Open Access

Telomere length is positively associated with the expression of IL‑6 and MIP‑1α in bone marrow mesenchymal stem cells of multiple myeloma

  • Authors:
    • Shengli Li
    • Yang Jiang
    • Ai Li
    • Xiaoli Liu
    • Xiangling Xing
    • Yanan Guo
    • Yaqi Xu
    • Yunliang Hao
    • Chengyun Zheng
  • View Affiliations

  • Published online on: June 29, 2017     https://doi.org/10.3892/mmr.2017.6885
  • Pages: 2497-2504
  • Copyright: © Li 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

Potential roles of mesenchymal stem cells (MSCs) in the pathogenesis of multiple myeloma (MM) are largely unknown. In the current study, the authors analyzed telomere length and the expressions of interleukin (IL)‑6 and macrophage inflammatory protein (MIP)‑1α in MSCs derived from the bone marrow (BM) of MM patients and controls. The current results demonstrated that there was no significant difference in cell surface expression of CD73 and CD90, and the capacity to differentiate into bone tissue were identified between the BM MSCs derived from MM patients and controls. Interestingly, telomere length (TL) and mRNA expressions of IL‑6 and MIP‑1α were significantly longer or higher in BM MSCs of MM than those of controls. Moreover, TL is positively associated with the expressions of IL‑6 and MIP‑1α at the mRNA level in BM MSCs of MM. Additionally, IL‑6 and MIP‑1α expression were significantly upregulated when MSCs from MM patients were cultured in the myeloma associated condition medium. The present study indicated that myeloma‑associated elongation of TL of BM MSCs may be a key element contributing to the increased IL‑6 and MIP‑1α expression, by which MSCs in the tumor microenvironment may facilitate MM and/or MM bone disease development.

Introduction

Multiple myeloma (MM) is an incurable B cell malignancy characterized by the clonal expansion of plasma cells within the bone marrow (BM). It is well established that the transformation of myeloma is not only dependent on genetic and epigenetic aberrations, but also on the interaction between the malignant plasma cells and the BM microenvironment (1,2). The BM microenvironment is composed of a noncellular compartment including the extracellular matrix and the liquid milieu, and cellular compartments of mesenchymal origin (3). Myeloma cells modify this microenvironment through their production of regulatory factors and direct cell-to-cell contact, to create a milieu that promotes myeloma cell survival (46) and augments osteoclast (OC) recruitment and OC mediated bone loss (7,8).

As an essential cell type in the BM microenvironment, bone marrow mesenchymal stem cell (MSCs) is a type of adult stem cell endowed with self-renew and differentiation capacities (9). Abnormalities of MSCs derived from MM patients (MM-MSCs) have been described in previous studies (1012). Compared with MSCs isolated from healthy donors, increased expressions of cytokines and extracellular matrix were detected in MM-MSCs (11,13,14). Additionally, decreased osteogenic potentiality and reduced efficiency of suppression of T-cell proliferation were also described (1517). As well, MM-MSCs exhibited an abnormal upregulation of microRNA-135b and promoted MM tumor growth (18,19). Moreover, MM-MSCs demonstrated non-recurrent genomic imbalances and hypomethylation of some genomic region resulted in upregulated several microRNAs, which are involved in regulation of senescence status and cell cycle characteristics of MM-MSCs (12,20).

Telomeres are composed of repetitive DNA sequences and associated proteins at the end of the chromosomes. They serve a dominant role in protecting the end of DNA from fusion, recombination and degradation (21). Telomeric DNA shortens itself with each cell division due to the end-replication limitation of chromosomes (22). Correlation is reported between telomere length and replicative capacity of human fibroblasts (23). However, like most normal somatic cells, telomerase activity has not been detected in human MSCs (24). Telomeres of MSCs shortens its length concomitantly with the increased age or cell division (25,26). Moreover, dysfunction of telomeres impairs function of MSCs and alters their gene expressions (26,27).

In the current study, the authors compared the telomere length (TL) in bone marrow MSCs from MM patients and age-matched controls. The results demonstrated that the TL in MM-MSCs was longer than that in controls, and MM-MSCs secreted higher levels of interleukin (IL)-6 and macrophage inflammatory protein (MIP)-1α than the controls. Moreover, the results indicated that the telomere length of MM-MSCs was closely correlated with the expressions of IL-6 or MIP-1α.

