotherapy (38). In numerous niches, stromal cells, including osteoblasts in normal BM, express podoplanin; this expression has been demonstrated to increase markedly under tumor conditions (24). In the present study, an increased level of podoplanin was observed in leukemic cells, which is consistent with previous studies of solid tumors (3941). Of note, CD38+ cells sustained a high podoplanin expression in the de novo AML and CR states following chemotherapy, and increased podo-planin is continuously required to maintain BM reconstruction or blast survival. The high expression of podoplanin in CD38+ cells, including leukocytes, may be associated with the release of podoplanin-soluble mediators. Cross-linkage between podoplanin-soluble mediator defensive action and surviving leukemic stem cells should be investigated in order to assist the development of targeted AML therapy.

Previously, Kim et al (42) reported that osteopontin (OPN) production by tumor cells, however not by stromal cells, enhances the propagation of tumor initiating cells in tumor environments, and that OPN silencing can delay tumor growth and extramedullary myelopoiesis. Like the diverse roles of OPN in tumor cells, the effects of podoplanin may alter depending on the environment; thus the present study investigated whether the inhibition of podoplanin was able to suppress leukemic blasts. A protective effect of podoplanin+ cells against apoptosis in blasts was detected, and further studies are required to identify cell type from podoplanin+ cells, which are associated with leukemic blasts. Stromal cell impairment leads to deficient hematopoiesis and chromosomal abnormalities, which may contribute to leukemogenesis (43,44), indicating the importance of micro-environment alteration in leukemia.

In the present study, leukemia-derived cells that express leukemia-related genes were markedly increased on podoplanin CD34+ cells. Podoplanin+ cells, which contain stromal cells, partly expressed hematopoietic-associated genes during differentiation; however, the mechanism through which this switching of podoplanin expression occurs, and the way it evolves to the progression of leukemic cells, remains unknown. Stromal cells appear to serve a role in AML by preventing apoptosis (45). Boyerinas et al (7) suggested that dormant leukemic cells are heavily regulated by the BM niche. By contrast, Flach et al (46) and Schepers et al (47) emphasized that DNA damage is responsible for the conversion of normal HSCs into malignant cells, and that LSC eventually leads to disruption of BM niches. Despite the controversy, understanding the association between LSCs and their surrounding environment is required for the treatment of AML.

Chemotherapy-resistant leukemic stem cells are typically observed in BM, and interact with stromal cells to promote blast retention (4750). Since the development of leukemia leads to alterations in microenvironmental factors, including immune and stromal cells, these alterations may directly or indirectly affect leukemic cells in a reciprocal manner (7,45,47). In the present study, a marked reduction in blast cell apoptosis was observed following co-culture with podoplanin+ cells, suggesting that blast cells rapidly promote cell proliferation, and have a protective role.

Further studies on syngeneic mouse models are required in order to gain insight into the function of podoplanin cells in leukemia, as well as to fully elucidate the functional properties of podoplanin+ stromal cells in the presence of cytokines or trafficking leukemia-associated mutant genes. The observations of the present study indicated that podoplanin+ cells in patients with leukemia are able to function as stromal cells, in order to protect against apoptosis and leukemic propagation with increased leukemic antigens.

Acknowledgments

The current study was supported by the Basic Science Research Program of the National Research Foundation of Korea funded by the Ministry of Education (grant. nos. 2014R1A1A2053407 and 2015R1D1A1A01059819).

References

1 

Pabst C, Krosl J, Fares I, Boucher G, Ruel R, Marinier A, Lemieux S, Hébert J and Sauvageau G: Identification of small molecules that support human leukemia stem cell activity ex vivo. Nat Methods. 11:436–442. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Shlush LI, Zandi S, Mitchell A, Chen WC, Brandwein JM, Gupta V, Kennedy JA, Schimmer AD, Schuh AC, Yee KW, et al: Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 506:328–333. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Zhang H, Mi JQ, Fang H, Wang Z, Wang C, Wu L, Zhang B, Minden M, Yang WT, Wang HW, et al: Preferential eradication of acute myelogenous leukemia stem cells by fenretinide. Proc Natl Acad Sci USA. 110:5606–5611. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Matsushita H, Nakajima H, Nakamura Y, Tsukamoto H, Tanaka Y, Jin G, Yabe M, Asai S, Ono R, Nosaka T, et al: C/EBPalpha and C/EBPvarepsilon induce the monocytic differentiation of myelomonocytic cells with the MLL-chimeric fusion gene. Oncogene. 27:6749–6760. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Wiley JM and Yeager AM: Predictive value of colony-forming unit assays for engraftment and leukemia-free survival after transplantation of chemopurged syngeneic bone marrow in rats. Exp Hematol. 19:179–84. 1991.PubMed/NCBI

