Altered susceptibility to apoptosis and N‑glycan profiles of hematopoietic KG1a cells following co‑culture with bone marrow‑derived stromal cells under hypoxic conditions

  • Authors:
    • Xingchen Pang
    • Yi Wang
    • Sijie Zhang
    • Zengqi Tan
    • Jia Guo
    • Feng Guan
    • Xiang Li
  • View Affiliations

  • Published online on: July 5, 2018     https://doi.org/10.3892/or.2018.6548
  • Pages: 1477-1486
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mesenchymal stromal cells are an important component of the bone marrow microenvironment (niche), where they support hematopoiesis via direct cell‑cell interactions with hematopoietic stem and progenitor cells, and by releasing soluble factors. Glycans, including N‑glycans, are involved in numerous biological processes, including inflammation, cell‑cell interactions, as well as cancer development and progression. Lectin‑based microarray analysis has provided a powerful new tool in recent years, for the investigation of aberrantly expressed N‑glycans and their functions in the bone marrow microenvironment. In the present study, we used an in vitro stromal/hematopoietic cell co‑culture system to examine the effects of stromal‑derived signals on apoptosis susceptibility of co‑cultured KG1a hematopoietic cells under hypoxic (1% O2) conditions. MALDI‑TOF/TOF‑MS analysis was used for the comparative global profiling of N‑glycans in KG1a cells and co‑cultured KG1a cells under hypoxia. KG1a cells became more susceptible to p53‑dependent apoptosis when co‑cultured with HS27A human stromal cells (derived from normal bone marrow) under hypoxia. We observed enhanced levels of core‑fucosylated N‑glycans (catalyzed by FUT8), bisecting GlcNAc (catalyzed by MGAT3), and their corresponding genes in co‑cultured cells. In addition we observed that overexpressing MGAT3 or FUT8 facilitated cell apoptosis in KG1a cells. Collectively, our data revealed the profiling of N‑glycans in KG1a cells before and after stroma contact. Our findings and future functional studies of core‑fucosylated N‑glycans and bisecting GlcNAc, will improve our understanding of the bone marrow microenvironment.

Introduction

The bone marrow microenvironment consists of a specialized population of cells that play essential roles in regulation, self-renewal and differentiation of adult stem cells (14). The microenvironment supports maturation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) and their release into the vascular system (57). Human bone marrow-derived stromal cell lines HS5 and HS27A, co-cultured with myeloid cells, are frequently used in studies of crosstalk between cells in the bone marrow microenvironment and hematopoietic cells (813). For example, HS5 and HS27A have been used to establish a xenotransplantation murine model of myelodysplastic syndromes (MDS), a group of clonal hematopoietic disorders characterized by dysregulation of programmed cell death (apoptosis) and ineffective hematopoiesis in both normal and clonal (transformed) hematopoietic cells (12,14). Kerbauy et al (14) observed engraftment of distinct clonal MDS-derived hematopoietic precursors when stromal cells (HS5 and HS27A combined) were co-injected via an intramedullary route. Our previous study found that intravenous co-administration of HS27A cells (but not HS5 cells) with HPCs from MDS patients facilitated engraftment of clonal CD34+ cells of any karyotype (12), indicating that HS27A cells were more effective than HS5 in supporting primitive clonal MDS precursors (12).

Glycans, a major category of biomolecules, are commonly attached to proteins and lipids to form glycoproteins, glycolipids, glycosaminoglycans and other types of glycoconjugates (1517). Glycans play key roles in a variety of biological processes, including cell adhesion, molecular trafficking, receptor activation and signal transduction (17,18). They are also involved in malignant hematopoiesis. CoCl2 (hypoxia mimic) induced expression of cell surface glycans recognized by both β-galactoside- and GlcNAc-binding lectins in HL-60 cells (19). Galectin-3 is developmentally expressed in human myeloid cells, and is strongly upregulated on the surface of late mature myeloid cells (20). Conversely, it also promotes proliferation and angiogenesis of endothelial cells differentiated from bone marrow mesenchymal stem cells (21). An important characteristic of the HSC microenvironment is that low oxygen (O2) tension and hypoxia (defined as O2 tension <2%) is essential for HSC function (2225). In vitro BM cells are always cultured under hypoxic conditions in order to maintain their primitive phenotype and self-renewal ability (26). In addition, the key regulator of cellular adaptation to hypoxic stress, hypoxia-inducible factor 1 α (HIF-1α) can be stabilized under hypoxia (25,26). No study to date has included global analysis of the expression of N-glycan in hematopoietic cells co-cultured with vs. without stromal cells under hypoxia.

An in vitro model in which KG1a hematopoietic cells were co-cultured with HS27A cells under hypoxia conditions (1% O2) was used in the present study. We examined the apoptosis rate of the KG1a cells, and the related apoptotic pathway. N-glycans linked to glycoproteins were released by PNGase F and analyzed by MALDI-TOF/TOF-MS, and selectively altered N-glycans were further analyzed by lectin staining. By presenting global analysis of patterns of altered N-glycans in hematopoietic cells before and after stroma contact under hypoxia, the present study provides valuable information for future investigations of the bone marrow microenvironment.