Materials and methods

Cell culture

The human myeloma cells RPMI-8226 were cultured in RPMI-1640 medium (Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) containing 10% fetal bovine serum with the addition of 2 mM L-glutamine (both from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and antibiotics (100 U/ml penicillin and 0.1 mg/ml streptomycin) in a humid atmosphere at 37°C and 5% CO2. After cell culture for 24 h, the conditioned medium of RPMI-8226 was collected and stored in −20°C.

Isolation and expansion of MSCs from bone marrows

Bone marrow (BM) samples were obtained from patients with MM or from controls (patients without a tumor, infection or autoimmune diseases; Table I). The current study was approved by the Ethics Committee of the Second Hospital of Shandong University (Jinan, China) and informed consent was obtained from all patients. Mononuclear cells (MNCs) were obtained from BM samples by density gradient separetion using Ficoll density gradient centrifugation. Finally, MNCs were cultured in Dulbecco's modified Eagle's medium (Hyclone; GE Healthcare Life Sciences) supplemented with 10% defined fetal bovine serum (Shanghai ExCell Biology, Inc., Shanghai, China), growth factors (R&D Systems, Inc., Minneapolis, MN, USA). At 72 or 96 h incubation, non-adherent cells were carefully removed, whereas the MSCs attached to the cell culture plate were defined as passage 0 (P0) MSCs. When the MSCs reached approximately 80% confluence, the cells were passaged (P1) by digestion using 0.05% trypsin-EDTA (Thermo Fisher Scientific, Inc.) The MSCs were collected near confluency at P4 for subsequent experiments.

Table I.

Clinical characteristics of patients with multiple myeloma (n=12).

Table I.

Clinical characteristics of patients with multiple myeloma (n=12).

SampleGenderAge (years)Disease stageaβ2-MG (mg/dl)BM plasma cells (%)Bone lesions
  1F68Early3.3158N
  2M65Early3.3651N
  3F69Late11.567Y
  4F63Early3.7451Y
  5M65Late12.465Y
  6M74Late6.7248N
  7M58Late7.7932Y
  8M57Late40.540Y
  9M59Late7.89  4Y
10M70Late943Y
11M58Late20.820N
12F59Early4.2532Y

a Early, I and II; late, III. ISS staging system was used. N, no; Y, yes; M, male; F, female; β2-MG, β2-microglobulin; BM, bone marrow.

Osteoblastic differentiation of MSCs

An osteocyte differentiation assay was performed according to the instructions provided in the StemPro® Osteogenesis Differentiation kit (cat no. A1007201; Thermo Fisher Scientific, Inc.). Briefly, MSCs were seeded at 1×104/cm2 in 6-well culture plates in osteoblast differentiation culture media for 21 days. Medium was changed 2–3 times a week. The osteocytes were stained with Alizarin red (cat no. A5533; Sigma-Aldrich, Merck KGaA, Darmstadt, Germany).

Immunophenotyping of bone marrow MSCs

The bone marrow MSCs were labeled with phycoerythrin-conjugated anti-CD73 (cat no. 550257), fluorescein isothiocyanate (FITC)-conjugated anti-CD90 (cat no. 555595), FITC-conjugated anti-CD105 (cat no. 561443) (all from BD Biosciences, Franklin Lakes, NJ, USA) and human leukocyte antigen G (HLA-G; cat no. 12-9957-73; eBioscience, Inc., San Diego, CA, USA). The fluorescence of the above MSC surface markers was detected and analyzed using flow cytometry with a BD FACSAria™ II system (BD Biosciences). Data were analyzed using the FlowJo software version 7.6 (FlowJo, LLC, Ashland, Oregon, USA).

Quantitative polymerase chain reaction (qPCR) and Flow-FISH for MSC telomere length assay

For qPCR detection, genomic DNA was isolated using QiAamp DNA Blood Mini kit (Qiagen GmbH, Hilden, Germany). Telomere length was determined by qPCR as described previously (28). The primer sequences for human telomere (Tel) and for β-globin (HBG) are listed in Table II. The relative telomere length was decided as the percentage of that in a MM-MSC. Flow-FISH of MSCs was performed as described previously (29).

