Effects of hypoxia‑inducible factor‑1α on the proliferation and apoptosis of human synovial mesenchymal stem cells

  • Authors:
    • Weiwei Zheng
    • Xueping Gu
    • Xingwei Sun
    • Dan Hu
  • View Affiliations

  • Published online on: September 9, 2019     https://doi.org/10.3892/mmr.2019.10656
  • Pages: 4315-4322
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hypoxia is a constant feature of the synovial microenvironment. How synovial mesenchymal stem cells (SMSCs) proliferate and differentiate in a hypoxic environment over a long period of time has aroused the interest of researchers. The aim of the present study was to explore the effects of hypoxia‑inducible factor‑1α (HIF‑1α) on the proliferation and apoptosis of human SMSCs. SMSCs were harvested and cultured under different concentration of oxygen, normoxia (21% O2), hypoxia (5% O2) and severe hypoxia (0.5% O2) to determine its effect on the expression of HIF‑1α. Then, the cells were collected and cell proliferation and apoptosis were detected at severe hypoxia (0.5% O2) and hypoxia (5% O2) conditions following HIF‑1α siRNA transfection. There were no significant changes in cellular proliferation or apoptosis when cultured in normoxia (21% O2), hypoxia (5% O2) or severe hypoxia (0.5% O2). However, the mRNA and protein expression of HIF‑1α were markedly upregulated in the hypoxic conditions. Further experiments suggested that the proliferation of SMSCs was obviously suppressed and apoptosis was markedly increased under severe hypoxic (0.5%) and hypoxic (5% O2) conditions following HIF‑1α siRNA transfection. In conclusion, HIF‑1α effectively improved the tolerance of SMSCs to hypoxia, which may promote cellular proliferation and prevent the apoptosis of SMSCs under hypoxic conditions.

Introduction

The study of mesenchymal stem cells (MSCs) has advanced considerably throughout the past few decades, and rapid progress has been made in utilizing MSCs to study disease pathogenesis, to discover biomarkers and novel targets, and to validate cell-based tissue engineering therapies (14). In the past few years, synovial mesenchymal stem cells (SMSCs) have attracted increasing attention as a promising cell source for tissue engineering as SMSCs have an intriguing multilineage developmental plasticity in vitro and in vivo (57). Synovial tissue can be obtained by minimally invasive surgery such as arthroscopy (8), and similar to adult MSCs from other sources, SMSCs can be isolated and expanded more efficiently in vitro (8,9). In addition to the ability to self-replicate and differentiate into multiple lineages, the immune-privileged nature of SMSCs implies their potential utilization in allogeneic cell-based settings (10,11). Articular synovium is avascular and exists in a low oxygen microenvironment. The oxygen tension ranges from 1 to 7% in the knee joints of different age groups (12,13). As oxygen tension seems to be a nonnegligible regulator of cell proliferation and differentiation of SMSCs, it is therefore essential to study the survival mechanism of SMSCs in a hypoxic environment.

Hypoxia-inducible factor-1α (HIF-1α) is a heterodimeric transcription factor that can be induced under hypoxic condition (14,15). The stimulation and activity of HIF-1α have been demonstrated to be mediated at different levels throughout the cell cycle (16). Some regulatory pathways, including Wnt, Notch, PTEN, JAK/STAT, LOX and FBW7 are involved in regulating the metabolism, angiogenesis, metastasis and invasion of MSCs via HIF-1α mediation (17). HIF-1α undergoes rapid degradation under normoxic conditions with a half-life of only approximately 5 min (18). In comparison, in hypoxic conditions, intermediate metabolites stabilize the expression of HIF-1α by inhibiting the activity of proliferol hydroxylase, and promotes its migration into the nucleus to combine with HIF-1β to form HIF-1 heterodimers (1921). Numerous studies have demonstrated that HIF-1α participates in the regulation of angiogenesis, cell growth and glucose metabolism (2224).