Materials and methods

Cell culture

KG1a cells (derived from acute myeloid leukemia) and bone marrow-derived stromal cell line HS27A are generous gifts from Professor H. Joachim Deeg (Fred-Hutchinson Cancer Research Center, Seattle, WA, USA). KG1a and HS27A were grown, propagated and subjected to experiments between passages 8 and 24, as previously described (27). Multiple aliquots from early passages were cryopreserved for later use.

Total protein extraction

Cells were harvested and lysed with T-PER Tissue Protein Extraction reagent (Thermo Fisher Scientific, Hudson, NH, USA) according to the manufacturer's protocol. In brief, cells were trypsinized, resuspended in phosphate-buffered saline (PBS), added with T-PER reagent containing 1% protease inhibitor cocktail (Biotool Ltd., Houston, TX, USA), incubated on ice for 30 min and homogenized. The sample was centrifuged for 15 min at 13,000 × g (4°C), and the supernatant was collected and stored at −80°C.

Enzyme-linked immunosorbent assay (ELISA)

Concentrations of secreted tumor necrosis factor α (TNFα) in culture supernatants were determined (in triplicate) using an ELISA kit (R&D Systems, Minneapolis, MN, USA) for human TNFα (detection limit 0.5 pg/ml). Color intensity of the chromogenic reaction was determined at 490 nm by a plate reader (Bio-Rad Laboratories, Hercules, CA, USA).

Western blot analysis

Proteins from each sample were separated by 10% SDS-PAGE and transferred onto polyvinylidene fluoride (PVDF) membranes using the Trans-Blot Turbo Transfer System (Bio-Rad Laboratories). Membranes were soaked in 5% (w/v) skim milk in TBST (20 mM Tris-HCl, 150 mM NaCl, 0.05% Tween-20, pH 8.0) for 2 h at 37°C, probed with primary antibodies against HIF-1α (1:1,000; cat. no. 3716; Cell Signaling Technology, Inc., Beverly, MA, USA), p53 (1:1,000; cat. no. 2524; Cell Signaling Technology, Inc.), Bax (1:1,000; cat. no. 2774; Cell Signaling Technology, Inc.), Bcl-2 (1:1,000; 2872; Cell Signaling Technology, Inc.), caspase-3 (1:1,000; cat. no. 9662; Cell Signaling Technology, Inc.), caspase-8 (1:1,000; cat. no. 9746; Cell Signaling Technology, Inc.), caspase-9 (1:1,000; cat. no. 9502; Cell Signaling Technology, Inc.), NF-κB (1:1,000; cat. no. 8242; Cell Signaling Technology, Inc.), phospho-NF-κB (1:1,000; cat. no. 3031; Cell Signaling Technology, Inc.) and β-tubulin (1:5,000; cat. no. T7816; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) overnight at 4°C, and incubated with the appropriate HRP-conjugated secondary antibody (1:5,000; cat. nos. A0208 and A0216; Beyotime Institute of Biotechnology, Shanghai, China). Bands were visualized using enhanced chemiluminescence detection kit Westar Nova (Cyanagen Srl, Bologna, Italy) with imaging by ChemiDoc™ XRS+ (Bio-Rad Laboratories).

Cell sorting

KG1a cells were co-cultured with HS27A cells for 24 h and added together with CD45+ magnetic beads. KG1a cells were isolated by magnetic-activated cell sorting (Miltenyi Biotec, Bergisch Gladbach, Germany) according to the manufacturer's protocol. Cell purity was assessed by staining with fluorescent-tagged anti-CD45 antibody and quantified by flow cytometry with the purity of sorted cells reaching >95%. Apoptosis was assessed by flow cytometry.

Caspase activity assay

Caspase activity assay was performed according to the manufacturer's instructions (Beyotime Institute of Biotechnology). Briefly, after treatment, isolated KG1a cells were lysed with lysis buffer and the supernatants were incubated with Ac-DEVD-pNA (200 µM) substrate for caspase-3 and Ac-IETD-pNA (200 µM) substrate for caspase-8, respectively. The reaction was assessed with a plate reader at 405 nm wavelength.

Quantitative real-time PCR

Total RNA was extracted as previously described (28). Primers were designed using the Primer-BLAST program (https://www.ncbi.nlm.nih.gov/tools/primer-blast/). First-strand cDNA was synthesized from total RNA using a OneScript Plus cDNA Synthesis kit (Abm Canada, Milton, ON, Canada). Quantitative real-time PCR was performed by SYBR-Green I dye detection with BrightGreen Express 2× qPCR Master Mix (Abm Canada). Gene expression was quantified by the 2−ΔΔCq method (29). All primer sequences are provided in Table I.