Table II.

The primer sequences for quantitative polymerase chain reaction and telomere length assessment.

Table II.

The primer sequences for quantitative polymerase chain reaction and telomere length assessment.

GeneSequence (5′-3′)
1IL-6 forward GAGGCACTGGCAGAAAACAACC
IL-6 reverse CCTCAAACTCCAAAAGACCAGTGATG
2MIP-1α forward GAATCATGCAGGTCTCCACTG
MIP-1α reverse CTCTAGGTCGCTGACATATTTC
3IDO forward TGGGGCAAAGGTCATGGAG
IDO reverse TTTCTTGGAGAGTTGGCAGTAAG
4hTERT forward CGGAAGAGTGTCTGGAGCAA
hTERT reverseGGATGA AGCGGAGTCTGGA
5GAPDH forward GCACCGTCAAGGCTGAGAAC
GAPDH reverse TGGTGAAGACGCCAGTGGA
6Tel1b CGGTTTGTTTGGGTTTGGGT-TTGGGTTTGGGTTTGGGTT
Tel 2b GGCTTGCCTTACCCTTACCCTTACCC-TTACCCTTACCCT
7HBG3 TGTGCTGGCCCATCACTTTG
HBG4 ACCAGCCACCACTTTCTGATAGG

[i] hTERT, human telomerase reverse transcriptase; IL, interleukin; MIP-1α, macrophage inflammatory protein-1α.

Cell cycle analysis

Cell cycle analysis was performed using the Cell Cycle and Apoptosis Analysis kit (cat no. C1052-1; Beyotime Institute of Biotechnology, Haimen, China) and flow cytometry with a BD FACSAria™ II system (BD Biosciences) was conducted following the manufacturer's instructions. The percentage of each phase in the cell cycle was calculated using cell cycle analysis software Modfit version LT 3.1 (Verity Software House, Topsham, ME, USA).

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

Total RNA was extracted using TRIzol reagent (Thermo Fisher Scientific, Inc.) following the manufacturer's instruction. cDNA was synthesized using the M-MLV enzyme (Thermo Fisher Scientific, Inc.) and an oligo-dT primer according to the protocol. qPCR was performed using SYBR-Green kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) on an ABI 7700 detector (Applied Biosystems; Thermo Fisher Scientific, Inc.). All sequences of primers are listed in Table II. Thermocycling conditions were as follows: At 50°C for 2 min, at 95°C for 10 min, followed by 45 cycles at 95°C for 15 sec and at 60°C for 1 min. The expression level of each gene was calculated according to the comparative Cq method and normalized to GAPDH expression (30).

Statistical analysis

The Student's t-test, paired t-test or Fisher's exact test were used to compare differences between groups. Pearson's correlation was applied to analysis for relationships between variables. All the results were presented using GraphPad Prism software (version, 5; GraphPad Software, Inc., La Jolla, CA, USA). The data are shown as mean ± standard error of the mean P<0.05 was considered to indicate a statistically significant difference.

Results

The phenotype, growth characteristics and osteoblastic differentiation of MSCs

As presented in Fig. 1, MSCs were cultured and analyzed of both MM patients (Fig. 1A) and the control group (Fig. 1B). The phenotypes were analyzed using flow cytometry. As presented in Table III, MSCs from both MM patients and controls were positive for CD73, CD90 and CD105, but negative for HLA-G. There were no significant differences between MM patients (6.08±0.69 days) and controls (8.62±1.33 days) on the days for obtaining 80% confluent MSCs cultures. The osteoblastic differentiation of MSCs was examined using Alizarin red staining. As indicated in Fig. 2, MSCs from three age-matched pairs of MM patients (Fig. 2A) and controls (Fig. 2B) displayed deposition of matrix mineralization.

Table III.

Flow cytometry analysis of mesenchymal stem cells from control and MM patients.

Table III.

Flow cytometry analysis of mesenchymal stem cells from control and MM patients.