HIF-1α has been reported to be abundantly expressed in synovial tissues of patients with rheumatoid arthritis, osteoarthritis and temporomandibular joint disorders, which are stimulated by a series of immune factors such as inflammation and oxidative stress response (2527). However, the expression of HIF-1α in normal human synovial membrane and the role of HIF-1α in synovial membrane adapting to different oxygen environments have not been reported. In particular, the effect of low oxygen tension on HIF-1α expression in SMSCs has not been characterized. Therefore, in order to determine the initial effects of HIF-1α in SMSCs, we investigated the expression of HIF-1α in SMSCs under different oxygen conditions and observed the effect of HIF-1α on the proliferation and apoptosis of human SMSCs in vitro.

Materials and methods

Tissue harvest and cell culture

Synovial tissue from six patients with a spectrum of knee conditions including ligament, meniscal, and cartilage injury were collected by arthroscopy. Ethical approval for this study was granted by the Institution Review Board of the Affiliated Hospital of Nanjing Medical University, and all study participants were recruited after providing informed written consent. SMSCs were isolated using an enzyme digestion procedure according to a previously described method (28). The culture medium (DMEM/Nutrient Mixture F-12 Ham supplemented with 20% FBS) was changed every 3 days. According to the manufacturer's protocol, 1.0×106 SMSCs were cultured for 21 days in adipogenic medium (Adipogenic Base Media, StemXVivo; R&D Systems) for adipogenesis detected by Oil red-O staining. The same method was used for osteogenesis with osteogenic medium (Osteogenic Base Media, StemXVivo; R&D Systems) and Alizarin red staining was conducted.

Hypoxia exposure

The 2nd passage of 1.0×105 SMSCs was cultured for 7 days under different ambient oxygen tension, including normoxia (21% O2), hypoxia (5% O2) or severe hypoxia (0.5% O2) at 37°C in a humidified incubator with 5% CO2.

Cell transfection

The 2nd passage of 1.0×105 cells were transfected with small interfering RNA (siRNA) as previously described (29). SMSCs were placed into 6-well plates for 24 h. Cells were transfected with specific siRNA (Ambion; Thermo Fisher Scientific, Inc.) targeting HIF-1α using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). After transfection for 48 h, SMSCs were collected for further analysis.

Proliferation assay

A Cell Counting Kit-8 (CCK-8) assay was used to evaluate cell viability. Briefly, the SMSCs were collected and seeded into 96-well plates at a dose of 5.0×103/ml. Then, after cell culture for 1 to 7 days, 10 µl of CCK-8 solution (Nanjing Jiancheng Biotechnology Institute) was added into each well. Cells were cultured at room temperature for 4 h in the dark. The absorbance was measured at 450 nm by a microplate reader (Bio-Rad, Inc.).

Flow cytometry

To determine the phenotypes of the SMSCs, flow cytometric analysis was used. The 2nd passage of 1.0×106 SMSCs was collected and the cells were suspended in PBS before being incubated with the following antibodies (Agilent Technologies, Inc.) for 90 min at 37°C: FITC-conjugated anti-human CD147 (cat. no. SZB10284; 1:500 dilution), CD90 (cat. no. bs-10430R; 1:200 dilution), CD105 (cat. no. bs-0579R; 1:10,000 dilution), CD44 (cat. no. K001677P; 1:500 dilution), CD117 (cat. no. 130-098-570; 1:500 dilution), CD34 (cat. no. bs-0765R-2; 1:200 dilution), CD14 (cat. no. K101533P; 1:200 dilution) and CD45 (cat. no. bs-10600R; 1:200 dilution). The cell phenotypes were analyzed using an FC 500 flow cytometer (BD Biosciences).