Table I.

Quantitative real-time PCR primer sequences.

Table I.

Quantitative real-time PCR primer sequences.

GenesPrimer sequences
FUT8F: 5′-TGTCCTGTACTTCATGCGCT-3′
R: 3′-TCCATGACCCTAATGGTCTTTT-5′
MGAT2F: 5′-AAAGAACACCTGCAGAACCG-3′
R: 3′-GGAATTGACAACGTCCTCGT-5′
MGAT3F: 5′-AGGAAGGAGATGAGGCACAG-3′
R: 3′-TTGCTGAGACCCAGCGG-5′
MGAT4BF: 5′-TCACTGCCGAAGTGTACTGTG-3′
R: 3′-CTGACACTCTGCACTCGCTC-5′
MGAT5F: 5′-GTGAGGGTAGCCGTCCATAG-3′
R: 3′-CAGCTTGGTTGCACTTGAGA-5′

[i] F, forward; R, reverse.

Apoptosis assay

Apoptosis of hematopoietic cells was assayed by Annexin V staining. KG1a cells were cultured under hypoxia for 24 h with or without HS27A stroma contact, on the basis of our previous studies (9,11). Twelve-well plates were centrifuged at 1,200 rpm (300 × g), the supernatant was discarded, and cells in each well were washed with cold PBS containing 2% bovine serum albumin (BSA) and assayed using an Annexin V-fluorescein isothiocyanate (FITC) apoptosis kit (Beyotime Institute of Biotechnology). Cells were then labeled with Allophycocyanin (APC)-conjugated anti-CD45 antibody (1:20; cat. no. 368512; BioLegend, San Diego, CA, USA) to distinguish hematopoietic cells (myeloid cells, CD45+) from stromal (CD45), and apoptosis was assayed by flow cytometry (model BD Accuri C6; BD Biosciences, San Jose, CA, USA) using Annexin V-FITC and propidium iodide (PI).

N-glycan mass profiling

N-glycans were separated as described in our previous study (30). In brief, we used a size-exclusion spin ultrafiltration unit (Amicon Ultra-0.5 10 KD; EMD Millipore; Merck KGaA, Darmstadt, Germany) to concentrate 2 mg total proteins. Proteins were denatured, centrifuged and then further digested with PNGase F (New England BioLabs, Ipswich, MA, USA) overnight at 37°C. Released N-glycans were collected and desalted using Sepharose 4B (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), as previously described (31), then characterized by MALDI-TOF/TOF-MS (ultrafleXtreme; Bruker Daltonik GmbH, Bremen, Germany) in positive-ion mode. Data were analyzed using the FlexAnalysis software program (Bruker Daltonik GmbH) and annotated using the GlycoWorkbench software program (http://code.google.com/p/glycoworkbench/). The stability of MS was estimated using coefficient of variation (CV) percentages based on relative intensity values. The relative variation was calculated by dividing the amount of a given type of N-glycan by the total N-glycan amount (30).

Flow cytometric (FACS) analysis of lectin binding affinity

Cells were harvested to a single cell suspension with trypsin or 0.2% EDTA, and washed three times with PBS. Aliquots (106 cells) were resuspended in 100 µl diluting Cy3-conjugated lectin (cat. no. L-1000, L-1020, L-1040 and L-1120; Vector Labs, Peterborough, UK) and APC-conjugated anti-human CD45 antibody (1:20; cat. no. 368512; BioLegend) in 0.1% BSA/PBS, and incubated on ice for 30 min in the dark. Lectins were used at a final concentration 100 µg/ml. Cells were washed twice with 1 ml PBS and analyzed by flow cytometry as above-mentioned.

Lectin staining

Lectin staining was performed as previously described (30). Cells were spun onto a microscope slide for 5 min at 300 × g, immobilized with 2% fresh paraformaldehyde for 15 min at room temperature, and blocked with 5% BSA in 1× PBS for 1 h at 37°C. Fixed cells were incubated with 20 µg/ml Cy3-conjugated lectin (LCA, PHA-E) in 5% BSA for 3 h in the dark, washed with 1× PBS, stained with 20 µg/ml DAPI in 1× PBS for 10 min, washed again with 1× PBS, and images were captured with a fluorescence microscope (model Eclipse E600; Nikon, Tokyo, Japan) at the same exposure time and gain factor.

Statistical analysis

Data are presented as the mean ± SEM. Statistical significance of differences between the means of two groups was evaluated by Student's t-test. Multiple group comparisons were evaluated by ANOVA with Bonferroni's post hoc test.

