ATF3 promotes migration and M1/M2 polarization of macrophages by activating tenascin‑C via Wnt/β‑catenin pathway

  • Authors:
    • Hao Sha
    • Dianzhong Zhang
    • Yunfei Zhang
    • Yanhua Wen
    • Yucai Wang
  • View Affiliations

  • Published online on: July 14, 2017     https://doi.org/10.3892/mmr.2017.6992
  • Pages: 3641-3647
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

There are different polarization states of macrophages, including the classically activated M1 phenotype and the alternatively activated M2 phenotype. These have different functions in the inflammation process. Activating transcription factor 3 (ATF3) is a key transcriptional regulator that inhibits the inflammatory response. However, the effects of ATF3 on migration and anti‑inflammatory control mechanisms of macrophages have not been thoroughly investigated. The present study investigated the effect of ATF3 on macrophage migration and M1/M2 polarization. Results revealed that overexpression of ATF3 promoted macrophage migration and the expression of the M2 phenotype markers [cluster of differentiation (CD) 163, mannose receptor C type 1, arginase 1 and peroxisome proliferator‑activated receptor γ] and inhibited expression of the M1 phenotype markers (monocyte chemoattractant protein‑1, inducible nitric oxide synthase, CD16 and tumor necrosis factor‑α), whereas knockdown of ATF3 resulted in a contrary effect. In addition, the wingless‑type MMTV integration site family member (Wnt)/β‑catenin signaling pathway was activated and the expression level of tenascin (TNC) was significantly upregulated by overexpression of ATF3. Additionally, inhibition of Wnt/β‑catenin signaling significantly attenuated the upregulatory effect of ATF3 on TNC. Finally, the effect of ATF3 on macrophage migration and markers of the M1 or M2 state was investigated using TNC‑specific siRNA. In conclusion, the results of the present study suggested that ATF3 promotes macrophage migration and reverses M1‑polarized macrophages to the M2 phenotype by upregulation of TNC via the Wnt/β‑catenin signaling pathway.

Introduction

Macrophages are essential components of innate immunity that serve a role in inflammation and host defense via production of pro- or anti-inflammatory mediators in response to various stimuli (1). Macrophages respond to stimulation in a polarized manner. The differentiation of macrophages to the classic activation (M1) phenotype is triggered by interferon (IFN)-γ, bacterial lipopolysaccharide (LPS), interleukin (IL)-1β, or tumor necrosis factor α (TNF-α), whereas IL-3 or IL-13 stimulate macrophage differentiation of the alternative activation phenotype (M2) (25). The M1 phenotype is characterized by the expression of high levels of pro-inflammatory cytokines. Conversely, M2 macrophages express an anti-inflammatory functional profile and are associated with wound repair and angiogenesis (6). Therefore, inflammatory associated diseases may result from a sustained pro-inflammatory reaction and failure of anti-inflammatory control mechanisms of macrophages.

Activating transcription factor 3 (ATF3), a member of the mammalian activating transcription factor/cAMP responsive element binding protein (ATF/CREB) family of transcription factors, is induced in a variety of stressed tissues, including mechanically and toxin-injured liver tissue, blood-deprived heart tissue and injured peripheral nerves (7,8). The transcriptional target of ATF3 varies in different cell types and conditions, which therefore leads to diverse effects on cell survival, proliferation and death (9). In neurons, ATF3 is frequently reported to be a novel neuronal marker of nerve injury, and induction of ATF3 expression enhances nerve regeneration (10,11). In cardiac myocytes, ATF3 is a novel cytoprotective factor in doxorubicin-induced apoptosis (12). Additionally, it is reported that ATF3 protects renal cells and b-cells against oxidative stress-induced cell death and apoptosis (13). Therefore, ATF3 is a protective factor in numerous tissues.