RT-qPCR

TRIzol reagent (Thermo Fisher Scientific, Inc.) was used to extract total RNA of the SMSCs. The target gene and an endogenous control β-actin were amplified by qPCR using the SYBR Green PCR kit (Takara Biotechnology Co., Ltd., Dalian, China). GAPDH was used as an internal reference. The primers for PCR were as follows: GLUT3 forward, 5′-CGGCTTCCTCATTACCTTC-3′ and reverse, 5′-GGCACGACTTAGACATTGG-3′; HIF-1α forward, 5′-TAAAGGAATTTCAATATTTGATGGG-3′ and reverse, 5′-AAAGGGTAAAGAACAAAACACACAG-3′; GAPDH forward, 5′-GGAGCGAGATCCCTCCAAAAT-3′ and reverse, 5′-GGCTGTTGTCATACTTCTCATGG-3′. The thermocycling conditions were 25°C for 5 min, 42°C for 60 min and at 95°C for 15 sec. Fold changes were calculated using the 2−ΔΔCq method (30) normalized to GAPDH.

Western blot analysis

SMSCs were harvested on ice in PBS and centrifuged at 1.3×104 g for 10 min. Total protein of the SMSCs was isolated using lysis buffer (Sigma-Aldrich; Merck-Millipore) and quantified using a bicinchoninic acid (BCA) assay (Beyotime Biotechnology, Inc., China). Then, 20 µg protein was electrophoresed on 10% gel with SDS-PAGE and transferred onto a PVDF membrane (Millipore, Bedford, MA, USA). The membrane was then blocked with 5% nonfat milk for 2 h at 4°C and was then incubated with GLUT3 (cat. no. bs-20225R; 1:500 dilution; BIOSS), HIF-1α (cat. no. K000487P; 1:500 dilution; Beijing Solarbio Science & Technology Co., Ltd.), cleaved caspase3 (cat. no. 1083-10; 1:500 dilution; BioVision, Inc.), Bax (cat. no. K002397P; 1:500 dilution; Beijing Solarbio Science & Technology Co., Ltd.) and Bcl-2 (cat. no. K003505P; 1:500 dilution; Beijing Solarbio Science & Technology Co., Ltd.) antibodies and GAPDH antibody (cat. no. G5262-1VL; 1:3,000 dilution; Sigma-Aldrich; Merck KGaA) overnight at 4°C. Then, the membranes were re-incubated with secondary antibodies (Cell Signaling Technology, Inc., USA). The signal was visualized using a photographic developer (Invitrogen Life Technologies, Inc., USA) and densitometry was performed using ImageJ (version 1.25; National Institutes of Health, Bethesda, MD, USA).

Statistical analysis

Data are reported as the means ± standard errors. SPSS 19.0 software (IBM Corp., Armonk, NY, USA) was used for statistical analysis. Parameters of cells under normoxic (21% O2), hypoxic (5% O2) and severe hypoxic (0.5% O2) conditions were determined by one-way ANOVA. Parameters of cells between groups following siRNA-induced HIF-1α knockdown were analyzed using a t-test. P<0.05 was considered statistically significant.

Results

Characterization of the SMSCs

Multilateral form fibre cells and a few spindle-shaped cells were observed among the primary cells and the 2nd passage of the SMSCs (Fig. 1A and B). Oil red O-positive lipid droplets could be obviously observed in the SMSCs after adipogenic induction for 3 weeks (Fig. 1C). Similarly, Alizarin red S staining showed that some mineralized nodules were effectively formed in the isolated SMSCs after three weeks of osteoinduction (Fig. 1D). These results demonstrated that the SMSCs exhibited the potential of multidirectional differentiation. CCK-8 assays demonstrated that the SMSCs increased over time in exponential growth, and the 2nd passage of the SMSCs exhibited the highest proliferation (Fig. 1E). The immunophenotypes were determined via flow cytometry. We found that the surface markers of CD44, CD90, CD105, CD147, CD14, CD34, CD45 and CD117 were expressed on average on 96.2, 94.8, 93.6, 98.1, 6.9, 7.6, 4.4 and 3.5% of the SMSCs, respectively (Fig. 1F), which fits the criteria that we previously reported (28).