6 

David H: Rudolf Virchow and modern aspects of tumor pathology. Pathol Res Pract. 183:356–364. 1988. View Article : Google Scholar : PubMed/NCBI

7 

Boyerinas B, Zafrir M, Yesilkanal AE, Price TT, Hyjek EM and Sipkins DA: Adhesion to osteopontin in the bone marrow niche regulates lymphoblastic leukemia cell dormancy. Blood. 121:4821–4831. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Wiseman DH, Greystoke BF and Somervaille TC: The variety of leukemic stem cells in myeloid malignancy. Oncogene. 33:3091–3098. 2014. View Article : Google Scholar

9 

Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, Diem K, Weninger W, Tschachler E, Alitalo K and Kerjaschki D: Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: Podoplanin as a specific marker for lymphatic endothelium. Am J Pathol. 154:385–394. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Schacht V, Ramirez MI, Hong YK, Hirakawa S, Feng D, Harvey N, Williams M, Dvorak AM, Dvorak HF, Oliver G and Detmar M: T1alpha/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. EMBO J. 22:3546–3556. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Kawase A, Ishii G, Nagai K, Ito T, Nagano T, Murata Y, Hishida T, Nishimura M, Yoshida J, Suzuki K and Ochiai A: Podoplanin expression by cancer associated fibroblasts predicts poor prognosis of lung adenocarcinoma. Int J Cancer. 123:1053–1059. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Yamanashi T, Nakanishi Y, Fujii G, Akishima-Fukasawa Y, Moriya Y, Kanai Y, Watanabe M and Hirohashi S: Podoplanin expression identified in stromal fibroblasts as a favorable prognostic marker in patients with colorectal carcinoma. Oncology. 77:53–62. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Kadota K, Huang CL, Liu D, Nakashima N, Yokomise H, Ueno M and Haba R: The clinical significance of the tumor cell D2-40 immunoreactivity in non-small cell lung cancer. Lung cancer. 70:88–93. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Lee JY, Park S, Kim DC, Yoon JH, Shin SH, Min WS and Kim HJ: A VEGFR-3 antagonist increases IFN-γ expression on low functioning NK cells in acute myeloid leukemia. J Clin Immunol. 33:826–837. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Conrad C, Niess H, Huss R, Huber S, von Luettichau I, Nelson PJ, Ott HC, Jauch KW and Bruns CJ: Multipotent mesenchymal stem cells acquire a lymphendothelial phenotype and enhance lymphatic regeneration in vivo. Circulation. 119:281–289. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Raica M, Cimpean AM and Ribatti D: The role of podoplanin in tumor progression and metastasis. Anticancer Res. 28:2997–3006. 2008.PubMed/NCBI

17 

Schacht V, Dadras SS, Johnson LA, Jackson DG, Hong YK and Detmar M: Up-regulation of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein, in human squamous cell carcinomas and germ cell tumors. Am J Pathol. 166:913–921. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Wicki A and Christofori G: The potential role of podoplanin in tumour invasion. Br J Cancer. 96:1–5. 2007. View Article : Google Scholar

19 

Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, Harris NL, Le Beau MM, Hellström-Lindberg E, Tefferi A and Bloomfield CD: The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: Rationale and important changes. Blood. 114:937–951. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Bae DS and Lee JK: Development of NK cell expansion methods using feeder cells from human myelogenous leukemia cell line. Blood Res. 49:154–161. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Lee JY, Park C, Cho YP, Lee E, Kim H, Kim P, Yun SH and Yoon YS: Podoplanin-expressing cells derived from bone marrow play a crucial role in postnatal lymphatic neovascularization. Circulation. 122:1413–1425. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