Antigen ExpressionMM patients (MEAN ± SEM)Control group (MEAN ± SEM) P-valuea
CD73 (%)90.9±0.0389.5±0.030.73
CD90 (%)75.4±0.0683.4±0.050.39
CD105 (%)62.6±10.459±80.81
HLA-G (%)1.1±4.40.9±0.50.77

a Comparison between patient group and control. CD, cluster of differentiation; HLA-G, human leukocyte antigen G; MM, multiple myeloma; SEM, standard error of the mean.

The TL and cell cycle of MSCs from MM patients and control group

The TL of the MSCs from 12 MM patients and 8 controls were compared by qPCR. As reported in Fig. 3A, the TL of MSCs from MM patients was significantly longer (0.98±0.11) than the controls (0.65±0.07; P=0.03). The authors further assessed the TL in MSCs using Flow-FISH. Similarly, the TL of MSCs was longer in the MM patients (10,860±807) than in the controls (5,227±1,143; P=0.015; Fig. 3B). Cell cycle characteristics of MM-MSCs were determined using flow cytometry. As presented in Fig. 3C, the percentage of MM-MSCs in G0/G1 was increased compared with controls (77.33±1.83 vs. 61.87±2.51; P<0.01), whereas the MM-MSCs in the S phase and G2/M phase fell to 14.36±1.36% (vs. 26.98±3.22; P<0.01) and 6.34±0.75% (vs. 10.95±1.14; P<0.01) respectively, when compared to those of controls.

The human telomerase catalytic subunit gene hTERT, which is the putative human telomerase catalytic subunit gene serving as an indicator of telomerase activity (31), were also measured using RT-qPCR method in normal and MM-MSCs. However, hTERT expression in those cells was not detected (data not shown). These results indicated that both normal and MM-MSCs lacked telomerase activity.

The expressions of IL-6, indoleamine 2,3-dioxygenase (IDO) and MIP-1α in MSCs from MM patients

The mRNA expressions of IL-6 and IDO in MSCs from MM patients and controls were investigated using RT-qPCR. The expression of IL-6 was significantly increased in MM-MSCs (P<0.05), while the expression of IDO decreased obviously compared with MSCs in control group (P<0.05; Fig. 3D). Moreover, the expression rate of MIP-1α from MM-MSCs was significantly higher than that from MSCs in control group (91.7 and 37.5% respectively, P<0.05).

The correlations between TL and IL-6, MIP-1α and IDO

As presented in Fig. 4, the TL of MM-MSCs was correlated with their expression of IL-6 (Fig. 4A) and MIP-1α (Fig. 4B). However, no significant correlation between the telomere length and the expression of IDO in MM-MSCs was found.

The conditioned medium of RPMI 8226 induces changes of gene expression and cell cycle in MSCs

MSCs isolated from three MM patients were cultured in myeloma condition culture medium (MCCM) (a mixture of myeloma cell line RPMI-8226 culture supernatant and MSCs complete medium) for 24 h, then the expressions of IL-6, MIP-1α and IDO were quantified using qPCR. When cultured in the MCCM, IL-6 (P<0.05) and MIP-1α (P<0.01) were significantly increased, while IDO was markedly downregulated (P<0.05), when compared with the MSCs cultured in regular MSC medium (Fig. 5A).

To explore whether MCCM affects the cell cycle of MM-MSCs, MSCs from three MM patients were cultured in MCCM. At 24 h culture, MSCs reported an increase of cells in G0/G1 phase and a decrease of the cells in S phase compared to the controls (P<0.05; Fig. 5B).

The changes of MSCs were not correlated with β2 microglobulin and bone marrow plasma cells

No significant correlations between TL and serum β2 microglobulin (β2-MG) (r=−0.24; P=0.45), and the percentage of plasma cell in BM (r=0.55; P=0.07) was identified. Moreover, these data did not present a correlation of IL-6 or MIP-1α expression with serum β2-MG, as well as the percentage of plasma cells in BM (data not shown).

Discussion

The direct and indirect interactions between BM-MSCs and MM cells not only mediate MM cell growth and survival, but also lead to the intrinsic abnormalities observed in MM-MSCs, such as genomic imbalances and an overexpression of IL-6 (32). In the present study, the authors indicated that bone marrow MSCs from MM are not different from control group in terms of phenotypic markers (e.g., CD73 and CD90) and capacity to differentiate into osteogenic tissues. However, the TL and expressions of IL-6 and MIP-1α were significantly increased in MM BM-MSCs than the controls. Moreover, the correlation between TL and IL-6 as well as MIP-1α expression in MM-MSCs were displayed.