Conversely, ATF3 is an inducible transcriptional repressor in innate immune cells, which regulates the magnitude and duration of inducible pro-inflammatory gene expression. Recently, it was revealed that ATF3 is an important transcriptional regulator that inhibits the inflammatory response by modulating the expression of cytokines and chemokines and demonstrated that ATF3 is a negative regulator of Toll-like receptor 4 (TLR4) signaling in macrophages (14). Activation of TLR4 by LPS induces the expression of ATF3, which subsequently inhibits the expression of various inflammatory genes induced by TLR signaling, including IL-6, IL-12β, and TNF-α. Additionally, ATF3 may modulate the expression levels of IFN-γ in macrophages by controlling basal and inducible levels of IFN-β, and the expression of genes downstream of IFN (15). Therefore, ATF3 is able to negatively regulate transcription of pro-inflammatory cytokines in macrophages. Understanding the exact role of ATF3 in macrophages in the context of inflammation is of primary concern, and may lead to the design of beneficial therapeutics for inflammatory associated diseases. However, the effects of ATF3 on recruitment and anti-inflammatory control mechanisms of macrophages remain to be investigated.

The present study investigated whether ATF3 exerted anti-inflammatory activities by modulating M1/M2 differentiation of macrophages. Macrophage migration and markers of M1/M2 macrophages were tested following overexpression of ATF3. Subsequently, the underlying mechanism of how overexpression of ATF3 modulates macrophage migration and M1/M2 polarization was investigated.

Materials and methods

Cell culture

Mouse macrophage RAW 264.7 cells (American Type Culture Collection, Manassas, VA, USA) were cultured in Dulbecco's modified Eagle's medium (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum, 100 U/ml streptomycin and 100 U/ml penicillin (Gibco; Thermo Fisher Scientific, Inc.) in a humidified atmosphere at 37°C and 5% CO2. Wntpalmitoyltransferase inhibitor (IWP-2,N-(6-M ethyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]-acetamide) was purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany) and 30 µM was used to treat the cells.

Plasmids and siRNA transfection

For overexpression of ATF3, ATF3 cDNA generated from reverse transcription-quantitative PCR (RT-qPCR; The primers used for the cloning were: 5′-AAAAAGCTTATGATGCTTCAACATCCAGG-3′ and 5′-TTTGAATTCTTAGCTCTGCAATGTTCCTT-3′) was subcloned into a pcDNA™3.1 plasmid (Invitrogen; Thermo Fisher Scientific, Inc.) between Hind3 and EcoR I sites to express ATF3 in abundance in E. coli DH5α cells to generate an ATF3 expression plasmid. The third generation of cells (5×105) were seeded into a 24-well plate and transfected with the pcDNA-ATF3 or the empty vector negative control plasmid, psDNA3.1(−), using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol at 37°C. After incubation for 48 h, cells were harvested and ATF3 expression was determined. To knockdown ATF3 and tenascin (TNC), The third generation of cells (5×105) were seeded into a 24-well plate and ATF3 siRNA, its negative control (sham), or TNC siRNA and its negative control siRNA (scramble) were transfected at 37°C using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.; 0.1 µM siRNA with 20 µl Lipofectamine). Following transfection for 48 h, the transfection medium was exchanged for normal medium and the cells were used in the subsequent experiments or harvested for ATF3 and TNC expression measurement.

Migration assays

Migration assays were performed using 8 µm pore size filters within 24-well transwell cell culture chambers with polycarbonate filters (Corning Life Sciences, Glendale, Arizona, USA) as previously described (16). A total of 5×105 cells were transfected with pcDNA-ATF3 or the psDNA3.1(−), and subsequently seeded into the upper chamber of the transwell. Transwells were either uncoated (5 µm pore size) or coated with Matrigel™ (BD Biosciences, Franklin Lakes, NJ, USA) diluted 1:50 (8 µm pore size). As a chemoattractant, monocyte chemoattractant protein-1 (MCP-1; 100 ng/ml; R&D Systems, Inc., Minneapolis, MN, USA) was present in the lower wells. After incubation for 18 h at 37°C in 5% CO2, cells in the lower chambers that passed through the filter were counted under a Carl Zeiss Primo Vert microscope (Carl Zeiss AG, Oberkochen, Germany).