Results

Co-culture with stromal cells enhances susceptibility of KG1a cells to apoptosis under a hypoxic condition

Hematopoietic cell line KG1a is resistant to apoptosis (32,33). However, apoptosis of KG1a was significantly increased when they were co-cultured with HS27A stromal cells under a hypoxic condition (Fig. 1A and B). We investigated the apoptosis of KG1a cells before and after co-culture with HS27A in the presence of 100 µM CoCl2. Treatment with CoCl2 mimics hypoxic conditions by inhibiting prolyl-4-hydroxylases involved in the degradation of HIF-1α (34,35). CoCl2 was used as a positive control in the present study. Apoptosis of KG1a cells co-cultured with HS27A cells was also increased by CoCl2 treatment (Fig. 1C and D). Concentration of TNFα, another factor that enhances susceptibility of KG1a cells to apoptosis, was increased in the co-culture system under hypoxic conditions (Fig. 1E). TNF receptor I (TNFR1) was also increased in KG1a cells following co-culture with stromal cells under hypoxic conditions (Fig. 1F and G).

Hypoxia induces the p53-dependent pathway in co-cultured KG1a cells

Our previous studies demonstrated upregulation of p53 levels in hematopoietic cells and primary MDS marrow cells in stromal contact culture (8,9). To investigate the possible involvement of a p53-dependent pathway in hypoxia-induced apoptosis in co-cultured KG1a cells, we examined expression of p53, caspase-3, caspase-8, caspase-9 and NF-κB in KG1a cells after co-culture with HS27A cells under hypoxic conditions and CoCl2 treatment. In co-cultured KG1a cells, p53, caspase-8, caspase-9 and phosphorylated NF-κB levels were elevated and caspase-3 and caspase-8 were cleaved and active (Fig. 2A and B). We further observed that the activities of caspase-3 and caspase-8 were also elevated in co-cultured KG1a cells, which appeared to be relevant with the increase in TNF-α and TNFR1 (Fig. 2C). Bax and Bcl-2 levels were respectively upregulated and downregulated in the co-cultured cells, as expected since their expression was affected by p53 through transcriptional regulation (36,37).

MALDI-TOF/TOF-MS profiles of N-glycans in KG1a cells before and after co-culture under hypoxic conditions

N-glycans are involved in a variety of biological processes, including cell-cell and cell-matrix interactions, receptor-mediated functions and specific protein functions. Many types of tumor cells display aberrant N-glycosylation patterns, and cancer-associated N-glycans are potential biomarkers for early detection and diagnosis of cancer (38). We compared MALDI-TOF/TOF-MS profiles of total N-glycans in KG1a cells before vs. after co-culture with HS27A cells under hypoxic conditions. MALDI-TOF/TOF-MS spectra of N-glycans with signal-to-noise ratios >5 from total glycoproteins were annotated using the GlycoWorkbench program (Fig. 3). Eight distinctive N-glycans were observed in KG1a cells before vs. after co-culture, at m/z 1257.476, 1419.534, 1485.526, 1581.592, 1688.598, 1743.650, 1905.711 and 2067.687, corresponding to eight proposed N-glycan structures including high-mannose, bi-antennary and tri-antennary types (Fig. 4). There were eight N-glycan structures found in both KG1a cells and co-cultured KG1a cells, two structures unique to co-cultured KG1a cells, and one structure unique to KG1a cells. Co-cultured KG1a cells showed enhancement of two complex type N-glycan structures with fucosylation.

Validation by real-time PCR of N-glycan biosynthesis-related genes

On the basis of our MS data, we examined the expression of N-glycan biosynthesis-related genes by real-time PCR. The glycosyltransferase genes involved in the synthesis of the N-glycan structures listed in Fig. 4 are α1,6-fucosyltransferase (FUT8) and β1,4-N-acetylglucosaminyltransferases 2, 3, 4A, 4B and 5 (MGAT2, MGAT3, MGAT4A, MGAT4B and MGAT5) (Fig. 5A). Among these six genes, FUT8, MGAT3 and MGAT5 showed increased expression in co-cultured KG1a cells, whereas the expression of MGAT2 and MGAT4B was not notably altered. These findings were consistent with our MS results. We then applied flow cytometric (FACS) analysis with specific lectins to further examine differentially expressed N-glycans in KG1a cells co-cultured with HS27A cells under hypoxic conditions. Co-cultured cells showed increased expression of N-glycan structures recognized by PHA-E (i.e., bisecting GlcNAc structures), LCA (core Fucα1-6GlcNAc) and WGA (multivalent Sia and (GlcNAc)n structures), and reduced expression of structures recognized by ConA (terminal mannose) (Fig. 5B-D).

Validation by flow cytometry of N-glycan expression differences in co-cultured KG1a cells under hypoxic conditions

To verify the function of FUT8 and MGAT3, we overexpressed these two genes in KG1a cells (Fig. 6A). Flow cytometric data revealed that the corresponding bisecting N-glycan or core fucosylation in KG1a cells that overexpressed MGAT3 or FUT8 was enhanced (Fig. 6B). Our data showed an enhanced apoptosis rate in KG1a cells that overexpressed MGAT3 or FUT8 under both hypoxic conditions and CoCl2 treatment (Fig. 6C and D). We suggest that modulation of glycogenes may play a role in regulating the apoptosis in KG1a cells.