The current results demonstrated that TL of MM-MSCs was significantly longer than that of controls, indicating a tumor or myeloma associated mechanism contributing to maintenance or elongation of TL. It is known that TLs shorten accordingly when cells divide. The authors' cell cycle analysis demonstrated that the percentage of MSCs in the G0/G1 phase was significantly increased in MM than in the controls, suggesting that there are more MM-MSCs in stable or non-dividing phase, which may in part explain why MM-MSCs present longer TL than the controls. In line with a previous report (24), these data suggested that MSCs from both controls and MM patients lacked telomerase activity. To date, the underlying the mechanisms of TL maintenance in MSCs is largely unknown. In MM cells lines, IL-6 and IGF-1 was demonstrated to increase telomerase activity via AKT-mediated phosphorylation of hTERT without alteration of the expression of hTERT at the level of mRNA or protein (33), indicating that MSCs may also apply this mechanism to maintain their TL in the microenvironment riched in inflammatory cytokines, such as IL-6.

Moreover, reduced percent of MSCs in S phase during cell cycle and increased in G0/G1 phase were identified following conditional culture. This result is in contrast with a recent report displaying that co-culture with MM cells produced a marked increase in S/G2/M phases of the cells (10). These differences may be due to divergences in the in vitro culture system applied or the myeloma condition medium, but not cell-cell direct contact in our system.

In the present study, the authors also compared MSC differentiation capacity between MM patients and controls, and did not identify markedly difference in differentiation into bone tissue between them. This result is controversial compared to the previous reports that indicate that the differentiation capacity of MSCs from MM patients is lower than those from the normal controls (19,34). It may be speculated that the primary reason causing the different results may be due to different cell passages applied in our and other studies. Since MSCs were used from passage 4, the in vitro culture or expansion procedures may overcome subtle function abnormalities of MM-MSCs. This issue will be addressed in future studies.

Changes in the expression of cytokines and growth factors from MSCs serve a prominent role in the growth and survival of multiple myeloma (1). High expression of IL-6 from MM-MSCs was detected. MM-MSCs were expressed a significantly higher level of IL-6 following culturing with the supernatants from MM cultures. Our findings are consistent with those previous reports that MM-MSCs overproduced IL-6 (35) and MM cells stimulated IL-6 expression in MSCs (4).

It has been demonstrated that MIP-1α contributes to the development of bone disease in MM via tumor survival promotion (36), osteoblast differentiation (7,8) and osteoblast functional inhibition (37). MIP-1α is indicated to be primarily produced by myeloma cells (8). Interestingly, MIP-1α was shown to also be expressed in MM-MSCs, and the expression of MIP-1α in MM-MSCs was higher than that in MSCs from the control. Moreover, MIP-1α expression in MM-MSCs was further increased under the stimulation of the supernatants isolated from MM cell line culture. Therefore, MIP-1α produced by MM-MSCs may also participate in the development of MM.

Previous studies have illustrated that the proliferative potential of human hematopoietic cells (38) and human fibroblasts (23) correlates with their telomere lengths. Similarly, the correlation between growth of MSCs and telomere length has also been suggested (39). Moreover, dysfunction of telomere impairs the function of mesenchymal progenitor cells and affects the expressions of various cytokines (27). Consistently, our findings of positive correlation of IL-6 and MIP-1α with telomere lengths in MM-MSCs highlight that telomere lengths affect cytokine expression in MM-MSCs, by which MM-MSCs facilitate MM development.

IDO is an important immunoregulatory molecule (40). Our results demonstrated that IDO was significantly lower in MM-MSCs than controls. Supernatants derived from MM cell line further downregulated IDO expression. The IDO expression may result in impaired immunomodulatory capacities of BM-MSCs in MM (15,17). Undoubtedly, the significance of such decreased IDO expression in MM-MSCs needs to be investigated in the future studies.