Western blotting

Total protein was lysed using radioimmunoprecipitation assay (RIPA) buffer (Beyotime Institute of Biotechnology, Nantong, China). After determining the protein concentration with a Bicinchoninic acid assay kit (Beyotime Institute of Biotechnology, Haimen, China), ~30 µg of proteins were loaded onto 10% gels and subjected to SDS-PAGE, prior to transfer onto polyvinylidene difluoride membranes (Invitrogen; Thermo Fisher Scientific, Inc.). Subsequently, membranes were blocked for 1 h in a blocking solution (5% skimmed milk, 0.05% Tween 20) at 37°C and probed with the following primary antibodies: Mouse anti-β-catenin (2698; 1:1,000), rabbit anti-c-myc 9402; 1:1,000) and anti-cyclin D1 (2292; 1:1,000; all from Cell Signaling Technology, Inc., Danvers, MA, USA), and mouse anti-ATF3 (sc81189; 1:500), rabbit anti-TNC (sc20932; 1:1,000), and mouse anti-β-actin (sc130300; 1:1,000; all from Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at 4°C overnight. Horseradish peroxidase-conjugated rabbit anti-mouse (sc358917; 1:3,000; Santa Cruz Biotechnology) or goat anti-rabbit (RPN4301; 1:5,000; GE Healthcare Life Sciences, Chalfont, UK) secondary antibodies were added to the membranes for 1 h at room temperature. Protein bands were detected using the Enhanced Chemiluminescence substrate detection system (Amersham Biosciences Corporation, Piscataway, USA). The intensities of the resulting bands were quantified using Carestream Molecular Imaging software version 5.0.2.30 (Carestream Health, Woodbridge, CT, USA) on a Gel Logic 2000 imaging system (Kodak, Rochester, NY, USA).

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

Reverse transcription PCR was performed on an Applied Biosystems® 7500 fast sequence detection system (Applied Biosystems; Thermo Fisher Scientific, Inc.). Briefly, total RNA was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and reverse transcribed using the MMLV Reverse Transcriptase kit (Takara Biotechnology Co., Ltd., Dalian, China) according to the manufacturer's protocol. qPCR was performed using SYBR Green reagent (Qiagen, Inc., Valencia, CA, USA). Cycling conditions were as follows: An initial predenaturation step at 95°C for 5 min, followed by 40 cycles of denaturation at 95°C for 15 sec, annealing at 58°C for 30 sec and extension at 72°C for 20 sec. The experiment was performed three times. The relative expression levels of the target genes were calculated using the 2−∆∆Cq method (17) and normalized to GAPDH. Forward and reverse sequences of the primers used for all target genes and GAPDH are listed in Table I.

Table I.

Primers used.

Table I.

Primers used.

GenePrimer sequences
MCP-1F: 5′-TCAGCCAGATGCAGTTAACGC-3′
R: 5′-TGGATGCATTAGCTTCAGATTTACG-3′
CD16F: 5′-GACAGTGTGACTCTGAAG-3′
R: 5′-GCACCTGTACTCTCCAC-3′
iNOSF: 5′-CCCTTCCGAATTTCTGGCAGCAGC-3′
R: 5′-GGCTGTCAGAGCCTCGTGGCTTTGG-3′
TNF-19αF: 5′-TTGACCTCAGCGCTGAGTTG-3′
R: 5′-CCTGTAGCCCACGTCGTAGC-3′
Arg-1F: 5′-CAGAAGAATGGAAGAGTCAG-3′
R: 5′-CAGATATGCAGGGAGTCACC-3′
CD163F: 5′-ATGGGTGGACACAGAATGGTT-3′
R: 5′-CAGGAGCGTTAGTGACAGCAG-3′
Mrc-1F: 5′-TCTTTTACGAGAAGTTGGGGTCAG-3′
F: 5′-ATCATTCCGTTCACAGAGGG-3′
PPARγR: 5′-GGAGATCTCCAGTGATATCGACCA-3′
F: 5′-ACGGCTTCTACGGATCGAAACT-3′
TNCR: 5′-GTTTGGAGACCGCAGAGAAGAA-3′
F: 5′-TGTCCCCATATCTGCCCATCA-3′
GAPDHR: 5′-AGGTCGGTGTGAACGGATTTC-3′
F: 5′-TGTAGACCATGTAGTTGAGGTCA-3′

[i] MCP-1, monocyte chemoattractant protein 1; CD, cluster of differentiation iNOs, inducible nitric oxide synthase; TNF-α, tumor necrosis factor-α; Arg-1, arginase 1; Mrc-1, mannose receptor C type 1; PPARγ, peroxisome proliferator-activated receptor γ; TNC, tenascin.

Statistical analysis

Data are expressed as the mean ± standard deviation. Comparisons between two groups were analyzed by unpaired Student's t-test. Experiments were repeated three times. P<0.05 was considered to indicate a statistically significant difference analyzed using SPSS version 18.0 (SPSS, Inc., Chicago, IL, USA).