23 

Koh CM: Preparation of cells for microscopy using cytospin. Methods Enzymol. 533:235–240. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Ariizumi T, Ogose A, Kawashima H, Hotta T, Li G, Xu Y, Umezu H, Sugai M and Endo N: Expression of podoplanin in human bone and bone tumors: New marker of osteogenic and chondrogenic bone tumors. Pathol Int. 60:193–202. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Gishizky ML and Witte ON: Initiation of deregulated growth of multipotent progenitor cells by bcr-abl in vitro. Science. 256:836–839. 1992. View Article : Google Scholar : PubMed/NCBI

26 

Cheng H, Hao S, Liu Y, Pang Y, Ma S, Dong F, Xu J, Zheng G, Li S, Yuan W and Cheng T: Leukemic marrow infiltration reveals a novel role for Egr3 as a potent inhibitor of normal hematopoietic stem cell proliferation. Blood. 126:1302–1313. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Drexler HG: Expression of FLT3 receptor and response to FLT3 ligand by leukemic cells. Leukemia. 10:588–599. 1996.PubMed/NCBI

28 

Rosnet O, Bühring HJ, Marchetto S, Rappold I, Lavagna C, Sainty D, Arnoulet C, Chabannon C, Kanz L, Hannum C and Birnbaum D: Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells. Leukemia. 10:238–248. 1996.PubMed/NCBI

29 

Meshinchi S and Appelbaum FR: Structural and functional alterations of FLT3 in acute myeloid leukemia. Clin Cancer Res. 15:4263–4269. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Patel JP, Gonen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J, van Vlierberghe P, Dolgalev I, Thomas S, Aminova O, et al: Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 366:1079–1089. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Miyoshi H, Shimizu K, Kozu T, Maseki N, Kaneko Y and Ohki M: t(8;21) breakpoints on chromosome 21 in acute myeloid leukemia are clustered within a limited region of a single gene, AML1. Proc Natl Acad Sci USA. 88:10431–10434. 1991. View Article : Google Scholar : PubMed/NCBI

32 

Kim HJ, Choi EJ, Sohn HJ, Park SH, Min WS and Kim TG: Combinatorial molecular marker assays of WT1, survivin and TERT at initial diagnosis of adult acute myeloid leukemia. Eur J Haematol. 91:411–422. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Suzuki S, Ishii G, Matsuwaki R, Neri S, Hashimoto H, Yamauchi C, Aokage K, Hishida T, Yoshida J, Kohno M, et al: Ezrin-expressing lung adenocarcinoma cells and podoplanin-positive fibroblasts form a malignant microenvironment. J Cancer Res Clin Oncol. 141:475–484. 2015. View Article : Google Scholar

34 

Chuang WY, Yeh CJ, Chao YK, Liu YH, Chang YS, Tseng CK, Chang HK, Wan YL and Hsueh C: Concordant podoplanin expression in cancer-associated fibroblasts and tumor cells is an adverse prognostic factor in esophageal squamous cell carcinoma. Int J Clin Exp Pathol. 7:4847–4856. 2014.PubMed/NCBI

35 

Dang Q, Liu J, Li J and Sun Y: Podoplanin: A novel regulator of tumor invasion and metastasis. Med Oncol. 31(24)2014. View Article : Google Scholar : PubMed/NCBI

36 

Lee JY and Kim HJ: (Lymph) angiogenic influences on hematopoietic cells in acute myeloid leukemia. Exp Mol Med. 46:e1222014. View Article : Google Scholar

37 

Lee JY, Park S, Min WS and Kim HJ: Restoration of natural killer cell cytotoxicity by VEGFR-3 inhibition in myelogenous leukemia. Cancer lett. 354:281–289. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Junttila MR and de Sauvage FJ: Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 501:346–354. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Chang YW, Hsieh PW, Chang YT, Lu MH, Huang TF, Chong KY, Liao HR, Cheng JC and Tseng CP: Identification of a novel platelet antagonist that binds to CLEC-2 and suppresses podoplanin-induced platelet aggregation and cancer metastasis. Oncotarget. 6:42733–42748. 2015.PubMed/NCBI

