Association of Irisin/FNDC5 with ERRα and PGC-1α Expression in NSCLC

Katarzyna Nowińska, Karolina Jabłońska, Urszula Ciesielska, Aleksandra Piotrowska, Katarzyna Haczkiewicz-Leśniak, Konrad Pawełczyk, Marzenna Podhorska-Okołów, Piotr Dzięgiel, Katarzyna Nowińska, Karolina Jabłońska, Urszula Ciesielska, Aleksandra Piotrowska, Katarzyna Haczkiewicz-Leśniak, Konrad Pawełczyk, Marzenna Podhorska-Okołów, Piotr Dzięgiel

Abstract

The rapid growth and division of cancer cells are associated with mitochondrial biogenesis or switching to glycolysis. ERRα, PGC-1α and irisin/FNDC5 are some of the proteins that can influence these processes. The aim of this study was to determine the correlation of these proteins in non-small cell lung cancer (NSCLC) and to investigate their association with clinicopathological parameters. Immunohistochemistry reactions were performed on tissue microarrays (860 NSCLC, 140 non-malignant lung tissue). The normal fibroblast cell line (IMR-90) and lung cancer cell lines (NCI-H1703 and NCI-H522) were used as co-cultures. The mRNA levels of FNDC5 and ESRRA (encoding ERRα) were assessed in IMR-90 cells after co-culture with lung cancer cells. We observed a decreased level of ERRα with an increase in tumor size (T), stages of the disease, and lymph node metastases (N). In the adenocarcinoma (AC) subtype, patients with a higher ERRα expression had significantly longer overall survival. A moderate positive correlation was observed between FNDC5 mRNA and ESRRA mRNA in NSCLCs. The expression of FNDC5 mRNA in IMR-90 cells increased after 24 h, and ESRRA gene expression increased after 48 h of co-culture. The ERRα receptor with PGC-1α participates in the control of FNDC5/irisin expression. Normal fibroblasts revealed an upregulation of the FNDC5 and ESRRA genes under the influence of lung cancer cells.

Keywords: ERRα; ESRRA; FNDC5; NSCLC; PGC-1α; irisin; non-small cell lung cancer.

Conflict of interest statement

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Figures

Figure 1
Figure 1
Immunohistochemical reactions (IHC) indicating lack of irisin (A) and ERRα (B) expression. Weak positive IHC reaction for PGC-1α (C) in non-malignant lung tissue (NMLT). Positive cytoplasmic IHC reaction indicating irisin expression in skeletal muscle (positive control—D). Irisin expression in NSCLC cancer cells and stromal cells (grade of malignancy G1—G, G2—J, G3—M). Nuclear expression of ERRα in kidney (positive control—E). ERRα expression in NSCLC cancer cells (G1—H, G2—K, G3—N). Positive cytoplasmic expression of PGC-1α in prostate (positive control—F). PGC-1α in NSCLC cancer cells and stromal cells (G1—I, G2—L, G3—O). Magnification ×200. Arrows indicate a positive reaction.
Figure 2
Figure 2
Comparison of irisin expression levels detected by immunohistochemistry (IHC) in NSCLC (n = 860) cells (A,C,E,G) and in stromal cells (B,D,F,H) according to the tumor size (A,B), malignancy grade (C,D), lymph node status (E,F), and tumor stage (G,H), * p ≤ 0.05, ** p ≤ 0.005, *** p ≤ 0.001. Kaplan–Meier survival curves show the prognostic impact of irisin expression levels in cancer cells (I) and stromal cells (J) on overall survival (OS) in patients with NSCLC. Patients were grouped according to the median value of expression levels.