Characteristics of the SMSCs under hypoxic conditions

To observe the effects of different oxygen concentration microenvironments on the proliferation and apoptosis of SMSCs, cells were cultured under oxygen environment of normoxia (21% O2), hypoxia (5% O2) and severe hypoxia (0.5% O2). Our findings suggested that there were no significant changes in cell viability or GLUT3 mRNA and protein expression following incubation under different oxygen conditions (Fig. 2A-C). Interestingly, the mRNA and protein expression of HIF-1α was significantly upregulated under hypoxic (5% O2) and severe hypoxic (0.5% O2) conditions (Fig. 2D and E). Additionally, no obvious changes were found in the protein expression levels of cleaved caspase 3, Bax and Bcl-2 (Fig. 3A). The level of apoptosis determined by flow cytometry also revealed no significant changes (Fig. 3B). These results indicated that HIF-1α may be engaged in regulating the proliferation and apoptosis of SMSCs.

Effect of HIF-1α on SMSCs under hypoxic condition

To characterize the specific mechanism of HIF-1α on SMSCs, siRNA-induced HIF-1α knockdown was conducted under severe hypoxic (0.5% O2) and hypoxic (5% O2) conditions. Our findings suggested that the cell viability and GLUT3 mRNA and protein expression were markedly suppressed following siRNA-induced HIF-1α knockdown (Fig. 4A-C). The mRNA and protein expression of HIF-1α were significantly suppressed following si-HIF-1α knockdown under severe hypoxic (0.5% O2) and hypoxic (5% O2) conditions (Fig. 4D and E). In addition, the protein expression of cleaved caspase 3 and Bax was significantly increased and the Bcl-2 expression was significantly decreased following HIF-1α knockdown (Fig. 5A). Flow cytometric analysis revealed that the apoptosis ratio was significantly increased following siRNA-induced HIF-1α knockdown (Fig. 5B). These results suggested that HIF-1α could effectively improve the tolerance to hypoxia of SMSCs, which might promote cellular proliferation and prevent the apoptosis of SMSCs under hypoxic conditions.

Discussion

Hypoxia occurs commonly in many types of mammalian tissues, such as synovium, cartilage and intervertebral discs, where cells with a poor O2 supply can adapt to these low oxygen tension conditions by activating survival pathways (3133). Synovium is a type of connective tissue covering most of the inner joint structure, including the inner surface of the joint fibrous capsule, intra-articular ligaments, tendons, and bone surface (34). Synovial mesenchymal stem cells (SMSCs) are the principal cell type of synovial tissue, which are related to the synthesis of hyaluronate and the secretion of synovial fluid (35,36). In the present study, to explore the effects of hypoxia on SMSCs, SMSCs were cultured under normoxic and hypoxic conditions. We found that there were no significant changes in the cellular proliferation and apoptosis of SMSCs under normal and hypoxic conditions. Apoptotic proteins, such as cleaved caspase 3, Bax and Bcl-2 also exhibited no significant change under normal and hypoxic conditions. However, the levels of HIF-1α mRNA and protein were significantly increased under hypoxic condition. These results demonstrated that HIF-1α may be expressed in large quantities under hypoxic conditions, which could improve the hypoxic tolerance and inhibit the spontaneous apoptosis of SMSCs.

To verify the specific role of HIF-1α in SMSCs under hypoxic conditions and to further explore the relationship between HIF-1α and the apoptosis of SMSCs, we conducted a selective inhibition of HIF-1α under 0.5% O2 and 5% O2 hypoxic conditions by siRNA interference. Our findings revealed that the cellular proliferation was significantly inhibited and the apoptosis ratio was significantly increased after HIF-1α knockdown. Western blot assays demonstrated that the protein expression levels of cleaved caspase 3 and Bax were obviously increased, and Bcl-2 expression was significantly inhibited. These results suggest that the HIF-1α gene plays a protective role in cellular proliferation and apoptosis of SMSCs in a hypoxic environment.