40 

Grau SJ, Trillsch F, Tonn JC, Goldbrunner RH, Noessner E, Nelson PJ and von Luettichau I: Podoplanin increases migration and angiogenesis in malignant glioma. Int J Clin Exp Pathol. 8:8663–8670. 2015.PubMed/NCBI

41 

Tanaka M, Kijima H, Shimada H, Makuuchi H, Ozawa S and Inokuchi S: Expression of podoplanin and vimentin is correlated with prognosis in esophageal squamous cell carcinoma. Mol Med Rep. 12:4029–4036. 2015.PubMed/NCBI

42 

Kim EK, Jeon I, Seo H, Park YJ, Song B, Lee KA, Jang Y, Chung Y and Kang CY: Tumor-derived osteopontin suppresses antitumor immunity by promoting extramedullary myelopoiesis. Cancer Res. 74:6705–6716. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Blau O, Baldus CD, Hofmann WK, Thiel G, Nolte F, Burmeister T, Türkmen S, Benlasfer O, Schümann E, Sindram A, et al: Mesenchymal stromal cells of myelodysplastic syndrome and acute myeloid leukemia patients have distinct genetic abnormalities compared with leukemic blasts. Blood. 118:5583–5592. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Geyh S, Oz S, Cadeddu RP, Fröbel J, Brückner B, Kündgen A, Fenk R, Bruns I, Zilkens C, Hermsen D, et al: Insufficient stromal support in MDS results from molecular and functional deficits of mesenchymal stromal cells. Leukemia. 27:1841–1851. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Konopleva M, Konoplev S, Hu W, Zaritskey AY, Afanasiev BV and Andreeff M: Stromal cells prevent apoptosis of AML cells by up-regulation of anti-apoptotic proteins. Leukemia. 16:1713–1724. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Flach J, Bakker ST, Mohrin M, Conroy PC, Pietras EM, Reynaud D, Alvarez S, Diolaiti ME, Ugarte F, Forsberg EC, et al: Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature. 512:198–202. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Schepers K, Pietras EM, Reynaud D, Flach J, Binnewies M, Garg T, Wagers AJ, Hsiao EC and Passegué E: Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell. 13:285–299. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, Nakamura R, Tanaka T, Tomiyama H, Saito N, et al: Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 25:1315–1321. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Saito Y, Uchida N, Tanaka S, Suzuki N, Tomizawa-Murasawa M, Sone A, Najima Y, Takagi S, Aoki Y, Wake A, et al: Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat Biotechnol. 28:275–280. 2010.PubMed/NCBI

50 

Bendall LJ, Kortlepel K and Gottlieb DJ: Human acute myeloid leukemia cells bind to bone marrow stroma via a combination of beta-1 and beta-2 integrin mechanisms. Blood. 82:3125–3132. 1993.PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 13 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee JY, Han AR, Lee SE, Min WS and Kim HJ: Co-culture with podoplanin+ cells protects leukemic blast cells with leukemia-associated antigens in the tumor microenvironment. Mol Med Rep 13: 3849-3857, 2016
APA
Lee, J.Y., Han, A., Lee, S., Min, W., & Kim, H. (2016). Co-culture with podoplanin+ cells protects leukemic blast cells with leukemia-associated antigens in the tumor microenvironment. Molecular Medicine Reports, 13, 3849-3857. https://doi.org/10.3892/mmr.2016.5009
MLA
Lee, J. Y., Han, A., Lee, S., Min, W., Kim, H."Co-culture with podoplanin+ cells protects leukemic blast cells with leukemia-associated antigens in the tumor microenvironment". Molecular Medicine Reports 13.5 (2016): 3849-3857.
Chicago
Lee, J. Y., Han, A., Lee, S., Min, W., Kim, H."Co-culture with podoplanin+ cells protects leukemic blast cells with leukemia-associated antigens in the tumor microenvironment". Molecular Medicine Reports 13, no. 5 (2016): 3849-3857. https://doi.org/10.3892/mmr.2016.5009