Figure 3
Figure 3
Comparison of ERRα receptor expression levels detected by immunohistochemistry (IHC) in non-small cell lung cancer NSCLC (n = 860, A,C,E,G) and adenocarcinoma subtype—AC (n = 344, B,D,F,H) according to the tumor size (A,B), the grade of malignancy (C,D), the lymph node status (E,F), and the tumor stage (G,H), * p ≤ 0.05, ** p ≤ 0.005, *** p ≤ 0.001.
Figure 4
Figure 4
Kaplan–Meier survival curves show the prognostic impact of ERRα expression levels on overall survival (OS) in patients with NSCLC (A), AC (D), SCC (G). Kaplan–Meier survival curves show the prognostic impact of PGC-1α expression levels in cancer cells on overall survival (OS) in patients with NSCLC (B), AC (E), SCC (H). Kaplan–Meier survival curves show the prognostic impact of PGC-1α expression levels in stromal cells on overall survival (OS) in patients with NSCLC (C), AC (F), SCC (I). Patients were grouped according to the median value of expression levels.
Figure 5
Figure 5
Comparison of PGC-1α expression levels detected by immunohistochemistry (IHC) in NSCLC (n = 860) cells (A,C,E,G) and in stromal cells (B,D,F,H) according to the tumor size (A,B), malignancy grade (C,D), lymph node status (E,F), and tumor stage (G,H). * p ≤ 0.05, ** p ≤ 0.005.
Figure 6
Figure 6
Correlations of ERRα receptor expression levels with diagnostic markers were strong positive—Ki-67 (A), moderate positive—EGFR (B), moderate positive—p63 (C) and weak positive—PD-L1 (D) in NSCLC (n = 860).
Figure 7
Figure 7
Correlations of PGC-1α expression levels in stromal cells with diagnostic markers were moderate positive—Ki-67 (A), weak positive—EGFR (B), weak positive—p63 (C) and weak positive—PD-L1 (D) in NSCLC (n = 860).
Figure 8
Figure 8
Correlations between irisin expressed in cancer cells with PGC-1α expressed in cancer cells (A) and stromal cells with PGC-1α (stromal cells) (B), and ERRα receptor (C). Correlations between ERRα and PGC-1α in NSCLC (n = 860) cancer cells (D).
Figure 9
Figure 9
Comparison between control (n = 16) and NSCLCs (n = 56) of FNDC5 mRNA (A) and ESRRA mRNA (B) expression levels. The moderate positive correlation between mRNA FNDC5 and mRNA ESRRA expression levels in NSCLC patients (C), * p ≤ 0.05, ** p ≤ 0.005.
Figure 10
Figure 10
Comparison of the expression level of FNDC5 mRNA after co-culture in IMR-90 cells in the empty insert (control) (A,B) and the insert with lung cancer cells [NCI-H1703 (C,D) and NCI-H522 (E,F)], * p ≤ 0.05, ** p ≤ 0.005, *** p < 0.001.
Figure 11
Figure 11
Comparison of the expression level of ESRRA mRNA after co-culture in IMR-90 cells in the empty insert (control) (A,B) and the insert with lung cancer cells [NCI-H1703 (C,D) and NCI-H522 (E,F)], * p ≤ 0.05, ** p ≤ 0.005.
Figure 12
Figure 12
Positive immunogold reaction (black dots—indicated by arrows) point to irisin/FNDC5 expression in the cell cytoplasm in NCI-H522 cells—magnification on the right (A), in NCI-H1703 cell mitochondria-M membrane—magnification on the right (B), in rough endoplasmic reticulum-RER and in cytoplasmic extensions of A549 cell (C), N-nucleus, magnification ×25,000.