Results

Overexpression of ATF3 promotes the migration of macrophages

To examine whether ATF3 regulates macrophage migration, the pcDNA-ATF3 plasmid and ATF3 siRNA were utilized to overexpress and knockdown the ATF3 protein, respectively (Fig. 1A). Cell viability was tested using an MTT assay and the ATF3 siRNA had no effect on cellular viability (data not show).

Subsequently, migration of macrophages was evaluated in the presence of chemotaxis-inducing agent MCP-1. Results demonstrated that overexpression of ATF3 significantly promoted the migration of macrophages compared with the empty vector [pcDNA3.1(−)], whereas knockdown of ATF3 significantly reduced migration compared with the sham group and the difference between the control and sham group were not significant (Fig. 1B).

Overexpression of ATF3 promotes macrophage differentiation of the M2 phenotype

To determine the function of ATF3 in the process of macrophage polarization, markers of the M1 phenotype [MCP-1, inducible nitric oxide synthase (iNOS), cluster of differentiation (CD) 16 and TNF-α] and the M2 phenotype [CD163, mannose receptor C type 1 (Mrc-1), arginase 1 (Arg-1) and peroxisome proliferator-activated receptor γ (PPARγ)] were measured. Results revealed that the mRNA expression levels of M1-associated genes encoding MCP-1, iNOS, CD16 and TNF-α, were reduced following transfection with pcDNA-ATF3 compared with the empty vector control group, and were enhanced following transfection with ATF3 siRNA compared with the sham group (P<0.05; Fig. 2A). In addition, overexpression of ATF3 in RAW 264.7 cells enhanced the expression levels of the genes encoding CD163, Mrc-1, Arg-1 and PPARγ compared with the empty vector control group, and these levels were then reduced by knockdown of ATF3 compared with the sham group (P<0.05; Fig. 2B). These results suggested that ATF3 promotes polarization of M2 in macrophages.

Overexpression of ATF3 activates the Wnt/β-catenin signaling pathway

β-catenin, encoded by the CTNNB1 gene, is a transcriptional co-activator and serves a role in the inflammatory response (18,19). To explore the mechanism of ATF3 regulation of macrophage migration and M2 polarization, the effect of ATF3 on Wnt/β-catenin signaling was investigated. As demonstrated in Fig. 3, overexpression of ATF3 resulted in enhanced expression levels of β-catenin and its target genes cyclin D1 and c-myc compared with cells transfected with the empty vector control. This suggested that overexpression of ATF3 induced the activation of the Wnt/β-catenin signaling pathway in macrophages.

TNC is activated by ATF3 via the Wnt/β-catenin signaling pathway

The gene encoding TNC is a canonical Wnt target (20), and serves a role in macrophage behavior and function (21,22). Therefore, the association between ATF3 and TNC was investigated. Results revealed that overexpression of AFT3 significantly upregulated the mRNA expression levels of TNC (Fig. 4A) and the TNC protein (Fig. 4B) compared with the empty vector control, and this effect was partially inhibited by IWP-2, an inhibitor of Wnt/β-catenin signaling, which suggested that ATF3 activates TNC via the Wnt/β-catenin signaling pathway.

ATF3 regulates the migration and polarization of M2 macrophages by upregulating TNC expression levels

To further investigate the role of TNC in ATF3-mediated macrophage migration and polarization, cells were transfected with pcDNA-ATF3 and knockdown of TNC using TNC siRNA (Fig. 5A), prior to determining cell viability and the expression levels of M2-associated genes. Results revealed that transfection of pcDNA-ATF3 significantly promoted macrophage migration compared with the empty vector control, whereas transfection with TNC siRNA reduced the migration induced by pcDNA-ATF3 compared with the scrambled control group (Fig. 5B). In addition, the expression levels of genes associated with the M1 phenotype that were downregulated by pcDNA-ATF3 were enhanced by transfection with TNC siRNA (Fig. 5C), whereas M2 gene expression levels that were upregulated by pcDNA-ATF3 were inhibited by TNC siRNA (Fig. 5D).

These results suggested that ATF3 regulates macrophage migration and M2 polarization, in part, by upregulation of TNC.