Discussion

Tumor/stromal cell interactions are frequently part of cell social networks, play essential roles in tumor development and growth, and provide useful therapeutic targets. Our 2010 studies were focused on an in vitro co-culture system involving two stromal cell lines (HS5 and HS27A) derived from a healthy bone marrow donor. Cells from patients with advanced MDS showed increased susceptibility to TNFα-induced apoptosis following stromal contact (8,9). Our 2013 in vivo study, based on the in vitro observations, involved injection of HS5 or HS27A cells, together with primary MDS marrow cells, into NOD.Cg-Prkdcscid Il2rgtm1Wjl (NSG) mice (12). HS27A cells, but not HS5 cells, facilitated the engraftment of clonal MDS cells in vivo. The present study focused on the crosstalk that occurs between hematopoietic and HS27A cells (but not HS5). Among the numerous factors present in the tumor microenvironment, hypoxia is a common feature of myeloid malignancies and has been shown to affect such HSC behaviors as apoptosis, proliferation, differentiation and resistance to chemotherapy. The role of hypoxia and its downstream signaling in hematopoiesis is controversial, since both promoting and inhibitory effects have been reported (39). Velasco-Hernandez et al found that deletion of HIF-1α in mouse hematopoietic cells promoted acute myeloid leukemia progression (40). Other studies indicated that hypoxia promoted leukemogenic niche metabolism and cytokine secretion (41,42). In the present study, we compared the apoptosis rates of hematopoietic KG1a cells alone or co-cultured with HS27A cells under hypoxic conditions. Our data clearly indicated that elevated caspase 8 activities were found in co-cultured KG1a cells. However, cleaved caspase-9 had no significant changes in co-cultured KG1a cells, even though the total amount of caspase-9 was increased, indicating that the co-culture may trigger the expression of total caspase-9 which possibly make the co-cultured KG1a cells becoming more sensitive to cell death signals (4345). Increased levels of p53 in the co-cultured cells revealed that the apoptotic pathway was p53-dependent.

Functional proteins in both normal and malignant cells are maintained by post-translational modifications (PTMs), which include phosphorylation, ubiquitination, methylation, N-acetylation and glycosylation (30). Glycosylation, the most common PTM, is involved in a wide variety of biological processes. Glycosylation of proteins (giving rise to glycoproteins) in eukaryotic cells is classified on the basis of various linkages of glycans to protein core regions e.g., N-linked glycan (GlcNAc linkage to Asn), O-linked glycan (O-GalNAc linkage to Ser/Thr), and other O-linked glycan varieties (e.g., O-linked mannose, O-linked GlcNAc) (30). Biological processes mediated by glycans include signal transduction, inflammation, virus/bacteria-host interactions, cell-cell interactions and cancer development and progression. Our MS analyses of N-glycan profiles of co-cultured KG1a cells under hypoxia revealed increases in bi- and tri-antennary type, GlcNAc type and fucose type N-glycans.

Synthesis of N-glycans is catalyzed by various enzymes termed glycosyltransferases. Fucosylation is catalyzed by FUT8, which transfers a fucose (Fuc) moiety from GDP-β-L-Fuc to the innermost GlcNAc residue of an N-glycan, resulting in α1,6-Fuc residue as core Fuc. FUT8 activity is upregulated during epithelial-mesenchymal transition (EMT) through transactivation of β-catenin/lymphoid enhancer-binding factor-1 (LEF-1) (46). The glycosyltransferase MGAT3 catalyzes transfer of GlcNAc to the core β-mannose residue of N-glycans having β1,4-linkage. Addition of bisecting GlcNAc residue to core β-mannose by MGAT3 inhibits activities of mannosidase II and other GlcNAc transferases (MGAT2, MGAT4). Li et al and Xu et al reported reduced levels of MGAT3 and its product (bisecting N-glycans) in a TGF-β1-induced EMT model (16,47). Takahashi et al observed that MGAT3 overexpression reduced the amount of poly-N-acetyllactosamine on N-glycans of EGFR, indicating that specific terminal structures of N-glycans regulated EGFR endocytosis through interaction with carbohydrate recognition molecules (48). MGAT3 exerts a stabilizing effect on E-cadherin at the cell membrane by inducing a delay in the turnover rate of the protein, thereby contributing to stable and functional adhesion, and blocking clathrin-dependent E-cadherin endocytosis (49). Remodeling of glycans by MGAT3 contributes to the retention of E-cadherin in membrane by mesalamine (5-ASA) (50). In contrast, MGAT5 promotes destabilization of E-cadherin, leading to its mislocalization, formation of unstable junctions and impairment of cell-cell adhesion (49). Inhibition of MGAT5 activity reduced liver fibrosis and suppressed TGF-β1-induced EMT in hepatocytes, as evidenced by reversal of EMT markers. Our model (co-cultured KG1a under hypoxia) showed increased expression of FUT8, MGAT3 and MGAT5, but no notable change of MGAT2 or MGAT4B. Increased binding affinity of lectins PHA-E and WGA in our model revealed elevated levels of bisecting-GlcNAc and (GlcNAc)n structures, in striking contrast to N-glycan structures in co-cultured KG1a under normoxia (data not shown). We then overexpressed FUT8 and MGAT3 genes in KG1a cells to ascertain whether modified glycogenes could impact the cell apoptosis of KG1a cells under hypoxia. Our data suggested that modified glycogenes of both FUT8 and MGAT3 could enhance cell apoptosis in KG1a cells. Our further studies will focus on the functional role of FUT8 and MGAT3 in primary cells, as well as the microenvironmental niche.