The particular mechanisms may be summarized as follows. In the adaptive response of cells to changes in oxygen, the activated HIF-1α induced by hypoxia was found to stimulate more than 100 downstream genes for mediating the process of cell proliferation and survival (37,38). These metabolic processes are involved in cell proliferation, migration, glucose metabolism and angiogenesis (39,40). Prior studies have noted that HIF-1α mediates adaptive metabolic responses to hypoxia by decreasing flux via the tricarboxylic acid cycle and increasing flux via the glycolytic pathway, in order to meet the energy demands of rapidly growing tissue (23,41). Therefore, cells under hypoxic conditions tend to burn more glucose in order to achieve adequate energy requirements for cell survival. The evidence suggests that HIF-1α mediates this metabolic transformation by inducing the overexpression of glucose transporters (GLUTs) and enzymes that are involved in the glycolysis pathway, thereby increasing glucose entry into cells (42,43).

In the present study, mRNA and protein expression of GLUT3 were markedly suppressed following HIF-1α knockdown, suggesting that HIF-1α may promote the glucose metabolic conversion of SMSCs under hypoxic condition. As mentioned in a literature review, the HIF-1α transcriptional induction of a variety of angiogenic factors, such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) promotes the synthesis of endogenous angiogenesis, which in turn promotes neovascularization to enrich cells for growth (44). Moreover, HIF-1α was found to promote cell migration into oxygen-rich regions through inducing secretion via transcriptional activation growth factors such as fibroblast growth factor 11 (FGF11), transforming growth factor β3 (TGF-β3), insulin-like growth factor (IGF) and epidermal growth factor (EGF) (4547).

In conclusion, we preliminarily explored the effects of HIF-1α on the proliferation and apoptosis of human SMSCs, and the results suggest that HIF-1α activation in SMSCs is probably one of the key mechanisms mediating the ability of SMSCs to adapt to hypoxic environments.

Acknowledgements

Not applicable.

Funding

This project was supported by Scientific Research Topics of Jiangsu Provincial Health Commission (grant no. H2018027).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

DH and XS conceived and designed the study. DH, XS, WZ and XG performed the experiments and analyzed the data. WZ and XG wrote the paper. DH and WZ reviewed and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Ethical approval for this study was granted by the Institution Review Board of the Affiliated Hospital of Nanjing Medical University, and all study participants were recruited after providing informed written consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Perez JR, Kouroupis D, Li DJ, Best TM, Kaplan L and Correa D: Tissue engineering and cell-based therapies for fractures and bone defects. Front Bioeng Biotechnol. 6:1052018. View Article : Google Scholar : PubMed/NCBI

2 

Golpanian S, Wolf A, Hatzistergos KE and Hare JM: Rebuilding the damaged heart: Mesenchymal stem cells, cell-based therapy, and engineered heart tissue. Physiol Rev. 96:1127–1168. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Heathman TR, Nienow AW, McCall MJ, Coopman K, Kara B and Hewitt CJ: The translation of cell-based therapies: Clinical landscape and manufacturing challenges. Regen Med. 10:49–64. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Leach JK and Whitehead J: Materials-directed differentiation of mesenchymal stem cells for tissue engineering and regeneration. ACS Biomater Sci Eng. 4:1115–1127. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Toyoda E, Sato M, Takahashi T, Maehara M, Nakamura Y, Mitani G, Takagaki T, Hamahashi K and Watanabe M: Multilineage-differentiating stress-enduring (Muse)-like cells exist in synovial tissue. Regen Ther. 10:17–26. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Hatakeyama A, Uchida S, Utsunomiya H, Tsukamoto M, Nakashima H, Nakamura E, Pascual-Garrido C, Sekiya I and Sakai A: Isolation and characterization of synovial mesenchymal stem cell derived from hip joints: A comparative analysis with a matched control knee group. Stem Cells Int. 2017:93123292017. View Article : Google Scholar : PubMed/NCBI