References

    1. Herbst R.S., Morgensztern D., Boshoff C. The Biology and Management of Non-Small Cell Lung Cancer. Nature. 2018;553:446–454. doi: 10.1038/nature25183.
    1. Pawelczyk K., Piotrowska A., Ciesielska U., Jablonska K., Gletzel-Plucinska N., Grzegrzolka J., Podhorska-Okolow M., Dziegiel P., Nowinska K. Role of PD-L1 Expression in Non-Small Cell Lung Cancer and Their Prognostic Significance According to Clinicopathological Factors and Diagnostic Markers. Int. J. Mol. Sci. 2019;20:824. doi: 10.3390/ijms20040824.
    1. Bader J.E., Voss K., Rathmell J.C. Targeting Metabolism to Improve the Tumor Microenvironment for Cancer Immunotherapy. Mol. Cell. 2020;78:1019–1033. doi: 10.1016/j.molcel.2020.05.034.
    1. Cruz-Bermúdez A., Laza-Briviesca R., Vicente-Blanco R.J., García-Grande A., Coronado M.J., Laine-Menéndez S., Alfaro C., Sanchez J.C., Franco F., Calvo V., et al. Cancer-Associated Fibroblasts Modify Lung Cancer Metabolism Involving ROS and TGF-β Signaling. Free Radic. Biol. Med. 2019;130:163–173. doi: 10.1016/j.freeradbiomed.2018.10.450.
    1. Deblois G., St-Pierre J., Giguère V. The PGC-1/ERR Signaling Axis in Cancer. Oncogene. 2013;32:3483–3490. doi: 10.1038/onc.2012.529.
    1. Dang C.V. Links between Metabolism and Cancer. Genes Dev. 2012;26:877–890. doi: 10.1101/gad.189365.112.
    1. Boström P., Wu J., Jedrychowski M.P., Korde A., Ye L., Lo J.C., Rasbach K.A., Boström E.A., Choi J.H., Long J.Z., et al. A PGC1α-Dependent Myokine That Drives Browning of White Fat and Thermogenesis. Nature. 2012;481:463–468. doi: 10.1038/nature10777.
    1. Pinkowska A., Podhorska-Okołów M., Dzięgiel P., Nowińska K. The Role of Irisin in Cancer Disease. Cells. 2021;10:1479. doi: 10.3390/cells10061479.
    1. Suchanski J., Tejchman A., Zacharski M., Piotrowska A., Grzegrzolka J., Chodaczek G., Nowinska K., Rys J., Dziegiel P., Kieda C., et al. Podoplanin Increases the Migration of Human Fibroblasts and Affects the Endothelial Cell Network Formation: A Possible Role for Cancer-Associated Fibroblasts in Breast Cancer Progression. PLoS ONE. 2017;12:e0184970. doi: 10.1371/journal.pone.0184970.
    1. Nowinska K., Jablonska K., Pawelczyk K., Piotrowska A., Partynska A., Gomulkiewicz A., Ciesielska U., Katnik E., Grzegrzolka J., Glatzel-Plucinska N., et al. Expression of Irisin/FNDC5 in Cancer Cells and Stromal Fibroblasts of Non-Small Cell Lung Cancer. Cancers. 2019;11:1538. doi: 10.3390/cancers11101538.
    1. Boström P.A., Fernández-Real J.M., Mantzoros C. Irisin in Humans: Recent Advances and Questions for Future Research. Metab.-Clin. Exp. 2014;63:178–180. doi: 10.1016/j.metabol.2013.11.009.
    1. Aydin S. Is Irisin a Decisive Protein in Cancer Cachexia and Death of Cancer Cells? Eur. Rev. Med. Pharmacol. Sci. 2016;20:3727–3729.
    1. Aydin S., Kuloglu T., Ozercan M.R., Albayrak S., Aydin S., Bakal U., Yilmaz M., Kalayci M., Yardim M., Sarac M., et al. Irisin Immunohistochemistry in Gastrointestinal System Cancers. Biotech. Histochem. 2016;91:242–250. doi: 10.3109/10520295.2015.1136988.
    1. Tekin S., Erden Y., Sandal S., Yilmaz B. Is Irisin an Anticarcinogenic Peptide? Med. Sci. Int. Med. J. 2014;4:2172–2180. doi: 10.5455/medscience.2014.03.8210.
    1. Kuloglu T., Celik O., Aydin S., Hanifi Ozercan I., Acet M., Aydin Y., Artas G., Turk A., Yardim M., Ozan G., et al. Irisin Immunostaining Characteristics of Breast and Ovarian Cancer Cells. Cell. Mol. Biol. 2016;62:40–44. doi: 10.14715/cmb/.
    1. Hofmann T., Elbelt U., Stengel A. Irisin as a Muscle-Derived Hormone Stimulating Thermogenesis—A Critical Update. Peptides. 2014;54:89–100. doi: 10.1016/j.peptides.2014.01.016.
    1. Gaggini M., Cabiati M., Del Turco S., Navarra T., De Simone P., Filipponi F., Del Ry S., Gastaldelli A., Basta G. Increased FNDC5/Irisin Expression in Human Hepatocellular Carcinoma. Peptides. 2017;88:62–66. doi: 10.1016/j.peptides.2016.12.014.
    1. Pinkowska A., Nowinska K., Ciesielska U., Podhorska-okolow M. Irisin Association with Ki-67, MCM3 and MT-I/II in Squamous Cell Carcinomas of the Larynx. Biomolecules. 2022;12:52. doi: 10.3390/biom12010052.
    1. Wozniak S., Nowinska K., Chabowski M., Dziegiel P. Significance of Irisin (FNDC5) Expression in Colorectal Cancer. Vivo. 2022;36:180–188. doi: 10.21873/invivo.12689.