In summary, in the present study, the authors find that MM-MSCs present longer telomere and higher IL-6 and MIP-1α expression, and telomere lengths are positively correlated with IL-6 and MIP-1α expression. These results indicated that MSCs in BM may be involved in pathothegenesis of MM and MM related bone diseases.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant nos. 81372545 and 81600176), the Fundamental Research Funds of Shandong University (grant no. 2014QY004-16 and 2015QY002-10), Youth Fund of Second Hospital of Shandong University (grant no. Y2013010029), the Natural Science Foundation of Shandong Province (grant no. ZR2016HB71) and the Shenzhen Strategic Emerging Industry Development Special Fund (grant no. JCYJ20150402105524048).

References

1 

Bianchi G and Munshi NC: Pathogenesis beyond the cancer clone(s) in multiple myeloma. Blood. 125:3049–3058. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Hideshima T, Mitsiades C, Tonon G, Richardson PG and Anderson KC: Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer. 7:585–598. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Manier S, Sacco A, Leleu X, Ghobrial IM and Roccaro AM: Bone marrow microenvironment in multiple myeloma progression. J Biomed Biotechnol. 2012:1574962012. View Article : Google Scholar : PubMed/NCBI

4 

Dankbar B, Padró T, Leo R, Feldmann B, Kropff M, Mesters RM, Serve H, Berdel WE and Kienast J: Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood. 95:2630–2636. 2000.PubMed/NCBI

5 

Gupta D, Treon SP, Shima Y, Hideshima T, Podar K, Tai YT, Lin B, Lentzsch S, Davies FE, Chauhan D, et al: Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: Therapeutic applications. Leukemia. 15:1950–1961. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Nefedova Y, Cheng P, Alsina M, Dalton WS and Gabrilovich DI: Involvement of Notch-1 signaling in bone marrow stroma-mediated de novo drug resistance of myeloma and other malignant lymphoid cell lines. Blood. 103:3503–3510. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Han JH, Choi SJ, Kurihara N, Koide M, Oba Y and Roodman GD: Macrophage inflammatory protein-1alpha is an osteoclastogenic factor in myeloma that is independent of receptor activator of nuclear factor kappaB ligand. Blood. 97:3349–3353. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Abe M, Hiura K, Wilde J, Moriyama K, Hashimoto T, Ozaki S, Wakatsuki S, Kosaka M, Kido S, Inoue D and Matsumoto T: Role for macrophage inflammatory protein (MIP)-1alpha and MIP-1beta in the development of osteolytic lesions in multiple myeloma. Blood. 100:2195–2202. 2002.PubMed/NCBI

9 

Bergfeld SA and DeClerck YA: Bone marrow-derived mesenchymal stem cells and the tumor microenvironment. Cancer Metastasis Rev. 29:249–261. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Kassen D, Moore S, Percy L, Herledan G, Bounds D, Rodriguez-Justo M, Croucher P and Yong K: The bone marrow stromal compartment in multiple myeloma patients retains capability for osteogenic differentiation in vitro: Defining the stromal defect in myeloma. Br J Haematol. 167:194–206. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Noll JE, Williams SA, Tong CM, Wang H, Quach JM, Purton LE, Pilkington K, To LB, Evdokiou A, Gronthos S and Zannettino AC: Myeloma plasma cells alter the bone marrow microenvironment by stimulating the proliferation of mesenchymal stromal cells. Haematologica. 99:163–171. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Berenstein R, Blau O, Nogai A, Waechter M, Slonova E, Schmidt-Hieber M, Kunitz A, Pezzutto A, Doerken B and Blau IW: Multiple myeloma cells alter the senescence phenotype of bone marrow mesenchymal stromal cells under participation of the DLK1-DIO3 genomic region. BMC Cancer. 15:682015. View Article : Google Scholar : PubMed/NCBI