7 

Zheng W, Gu X, Sun X, Wu Q and Dan H: FAK mediates BMP9-induced osteogenic differentiation via Wnt and MAPK signaling pathway in synovial mesenchymal stem cells. Artifl Cells Nanomed Biotechnol. 47:2641–2649. 2019. View Article : Google Scholar

8 

Baboolal TG, Khalil-Khan A, Theodorides AA, Wall O, Jones E and McGonagle D: A novel arthroscopic technique for intraoperative mobilization of synovial mesenchymal stem cells. Am J Sports Med. 46:3532–3540. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Santhagunam A, Dos Santos F, Madeira C, Salgueiro JB and Cabral JM: Isolation and ex vivo expansion of synovial mesenchymal stromal cells for cartilage repair. Cytotherapy. 16:440–453. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Ferro T, Santhagunam A, Madeira C, Salgueiro JB, da Silva CL and Cabral JMS: Successful isolation and ex vivo expansion of human mesenchymal stem/stromal cells obtained from different synovial tissue-derived (biopsy) samples. J Cell Physiol. 234:3973–3984. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Segawa Y, Muneta T, Makino H, Nimura A, Mochizuki T, Ju YJ, Ezura Y, Umezawa A and Sekiya I: Mesenchymal stem cells derived from synovium, meniscus, anterior cruciate ligament, and articular chondrocytes share similar gene expression profiles. J Orthop Res. 27:435–441. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Juranek I, Stern R and Soltes L: Hyaluronan peroxidation is required for normal synovial function: An hypothesis. Med Hypotheses. 82:662–666. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Fearon U, Canavan M, Biniecka M and Veale DJ: Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol. 12:385–397. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Schipani E, Mangiavini L and Merceron C: HIF-1α and growth plate development: What we really know. Bonekey Rep. 4:7302015. View Article : Google Scholar : PubMed/NCBI

15 

Schönenberger MJ and Kovacs WJ: Hypoxia signaling pathways: Modulators of oxygen-related organelles. Front Cell Dev Biol. 3:422015. View Article : Google Scholar : PubMed/NCBI

16 

Esteve JM, Launay JM, Kellermann O and Maroteaux L: Functions of serotonin in hypoxic pulmonary vascular remodeling. Cell Biochem Biophys. 47:33–44. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Patel J, Landers K, Mortimer RH and Richard K: Regulation of hypoxia inducible factors (HIF) in hypoxia and normoxia during placental development. Placenta. 31:951–957. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Acker T and Plate KH: Hypoxia and hypoxia inducible factors (HIF) as important regulators of tumor physiology. Cancer Treat Res. 117:219–248. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Warfel NA, Dolloff NG, Dicker DT, Malysz J and El-Deiry WS: CDK1 stabilizes HIF-1α via direct phosphorylation of Ser668 to promote tumor growth. Cell Cycle. 12:3689–3701. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Alonso D, Serrano E, Bermejo FJ and Corral RS: HIF-1α-regulated MIF activation and Nox2-dependent ROS generation promote Leishmania amazonensis killing by macrophages under hypoxia. Cell Immunol. 335:15–21. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Liu J, Zhang C, Zhao Y, Yue X, Wu H, Huang S, Chen J, Tomsky K, Xie H, Khella CA, et al: Parkin targets HIF-1α for ubiquitination and degradation to inhibit breast tumor progression. Nat Commun. 8:18232017. View Article : Google Scholar : PubMed/NCBI