In conclusion, the present study provided the first demonstration of hypoxia-induced apoptosis in KG1a hematopoietic cells following contact with HS27A stromal cells, and N-glycan profiles of KG1a cells before and after stromal contact under hypoxia. Our findings are useful references for future studies based on co-culture models, and may help elucidate the roles of hematopoietic and stromal cells in bone-related diseases. Our ongoing studies will focus on the functions of N-glycans in co-culture systems and the clinical relevance of these N-glycans.

Acknowledgements

The authors are grateful to Dr S. Anderson for the English language editing of the manuscript.

Funding

The present study was supported in part by grants from the National Natural Science Foundation of China (nos. 81470294, 31400691 and 81770123), the Natural Science Foundation of Jiangsu Province (BK20140169), the Fundamental Research Funds for the Central Universities (JUSRP51619B) and the 13115 Key Projects of Scientific and Technical Innovation of Shaan'xi Province (2010ZDKG-53).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

XL and YW designed the study and wrote the manuscript. XP, SZ, XL, ZT and JG performed the experiments. FG critiqued the manuscript and was also involved in the conception and design of the study. All authors have read and approved the final manuscript. All authors agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HSCs

hematopoietic stem cells

HPCs

hematopoietic progenitor cells

MDS

myelodysplastic syndrome

HIF-1α

hypoxia-inducible factor 1α

TNFα

tumor necrosis factor α

PTMs

post-translational modifications

EMT

epithelial-mesenchymal transition

LEF-1

lymphoid enhancer-binding factor-1

References

1 

Trentin JJ: Determination of bone marrow stem cell differentiation by stromal hemopoietic inductive microenvironments (HIM). Am J Pathol. 65:621–628. 1971.PubMed/NCBI

2 

Bennett M and Kumar V: 89Sr-induced bone marrow aplasia: Effects on seed (stem cells) and soil (inductive microenvironment). Lab Invest. 49:235–236. 1983.PubMed/NCBI

3 

Morrison SJ and Scadden DT: The bone marrow niche for haematopoietic stem cells. Nature. 505:327–334. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Simmons PJ and Torok-Storb B: Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood. 78:55–62. 1991.PubMed/NCBI

5 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Manabe A, Coustan-Smith E, Behm FG, Raimondi SC and Campana D: Bone marrow-derived stromal cells prevent apoptotic cell death in B-lineage acute lymphoblastic leukemia. Blood. 79:2370–2377. 1992.PubMed/NCBI

7 

Ayala F, Dewar R, Kieran M and Kalluri R: Contribution of bone microenvironment to leukemogenesis and leukemia progression. Leukemia. 23:2233–2241. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Marcondes AM, Li X, Gooley TA, Milless B and Deeg HJ: Identification of DJ-1/PARK-7 as a determinant of stroma-dependent and TNF-alpha-induced apoptosis in MDS using mass spectrometry and phosphopeptide analysis. Blood. 115:1993–2002. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Li X, Marcondes AM, Gooley TA and Deeg HJ: The helix-loop-helix transcription factor TWIST is dysregulated in myelodysplastic syndromes. Blood. 116:2304–2314. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Marcondes AM, Li X, Tabellini L, Bartenstein M, Kabacka J, Sale GE, Hansen JA, Dinarello CA and Deeg HJ: Inhibition of IL-32 activation by α-1 antitrypsin suppresses alloreactivity and increases survival in an allogeneic murine marrow transplantation model. Blood. 118:5031–5039. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Li X, Xu F, Chang C, Byon J, Papayannopoulou T, Deeg HJ and Marcondes AM: Transcriptional regulation of miR-10a/b by TWIST-1 in myelodysplastic syndromes. Haematologica. 98:414–419. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Li X, Marcondes AM, Ragoczy T, Telling A and Deeg HJ: Effect of intravenous coadministration of human stroma cell lines on engraftment of long-term repopulating clonal myelodysplastic syndrome cells in immunodeficient mice. Blood Cancer J. 3:e1132013. View Article : Google Scholar : PubMed/NCBI