13 

Wallace SR, Oken MM, Lunetta KL, Panoskaltsis-Mortari A and Masellis AM: Abnormalities of bone marrow mesenchymal cells in multiple myeloma patients. Cancer. 91:1219–1230. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Corre J, Labat E, Espagnolle N, Hébraud B, Avet-Loiseau H, Roussel M, Huynh A, Gadelorge M, Cordelier P, Klein B, et al: Bioactivity and prognostic significance of growth differentiation factor GDF15 secreted by bone marrow mesenchymal stem cells in multiple myeloma. Cancer Res. 72:1395–1406. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Arnulf B, Lecourt S, Soulier J, Ternaux B, Lacassagne MN, Crinquette A, Dessoly J, Sciaini AK, Benbunan M, Chomienne C, et al: Phenotypic and functional characterization of bone marrow mesenchymal stem cells derived from patients with multiple myeloma. Leukemia. 21:158–163. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Li B, Shi M, Li J, Zhang H, Chen B, Chen L, Gao W, Giuliani N and Zhao RC: Elevated tumor necrosis factor-alpha suppresses TAZ expression and impairs osteogenic potential of Flk-1+ mesenchymal stem cells in patients with multiple myeloma. Stem Cells Dev. 16:921–930. 2007. View Article : Google Scholar : PubMed/NCBI

17 

André T, Najar M, Stamatopoulos B, Pieters K, Pradier O, Bron D, Meuleman N and Lagneaux L: Immune impairments in multiple myeloma bone marrow mesenchymal stromal cells. Cancer Immunol Immunother. 64:213–224. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Roccaro AM, Sacco A, Maiso P, Azab AK, Tai YT, Reagan M, Azab F, Flores LM, Campigotto F, Weller E, et al: BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest. 123:1542–1555. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Xu S, Santini Cecilia G, De Veirman K, Vande Broek I, Leleu X, De Becker A, Van Camp B, Vanderkerken K and Van Riet I: Upregulation of miR-135b is involved in the impaired osteogenic differentiation of mesenchymal stem cells derived from multiple myeloma patients. PLoS One. 8:e797522013. View Article : Google Scholar : PubMed/NCBI

20 

Garayoa M, Garcia JL, Santamaria C, Garcia-Gomez A, Blanco JF, Pandiella A, Hernández JM, Sanchez-Guijo FM, del Cañizo MC, Gutiérrez NC and Miguel San JF: Mesenchymal stem cells from multiple myeloma patients display distinct genomic profile as compared with those from normal donors. Leukemia. 23:1515–1527. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Blackburn EH: Structure and function of telomeres. Nature. 350:569–573. 1991. View Article : Google Scholar : PubMed/NCBI

22 

Harley CB, Futcher AB and Greider CW: Telomeres shorten during ageing of human fibroblasts. Nature. 345:458–460. 1990. View Article : Google Scholar : PubMed/NCBI

23 

Allsopp RC, Vaziri H, Patterson C, Goldstein S, Younglai EV, Futcher AB, Greider CW and Harley CB: Telomere length predicts replicative capacity of human fibroblasts. Proc Natl Acad Sci USA. 89:10114–10118. 1992. View Article : Google Scholar : PubMed/NCBI

24 

Zimmermann S, Voss M, Kaiser S, Kapp U, Waller CF and Martens UM: Lack of telomerase activity in human mesenchymal stem cells. Leukemia. 17:1146–1149. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Choumerianou DM, Martimianaki G, Stiakaki E, Kalmanti L, Kalmanti M and Dimitriou H: Comparative study of stemness characteristics of mesenchymal cells from bone marrow of children and adults. Cytotherapy. 12:881–887. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Banfi A, Bianchi G, Notaro R, Luzzatto L, Cancedda R and Quarto R: Replicative aging and gene expression in long-term cultures of human bone marrow stromal cells. Tissue Eng. 8:901–910. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Ju Z, Jiang H, Jaworski M, Rathinam C, Gompf A, Klein C, Trumpp A and Rudolph KL: Telomere dysfunction induces environmental alterations limiting hematopoietic stem cell function and engraftment. Nat Med. 13:742–747. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Cawthon RM: Telomere measurement by quantitative PCR. Nucleic Acids Res. 30:e472002. View Article : Google Scholar : PubMed/NCBI

29 

Ci X, Li B, Ma X, Kong F, Zheng C, Björkholm M, Jia J and Xu D: Bortezomib-mediated down-regulation of telomerase and disruption of telomere homeostasis contributes to apoptosis of malignant cells. Oncotarget. 6:38079–38092. 2015. View Article : Google Scholar : PubMed/NCBI