Discussion

Macrophages are primary producers of pro-inflammatory mediators and the migration of macrophages from the circulation into injured tissues serves a crucial role in wound healing. Macrophage polarization is closely associated with homeostatic tissue remodeling, resolution of inflammation, remodeling and tissue repair (23). ATF3 is a transcriptional modulator, induced by LPS and the TLR-dependent injury response, that negatively regulates numerous pro-inflammatory cytokines and chemokines in macrophages (24). Previous reviews have reported that ATF3 modulates the expression levels of a number of inflammatory genes (25). Therefore, the present study investigated the effect of ATF3 on macrophage migration and polarization.

Migration of macrophages serves a role in the onset and course of inflammation. Chen et al (26) revealed that the epithelium-derived exosomal ATF3 inhibited the expression of monocyte chemoattractant protein 1 and macrophage migration, and Zmuda et al (27) suggested that ATF3 knockout islets inhibited macrophage recruitment in vivo. The present study revealed that overexpression of ATF3 promoted M2 marker expression and suppressed the expression levels of M1-associated markers. This suggested that ATF3 may reverse M1-polarized macrophages to M2 phenotypes, and that the ATF3-mediated anti-inflammatory function is closely associated with macrophage phenotype. It is evident that ATF3 serves an important role in injury in numerous tissues, and it was revealed that ATF3 may protect against acute kidney and lung injury (28,29). M2 macrophages exhibit immunoregulatory functions including defense against infection, promotion of angiogenesis and wound healing (30). The results of the present study suggested that ATF3 may be a protective regulator for injured tissues by promoting the polarization of M2 macrophages.

ATF3 serves a role in the cellular adaptive-response network in response to signals perturbing homeostasis (25). Previous data has suggested that ATF3 activates the Wnt/β-catenin signaling pathway in human breast cancer cells (31), which concurs with the results of the present study, as overexpression of ATF3 activated the Wnt/β-catenin pathway in macrophages. Various components of the Wnt/β-catenin signaling pathway are involved in the inflammatory response, including in inflammatory conditions in humans and in the LPS-treated macrophage cell line (32). Wnt upregulates the expression levels of TNC (20), which is a large hexameric extracellular matrix glycoprotein that is highly expressed during embryonic development, cancer invasion and wound healing. It has been reported that TNC may be expressed in macrophages and regulates their behavior and function (21,22). TNC has been suggested to act as pro-inflammatory modulator in various diseases (33,34) and accelerates macrophage migration (35). The results of the present study demonstrated that overexpression of ATF3 upregulated TNC expression levels via the Wnt/β-catenin signaling pathway. In addition, the present study demonstrated that TNC was an effector for ATF3 in modulating macrophage migration and M2 polarization.

In conclusion, the present study revealed that overexpression of ATF3 in macrophage cells promoted their migration to chemotaxis-inducing agent MCP-1, and influenced the M1/M2 phenotype. These results emphasize that ATF3 expression levels affect macrophages, partially by upregulating TNC via the Wnt/β-catenin signaling pathway. The present study may provide an insight into the positive regulation of ATF3 on macrophage migration, and tissue regeneration via modulation of the M2 macrophage.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81001196) and Shaanxi Social Development for Science and Technology Project (grant no. 2016SF-330).

Glossary

Abbreviations

Abbreviations:

ATF3

activating transcription factor 3

TNC

tenascin

IFN-γ

interferon-γ

TNF-α

tumor necrosis factor-α

ATF/CREB

activating transcription factor/cAMP responsive element binding protein

TLR4

Toll-like receptor 4

References

1 

Gordon S and Martinez FO: Alternative activation of macrophages: Mechanism and functions. Immunity. 32:593–604. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Martinez FO, Sica A, Mantovani A and Locati M: Macrophage activation and polarization. Front Biosci. 13:453–461. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Kuroda E, Ho V, Ruschmann J, Antignano F, Hamilton M, Rauh MJ, Antov A, Flavell RA, Sly LM and Krystal G: SHIP represses the generation of IL-3-induced M2 macrophages by inhibiting IL-4 production from basophils. J Immunol. 183:3652–3660. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Odegaard JI and Chawla A: Mechanisms of macrophage activation in obesity-induced insulin resistance. Nat Clin Pract Endocrinol Metab. 4:619–626. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Porcheray F, Viaud S, Rimaniol AC, Léone C, Samah B, Dereuddre-Bosquet N, Dormont D and Gras G: Macrophage activation switching: An asset for the resolution of inflammation. Clin Exp Immunol. 142:481–489. 2005.PubMed/NCBI