13 

Li X and Deeg HJ: Murine xenogeneic models of myelodysplastic syndrome: An essential role for stroma cells. Exp Hematol. 42:4–10. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Kerbauy DM, Lesnikov V, Torok-Storb B, Bryant E and Deeg HJ: Engraftment of distinct clonal MDS-derived hematopoietic precursors in NOD/SCID-beta2-microglobulin-deficient mice after intramedullary transplantation of hematopoietic and stromal cells. Blood. 104:2202–2203. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Li M, Song L and Qin X: Glycan changes: Cancer metastasis and anti-cancer vaccines. J Biosci. 35:665–673. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Li X, Wang X, Tan Z, Chen S and Guan F: Role of glycans in cancer cells undergoing epithelial-mesenchymal transition. Front Oncol. 6:332016. View Article : Google Scholar : PubMed/NCBI

17 

Dube DH and Bertozzi CR: Glycans in cancer and inflammation-potential for therapeutics and diagnostics. Nat Rev Drug Discov. 4:477–488. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Groux-Degroote S, Guérardel Y and Delannoy P: Gangliosides: Structures, biosynthesis, analysis, and roles in cancer. Chembiochem. 18:1146–1154. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Timoshenko AV, Lanteigne J and Kozak K: Extracellular stress stimuli alter galectin expression profiles and adhesion characteristics of HL-60 cells. Mol Cell Biochem. 413:137–143. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Le Marer N: GALECTIN-3 expression in differentiating human myeloid cells. Cell Biol Int. 24:245–251. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Wan SY, Zhang TF and Ding Y: Galectin-3 enhances proliferation and angiogenesis of endothelial cells differentiated from bone marrow mesenchymal stem cells. Transplant Proc. 43:3933–3938. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Mohyeldin A, Garzón-Muvdi T and Quiñones-Hinojosa A: Oxygen in stem cell biology: A critical component of the stem cell niche. Cell Stem Cell. 7:150–161. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Wenger RH: Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression. FASEB J. 16:1151–1162. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Semenza GL: Intratumoral hypoxia, radiation resistance, and HIF-1. Cancer Cell. 5:405–406. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 3:721–732. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Danet GH, Pan Y, Luongo JL, Bonnet DA and Simon MC: Expansion of human SCID-repopulating cells under hypoxic conditions. J Clin Invest. 112:126–135. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Li X, Wan T, Zhang S, Li D and Han X: Quantitative proteomic analysis and comparison of two bone marrow stromal cell lines using the SILAC method. Exp Hematol. 44:1059–1071. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Li X, Li D, Pang X, Yang G, Deeg HJ and Guan F: Quantitative analysis of glycans, related genes, and proteins in two human bone marrow stromal cell lines using an integrated strategy. Exp Hematol. 43:760–769.e7. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Tan Z, Lu W, Li X, Yang G, Guo J, Yu H, Li Z and Guan F: Altered N-Glycan expression profile in epithelial-to-mesenchymal transition of NMuMG cells revealed by an integrated strategy using mass spectrometry and glycogene and lectin microarray analysis. J Proteome Res. 13:2783–2795. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Guan F, Tan Z, Li X, Pang X, Zhu Y, Li D and Yang G: A lectin-based isolation/enrichment strategy for improved coverage of N-glycan analysis. Carbohydr Res. 416:7–13. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Ying SX, Seal S, Abbassi N, Hockenbery DM, Kiem HP, Li X, Pagel JM, Gopal AK and Deeg HJ: Differential effects of bexarotene on intrinsic and extrinsic pathways in TRAIL-induced apoptosis in two myeloid leukemia cell lines. Leuk Lymphoma. 48:1003–1014. 2007. View Article : Google Scholar : PubMed/NCBI

33 

de Thonel A, Bettaïeb A, Jean C, Laurent G and Quillet-Mary A: Role of protein kinase C zeta isoform in Fas resistance of immature myeloid KG1a leukemic cells. Blood. 98:3770–3777. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Lopez-Sánchez LM, Jimenez C, Valverde A, Hernandez V, Peñarando J, Martinez A, Lopez-Pedrera C, Muñoz-Castañeda JR, De la Haba-Rodríguez JR, Aranda E and Rodriguez-Ariza A: CoCl2, a mimic of hypoxia, induces formation of polyploid giant cells with stem characteristics in colon cancer. PLoS One. 9:e991432014. View Article : Google Scholar : PubMed/NCBI

35 

Guan F, Schaffer L, Handa K and Hakomori SI: Functional role of gangliotetraosylceramide in epithelial-to-mesenchymal transition process induced by hypoxia and by TGF-{beta}. FASEB J. 24:4889–4903. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Ohtsuka T, Ryu H, Minamishima YA, Macip S, Sagara J, Nakayama KI, Aaronson SA and Lee SW: ASC is a Bax adaptor and regulates the p53-Bax mitochondrial apoptosis pathway. Nat Cell Biol. 6:121–128. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Marchenko ND and Moll UM: Mitochondrial death functions of p53. Mol Cell Oncol. 1:e9559952014. View Article : Google Scholar : PubMed/NCBI