30 

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 : PubMed/NCBI

31 

Meyerson M, Counter CM, Eaton EN, Ellisen LW, Steiner P, Caddle SD, Ziaugra L, Beijersbergen RL, Davidoff MJ, Liu Q, et al: hEST2, the putative human telomerase catalytic subunit gene, is up-regulated in tumor cells and during immortalization. Cell. 90:785–795. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Reagan MR and Ghobrial IM: Multiple myeloma mesenchymal stem cells: Characterization, origin, and tumor-promoting effects. Clin Cancer Res. 18:342–349. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Akiyama M, Hideshima T, Hayashi T, Tai YT, Mitsiades CS, Mitsiades N, Chauhan D, Richardson P, Munshi NC and Anderson KC: Cytokines modulate telomerase activity in a human multiple myeloma cell line. Cancer Res. 62:3876–3882. 2002.PubMed/NCBI

34 

Bolzoni M, Donofrio G, Storti P, Guasco D, Toscani D, Lazzaretti M, Bonomini S, Agnelli L, Capocefalo A, Dalla Palma B, et al: Myeloma cells inhibit non-canonical wnt co-receptor ror2 expression in human bone marrow osteoprogenitor cells: Effect of wnt5a/ror2 pathway activation on the osteogenic differentiation impairment induced by myeloma cells. Leukemia. 27:451–463. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Li B, Fu J, Chen P and Zhuang W: Impairment in immunomodulatory function of mesenchymal stem cells from multiple myeloma patients. Arch Med Res. 41:623–633. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Lentzsch S, Gries M, Janz M, Bargou R, Dörken B and Mapara MY: Macrophage inflammatory protein 1-alpha (MIP-1 alpha) triggers migration and signaling cascades mediating survival and proliferation in multiple myeloma (MM) cells. Blood. 101:3568–3573. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Vallet S, Pozzi S, Patel K, Vaghela N, Fulciniti MT, Veiby P, Hideshima T, Santo L, Cirstea D, Scadden DT, et al: A novel role for CCL3 (MIP-1α) in myeloma-induced bone disease via osteocalcin downregulation and inhibition of osteoblast function. Leukemia. 25:1174–1181. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Van Ziffle JA, Baerlocher GM and Lansdorp PM: Telomere length in subpopulations of human hematopoietic cells. Stem Cells. 21:654–660. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Samsonraj RM, Raghunath M, Hui JH, Ling L, Nurcombe V and Cool SM: Telomere length analysis of human mesenchymal stem cells by quantitative PCR. Gene. 519:348–355. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Ge W, Jiang J, Arp J, Liu W, Garcia B and Wang H: Regulatory T-cell generation and kidney allograft tolerance induced by mesenchymal stem cells associated with indoleamine 2,3-dioxygenase expression. Transplantation. 90:1312–1320. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 16 Issue 3

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
Li S, Jiang Y, Li A, Liu X, Xing X, Guo Y, Xu Y, Hao Y and Zheng C: Telomere length is positively associated with the expression of IL‑6 and MIP‑1α in bone marrow mesenchymal stem cells of multiple myeloma. Mol Med Rep 16: 2497-2504, 2017
APA
Li, S., Jiang, Y., Li, A., Liu, X., Xing, X., Guo, Y. ... Zheng, C. (2017). Telomere length is positively associated with the expression of IL‑6 and MIP‑1α in bone marrow mesenchymal stem cells of multiple myeloma. Molecular Medicine Reports, 16, 2497-2504. https://doi.org/10.3892/mmr.2017.6885
MLA
Li, S., Jiang, Y., Li, A., Liu, X., Xing, X., Guo, Y., Xu, Y., Hao, Y., Zheng, C."Telomere length is positively associated with the expression of IL‑6 and MIP‑1α in bone marrow mesenchymal stem cells of multiple myeloma". Molecular Medicine Reports 16.3 (2017): 2497-2504.
Chicago
Li, S., Jiang, Y., Li, A., Liu, X., Xing, X., Guo, Y., Xu, Y., Hao, Y., Zheng, C."Telomere length is positively associated with the expression of IL‑6 and MIP‑1α in bone marrow mesenchymal stem cells of multiple myeloma". Molecular Medicine Reports 16, no. 3 (2017): 2497-2504. https://doi.org/10.3892/mmr.2017.6885