6 

Gordon S: Alternative activation of macrophages. Nat Rev Immunol. 3:23–35. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Hai T and Hartman MG: The molecular biology and nomenclature of the activating transcription factor/cAMP responsive element binding family of transcription factors: Activating transcription factor proteins and homeostasis. Gene. 273:1–11. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Hai T, Wolfgang CD, Marsee DK, Allen AE and Sivaprasad U: ATF3 and stress responses. Gene Expr. 7:321–335. 1999.PubMed/NCBI

9 

Thompson MR, Xu D and Williams BR: ATF3 transcription factor and its emerging roles in immunity and cancer. J Mol Med (Berl). 87:1053–1060. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Tsujino H, Kondo E, Fukuoka T, Dai Y, Tokunaga A, Miki K, Yonenobu K, Ochi T and Noguchi K: Activating transcription factor 3 (ATF3) induction by axotomy in sensory and motoneurons: A novel neuronal marker of nerve injury. Mol Cell Neurosci. 15:170–182. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Averill S, Michael GJ, Shortland PJ, Leavesley RC, King VR, Bradbury EJ, McMahon SB and Priestley JV: NGF and GDNF ameliorate the increase in ATF3 expression which occurs in dorsal root ganglion cells in response to peripheral nerve injury. Eur J Neurosci. 19:1437–1445. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Nobori K, Ito H, Tamamori-Adachi M, Adachi S, Ono Y, Kawauchi J, Kitajima S, Marumo F and Isobe M: ATF3 inhibits doxorubicin-induced apoptosis in cardiac myocytes: A novel cardioprotective role of ATF3. J Mol Cell Cardiol. 34:1387–1397. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Yoshida T, Sugiura H, Mitobe M, Tsuchiya K, Shirota S, Nishimura S, Shiohira S, Ito H, Nobori K, Gullans SR, et al: ATF3 protects against renal ischemia-reperfusion injury. J Am Soc Nephrol. 19:217–224. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Nguyen CT, Kim EH, Luong TT, Pyo S and Rhee DK: TLR4 mediates pneumolysin-induced ATF3 expression through the JNK/p38 pathway in Streptococcus pneumoniae-infected RAW 264.7 cells. Mol Cells. 38:58–64. 2015.PubMed/NCBI

15 

Labzin LI, Schmidt SV, Masters SL, Beyer M, Krebs W, Klee K, Stahl R, Lütjohann D, Schultze JL, Latz E and De Nardo D: ATF3 is a key regulator of macrophage IFN responses. J Immunol. 195:4446–4455. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Lighvani S, Baik N, Diggs JE, Khaldoyanidi S, Parmer RJ and Miles LA: Regulation of macrophage migration by a novel plasminogen receptor Plg-R KT. Blood. 118:5622–5630. 2011. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Kim JS, Yeo S, Shin DG, Bae YS, Lee JJ, Chin BR, Lee CH and Baek SH: Glycogen synthase kinase 3beta and beta-catenin pathway is involved in toll-like receptor 4-mediated NADPH oxidase 1 expression in macrophages. FEBS J. 277:2830–2837. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Kraus C, Liehr T, Hülsken J, Behrens J, Birchmeier W, Grzeschik KH and Ballhausen WG: Localization of the human beta-catenin gene (CTNNB1) to 3p21: A region implicated in tumor development. Genomics. 23:272–274. 1994. View Article : Google Scholar : PubMed/NCBI

20 

Pedersen EA, Scannell CA, Menon R and Lawlor ER: Tenascin C is a canonical Wnt target gene in Ewing sarcoma and its expression is potentiated by R-spondin. Cancer Res. 74:3085. 2014. View Article : Google Scholar

21 

Kimura T, Tajiri K, Hlroe M, et al: Tenascin-C regulates macrophage behavior during tissue repair after myocardial infarction in mouse model. Molecular Biology Of The CellAmer Soc Cell Biology. Bethesda, MD: pp. 20814–2755. 2014