22 

Mukund V, Saddala MS, Farran B, Mannavarapu M, Alam A and Nagaraju GP: Molecular docking studies of angiogenesis target protein HIF-1α and genistein in breast cancer. Gene. 701:169–172. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Palmer CS, Ostrowski M, Balderson B, Christian N and Crowe SM: Glucose metabolism regulates T cell activation, differentiation, and functions. Front Immunol. 6:12015. View Article : Google Scholar : PubMed/NCBI

24 

Cho SH, Raybuck AL, Blagih J, Kemboi E, Haase VH, Jones RG and Boothby MR: Hypoxia-inducible factors in CD4+ T cells promote metabolism, switch cytokine secretion, and T cell help in humoral immunity. Proc Natl Acad Sci USA. 116:8975–8984. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Pfander D and Gelse K: Hypoxia and osteoarthritis: How chondrocytes survive hypoxic environments. Curr Opin Rheumatol. 19:457–462. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Jiang SJ, Li W, Li YJ, Fang W and Long X: Dickkopfrelated protein 1 induces angiogenesis by upregulating vascular endothelial growth factor in the synovial fibroblasts of patients with temporomandibular joint disorders. Mol Med Rep. 12:4959–4966. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Fleming AM, Zhu J, Ding Y and Burrows CJ: Location dependence of the transcriptional response of a potential G-quadruplex in gene promoters under oxidative stress. Nucleic Acids Res. 47:5049–5060. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Zheng W, Yang M, Wu C and Su X: Experimental study on Osteogenesis of synovium-derived mesenchymal stem cells in vitro and in vivo. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 30:102–109. 2016.(In Chinese). PubMed/NCBI

29 

Lv B, Hua T, Li F, Han J, Fang J, Xu L, Sun C, Zhang Z, Feng Z and Jiang X: Hypoxia-inducible factor 1 a protects mesenchymal stem cells against oxygen-glucose deprivation-induced injury via autophagy induction and PI3K/AKT/mTOR signaling pathway. Am J Transl Res. 9:2492–2499. 2017.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 

Fernández-Torres J, Martínez-Nava GA, Gutiérrez-Ruíz MC, Gómez-Quiroz LE and Gutiérrez M: Role of HIF-1α signaling pathway in osteoarthritis: A systematic review. Rev Bras Reumatol Engl Ed. 57:162–173. 2017.(In English, Portuguese). PubMed/NCBI

32 

Daniel SK, Sullivan KM, Labadie KP and Pillarisetty VG: Hypoxia as a barrier to immunotherapy in pancreatic adenocarcinoma. Clin Transl Med. 8:102019. View Article : Google Scholar : PubMed/NCBI

33 

Ichihara S, Li P, Mise N, Suzuki Y, Izuoka K, Nakajima T, Gonzalez F and Ichihara G: Ablation of aryl hydrocarbon receptor promotes angiotensin II-induced cardiac fibrosis through enhanced c-Jun/HIF-1α signaling. Arch Toxicol. 93:1543–1553. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Radenska-Lopovok SG: Immunomorphological characteristics of the synovial membrane in rheumatic diseases. Arkh Patol. 78:64–68. 2016.(In Russian). View Article : Google Scholar : PubMed/NCBI

35 

Sekiya I, Koga H, Otabe K, Nakagawa Y, Katano H, Ozeki N, Mizuno M, Horie M, Kohno Y, Katagiri K, et al: Additional use of synovial mesenchymal stem cell transplantation following surgical repair of a complex degenerative tear of the medial meniscus of the knee: A case report. Cell Transplant. Jul 17–2019.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

36 

Desando G, Bartolotti I, Cavallo C, Schiavinato A, Secchieri C, Kon E, Filardo G, Paro M and Grigolo B: Short-term homing of hyaluronan-primed cells: Therapeutic implications for osteoarthritis treatment. Tissue Eng Part C Methods. 24:121–133. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Xu W, Xu R, Li X, Zhang H, Wang X and Zhu J: Downregulating hypoxia-inducible factor-1α expression with perfluorooctyl-bromide nanoparticles reduces early brain injury following experimental subarachnoid hemorrhage in rats. Am J Transl Res. 8:2114–2126. 2016.PubMed/NCBI