38 

Etxebarria J and Reichardt NC: Methods for the absolute quantification of N-glycan biomarkers. Biochim Biophys Acta. 1860:1676–1687. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Korn C and Méndez-Ferrer S: Myeloid malignancies and the microenvironment. Blood. 129:811–822. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Velasco-Hernandez T, Hyrenius-Wittsten A, Rehn M, Bryder D and Cammenga J: HIF-1α can act as a tumor suppressor gene in murine acute myeloid leukemia. Blood. 124:3597–3607. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Drolle H, Wagner M, Vasold J, Kütt A, Deniffel C, Sotlar K, Sironi S, Herold T, Rieger C and Fiegl M: Hypoxia regulates proliferation of acute myeloid leukemia and sensitivity against chemotherapy. Leuk Res. 39:779–785. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Kuschel A, Simon P and Tug S: Functional regulation of HIF-1α under normoxia-is there more than post-translational regulation? J Cell Physiol. 227:514–524. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Liedtke C, Gröger N, Manns MP and Trautwein C: Interferon-alpha enhances TRAIL-mediated apoptosis by up-regulating caspase-8 transcription in human hepatoma cells. J Hepatol. 44:342–349. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Ruiz-Ruiz C, Ruiz de Almodóvar C, Rodríguez A, Ortiz-Ferrón G, Redondo JM and López-Rivas A: The up-regulation of human caspase-8 by interferon-gamma in breast tumor cells requires the induction and action of the transcription factor interferon regulatory factor-1. J Biol Chem. 279:19712–19720. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Gomyo Y, Sasaki J, Branch C, Roth JA and Mukhopadhyay T: 5-aza-2′-deoxycytidine upregulates caspase-9 expression cooperating with p53-induced apoptosis in human lung cancer cells. Oncogene. 23:6779–6787. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Chen CY, Jan YH, Juan YH, Yang CJ, Huang MS, Yu CJ, Yang PC, Hsiao M, Hsu TL and Wong CH: Fucosyltransferase 8 as a functional regulator of nonsmall cell lung cancer. Proc Natl Acad Sci USA. 110:630–635. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Xu Q, Isaji T, Lu Y, Gu W, Kondo M, Fukuda T, Du Y and Gu J: Roles of N-acetylglucosaminyltransferase III in epithelial-to-mesenchymal transition induced by transforming growth factor β1 (TGF- β1) in epithelial cell lines. J Biol Chem. 287:16563–16574. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Takahashi M, Kizuka Y, Ohtsubo K, Gu J and Taniguchi N: Disease-associated glycans on cell surface proteins. Mol Aspects Med. 51:56–70. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Pinho SS, Figueiredo J, Cabral J, Carvalho S, Dourado J, Magalhães A, Gärtner F, Mendonfa AM, Isaji T, Gu J, et al: E-cadherin and adherens-junctions stability in gastric carcinoma: Functional implications of glycosyltransferases involving N-glycan branching biosynthesis, N-acetylglucosaminyltransferases III and V. Biochim Biophys Acta. 1830:2690–2700. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Khare V, Lang M, Dammann K, Campregher C, Lyakhovich A and Gasche C: Modulation of N-glycosylation by mesalamine facilitates membranous E-cadherin expression in colon epithelial cells. Biochem Pharmacol. 87:312–320. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 40 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pang X, Wang Y, Zhang S, Tan Z, Guo J, Guan F and Li X: Altered susceptibility to apoptosis and N‑glycan profiles of hematopoietic KG1a cells following co‑culture with bone marrow‑derived stromal cells under hypoxic conditions. Oncol Rep 40: 1477-1486, 2018
APA
Pang, X., Wang, Y., Zhang, S., Tan, Z., Guo, J., Guan, F., & Li, X. (2018). Altered susceptibility to apoptosis and N‑glycan profiles of hematopoietic KG1a cells following co‑culture with bone marrow‑derived stromal cells under hypoxic conditions. Oncology Reports, 40, 1477-1486. https://doi.org/10.3892/or.2018.6548
MLA
Pang, X., Wang, Y., Zhang, S., Tan, Z., Guo, J., Guan, F., Li, X."Altered susceptibility to apoptosis and N‑glycan profiles of hematopoietic KG1a cells following co‑culture with bone marrow‑derived stromal cells under hypoxic conditions". Oncology Reports 40.3 (2018): 1477-1486.
Chicago
Pang, X., Wang, Y., Zhang, S., Tan, Z., Guo, J., Guan, F., Li, X."Altered susceptibility to apoptosis and N‑glycan profiles of hematopoietic KG1a cells following co‑culture with bone marrow‑derived stromal cells under hypoxic conditions". Oncology Reports 40, no. 3 (2018): 1477-1486. https://doi.org/10.3892/or.2018.6548