22 

Shimojo N, Hashizume R, Kanayama K, Hara M, Suzuki Y, Nishioka T, Hiroe M, Yoshida T and Imanaka-Yoshida K: Tenascin-C may accelerate cardiac fibrosis by activating macrophages via the integrin αVβ3/nuclear factor-κB/interleukin-6 axis. Hypertension. 66:757–766. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Mantovani A, Biswas SK, Galdiero MR, Sica A and Locati M: Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol. 229:176–185. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Gilchrist M, Thorsson V, Li B, Rust AG, Korb M, Roach JC, Kennedy K, Hai T, Bolouri H and Aderem A: Systems biology approaches identify ATF3 as a negative regulator of Toll-like receptor 4. Nature. 441:173–178. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Hai T, Wolford CC and Chang YS: ATF3, a hub of the cellular adaptive-response network, in the pathogenesis of diseases: Is modulation of inflammation a unifying component? Gene Expr. 15:1–11. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Chen HH, Lai PF, Lan YF, Cheng CF, Zhong WB, Lin YF, Chen TW and Lin H: Exosomal ATF3 RNA attenuates pro-inflammatory gene MCP-1 transcription in renal ischemia-reperfusion. J Cell Physiol. 229:1202–1211. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Zmuda EJ, Qi L, Zhu MX, Mirmira RG, Montminy MR and Hai T: The roles of ATF3, an adaptive-response gene, in high-fat-diet-induced diabetes and pancreatic beta-cell dysfunction. Mol Endocrinol. 24:1423–1433. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Li HF, Cheng CF, Liao WJ, Lin H and Yang RB: ATF3-mediated epigenetic regulation protects against acute kidney injury. J Am Soc Nephrol. 21:1003–1013. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Shan Y, Akram A, Amatullah H, Zhou DY, Gali PL, Maron-Gutierrez T, González-López A, Zhou L, Rocco PR, Hwang D, et al: ATF3 protects pulmonary resident cells from acute and ventilator-induced lung injury by preventing Nrf2 degradation. Antioxid Redox Signal. 22:651–668. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Murray PJ and Wynn TA: Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 11:723–737. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Yan L, Della Coletta L, Powell KL, Shen J, Thames H, Aldaz CM and MacLeod MC: Activation of the canonical Wnt/β-catenin pathway in ATF3-induced mammary tumors. PLoS One. 6:e165152011. View Article : Google Scholar : PubMed/NCBI

32 

Lee H, Bae S, Choi BW and Yoon Y: WNT/β-catenin pathway is modulated in asthma patients and LPS-stimulated RAW264.7 macrophage cell line. Immunopharmacol Immunotoxicol. 34:56–65. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Carey WA, Taylor GD, Dean WB and Bristow JD: Tenascin-C deficiency attenuates TGF-ß-mediated fibrosis following murine lung injury. Am J Physiol Lung Cell Mol Physiol. 299:L785–L793. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Midwood K, Sacre S, Piccinini AM, Inglis J, Trebaul A, Chan E, Drexler S, Sofat N, Kashiwagi M, Orend G, et al: Tenascin-C is an endogenous activator of Toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease. Nature Med. 15:774–780. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Shimojo N, Hashizume R, Kanayama K, Suzuki Y, Hara M, Nishioka T, Yoshida T and Yoshida KI: A functional role of tenascin-C in angiotensin II-induced cardiac fibrosis. Circulation. 130:A96322014.

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
Sha H, Zhang D, Zhang Y, Wen Y and Wang Y: ATF3 promotes migration and M1/M2 polarization of macrophages by activating tenascin‑C via Wnt/β‑catenin pathway. Mol Med Rep 16: 3641-3647, 2017
APA
Sha, H., Zhang, D., Zhang, Y., Wen, Y., & Wang, Y. (2017). ATF3 promotes migration and M1/M2 polarization of macrophages by activating tenascin‑C via Wnt/β‑catenin pathway. Molecular Medicine Reports, 16, 3641-3647. https://doi.org/10.3892/mmr.2017.6992
MLA
Sha, H., Zhang, D., Zhang, Y., Wen, Y., Wang, Y."ATF3 promotes migration and M1/M2 polarization of macrophages by activating tenascin‑C via Wnt/β‑catenin pathway". Molecular Medicine Reports 16.3 (2017): 3641-3647.
Chicago
Sha, H., Zhang, D., Zhang, Y., Wen, Y., Wang, Y."ATF3 promotes migration and M1/M2 polarization of macrophages by activating tenascin‑C via Wnt/β‑catenin pathway". Molecular Medicine Reports 16, no. 3 (2017): 3641-3647. https://doi.org/10.3892/mmr.2017.6992