38 

Masoud GN and Li W: HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta Pharm Sin B. 5:378–389. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Miska J, Lee-Chang C, Rashidi A, Muroski ME, Chang AL, Lopez-Rosas A, Zhang P, Panek WK, Cordero A, Han Y, et al: HIF-1α is a metabolic switch between glycolytic-driven migration and oxidative phosphorylation-driven immunosuppression of tregs in glioblastoma. Cell Rep. 27:226–237.e4. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Bensaad K and Harris AL: Hypoxia and metabolism in cancer. Adv Exp Med Biol. 772:1–39. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Zhao L, Mao Y, Zhao Y, Cao Y and Chen X: Role of multifaceted regulators in cancer glucose metabolism and their clinical significance. Oncotarget. 7:31572–31585. 2016.PubMed/NCBI

42 

Pereira KM, Chaves FN, Viana TS, Carvalho FS, Costa FW, Alves AP and Sousa FB: Oxygen metabolism in oral cancer: HIF and GLUTs (Review). Oncol Lett. 6:311–316. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Chen Z, Tian D, Liao X, Zhang Y, Xiao J, Chen W, Liu Q, Chen Y, Li D, Zhu L and Cai S: Apigenin combined with gefitinib blocks autophagy flux and induces apoptotic cell death through inhibition of HIF-1α, c-Myc, p-EGFR, and glucose metabolism in EGFR L858R+T790M-mutated H1975 cells. Front Pharmacol. 10:2602019. View Article : Google Scholar : PubMed/NCBI

44 

Fu YC and Xin ZM: Inhibited corneal neovascularization in rabbits following corneal alkali burn by double-target interference for VEGF and HIF-1α. Biosci Rep. 39(pii): BSR201805522019. View Article : Google Scholar : PubMed/NCBI

45 

Szojka ARA, Lyons BD, Moore CN, Liang Y, Kunze M, Idrees E, Mulet-Sierra A, Jomha NM and Adesida AB: Hypoxia and TGF-β3 synergistically mediate inner meniscus-like matrix formation by fibrochondrocytes. Tissue Eng Part A. 25:446–456. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Knowles HJ: Hypoxia-induced fibroblast growth factor 11 stimulates osteoclast-mediated resorption of bone. Calcif Tissue Int. 100:382–391. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Zhao FL and Qin CF: EGF promotes HIF-1α expression in colorectal cancer cells and tumor metastasis by regulating phosphorylation of STAT3. Eur Rev Med Pharmacol Sci. 23:1055–1062. 2019.PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 20 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
Zheng W, Gu X, Sun X and Hu D: Effects of hypoxia‑inducible factor‑1α on the proliferation and apoptosis of human synovial mesenchymal stem cells. Mol Med Rep 20: 4315-4322, 2019
APA
Zheng, W., Gu, X., Sun, X., & Hu, D. (2019). Effects of hypoxia‑inducible factor‑1α on the proliferation and apoptosis of human synovial mesenchymal stem cells. Molecular Medicine Reports, 20, 4315-4322. https://doi.org/10.3892/mmr.2019.10656
MLA
Zheng, W., Gu, X., Sun, X., Hu, D."Effects of hypoxia‑inducible factor‑1α on the proliferation and apoptosis of human synovial mesenchymal stem cells". Molecular Medicine Reports 20.5 (2019): 4315-4322.
Chicago
Zheng, W., Gu, X., Sun, X., Hu, D."Effects of hypoxia‑inducible factor‑1α on the proliferation and apoptosis of human synovial mesenchymal stem cells". Molecular Medicine Reports 20, no. 5 (2019): 4315-4322. https://doi.org/10.3892/mmr.2019.10656