    1. Park E.J., Myint P.K., Ito A., Appiah M.G., Darkwah S., Kawamoto E., Shimaoka M. Integrin-Ligand Interactions in Inflammation, Cancer, and Metabolic Disease: Insights Into the Multifaceted Roles of an Emerging Ligand Irisin. Front. Cell Dev. Biol. 2020;8:588066. doi: 10.3389/fcell.2020.588066.
    1. Lee H.J., Lee J.O., Kim N., Kim J.K., Kim H.I., Lee Y.W., Kim S.J., Choi J.-I., Oh Y., Kim J.H., et al. Irisin, a Novel Myokine, Regulates Glucose Uptake in Skeletal Muscle Cells via AMPK. Mol. Endocrinol. 2015;29:873–881. doi: 10.1210/me.2014-1353.
    1. Wrann C.D., White J.P., Salogiannnis J., Laznik-Bogoslavski D., Wu J., Ma D., Lin J.D., Greenberg M.E., Spiegelman B.M. Exercise Induces Hippocampal BDNF through a PGC-1α/FNDC5 Pathway. Cell Metab. 2013;18:649–659. doi: 10.1016/j.cmet.2013.09.008.
    1. Xu B. BDNF (I)Rising from Exercise. Cell Metab. 2013;18:612–614. doi: 10.1016/j.cmet.2013.10.008.
    1. Arany Z., Foo S.Y., Ma Y., Ruas J.L., Bommi-Reddy A., Girnun G., Cooper M., Laznik D., Chinsomboon J., Rangwala S.M., et al. HIF-Independent Regulation of VEGF and Angiogenesis by the Transcriptional Coactivator PGC-1α. Nature. 2008;451:1008–1012. doi: 10.1038/nature06613.
    1. Zhang Z., Teng C.T. Estrogen Receptor α and Estrogen Receptor-Related Receptor A1 Compete for Binding and Coactivator. Mol. Cell. Endocrinol. 2001;172:223–233. doi: 10.1016/S0303-7207(00)00372-5.
    1. Bonnelye E., Vanacker J.M., Dittmar T., Begue A., Desbiens X., Denhardt D.T., Aubin J.E., Laudet V., Fournier B. The ERR-1 Orphan Receptor Is a Transcriptional Activator Expressed during Bone Development. Mol. Endocrinol. 1997;11:905–916. doi: 10.1210/mend.11.7.9948.
    1. Lynch C., Zhao J., Xia M. Cell-Based Assays to Identify ERR and ERR/PGC Modulators. Methods Mol. Biol. 2022;2474:3–9. doi: 10.1007/978-1-0716-2213-1_1.
    1. Ning Z., Du X., Zhang J., Yang K., Miao L., Zhu Y., Yuan H., Wang L., Klocker H., Shi J. PGE2 Modulates the Transcriptional Activity of ERRa in Prostate Stromal Cells. Endocrine. 2014;47:901–912. doi: 10.1007/s12020-014-0261-7.
    1. Wu Y.M., Chen Z.J., Liu H., Wei W.D., Lu L.L., Yang X.L., Liang W.T., Liu T., Liu H.L., Du J., et al. Inhibition of ERRa Suppresses Epithelial Mesenchymal Transition of Triple Negative Breast Cancer Cells by Directly Targeting Fibronectin. Oncotarget. 2015;6:25588–25601. doi: 10.18632/oncotarget.4436.
    1. Li P., Wang J., Wu D., Ren X., Wu W., Zuo R., Zeng Q., Wang B., He X., Yuan J., et al. ERRα Is an Aggressive Factor in Lung Adenocarcinoma Indicating Poor Prognostic Outcomes. Cancer Manag. Res. 2019;11:8111–8123. doi: 10.2147/CMAR.S204732.
    1. Shao L., Li H., Chen J., Song H., Zhang Y., Wu F., Wang W., Zhang W., Wang F., Li H., et al. Irisin Suppresses the Migration, Proliferation, and Invasion of Lung Cancer Cells via Inhibition of Epithelial-to-Mesenchymal Transition. Biochem. Biophys. Res. Commun. 2016;485:598–605. doi: 10.1016/j.bbrc.2016.12.084.
    1. Wahab F., Khan I.U., Polo I.R., Zubair H., Drummer C., Shahab M., Behr R. Irisin in the Primate Hypothalamus and Its Effect on GnRH In Vitro. J. Endocrinol. 2019;241:175–187. doi: 10.1530/JOE-18-0574.
    1. Herzig S., Shaw R.J. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 2018;19:121–135. doi: 10.1038/nrm.2017.95.
    1. Jablonska K., Nowinska K., Piotrowska A., Partynska A., Katnik E., Pawelczyk K., Kmiecik A., Glatzel-Plucinska N., Podhorska-Okolow M., Dziegiel P. Prognostic Impact of Melatonin Receptors MT1 and MT2 in Non-Small Cell Lung Cancer (NSCLC) Cancers. 2019;11:1001. doi: 10.3390/cancers11071001.
    1. Mukherjee T.K., Malik P., Hoidal J.R. The Emerging Role of Estrogen Related Receptorα in Complications of Non-Small Cell Lung Cancers (Review) Oncol. Lett. 2021;21:258. doi: 10.3892/ol.2021.12519.
    1. Yoriki K., Mori T., Kokabu T., Matsushima H., Umemura S., Tarumi Y., Kitawaki J. Estrogen-Related Receptor Alpha Induces Epithelial-Mesenchymal Transition through Cancer-Stromal Interactions in Endometrial Cancer. Sci. Rep. 2019;9:6697. doi: 10.1038/s41598-019-43261-z.
    1. Olsen C.J., Moreira J., Lukanidin E.M., Ambartsumian N.S. Human Mammary Fibroblasts Stimulate Invasion of Breast Cancer Cells in a Three-Dimensional Culture and Increase Stroma Development in Mouse Xenografts. BMC Cancer. 2010;10:444. doi: 10.1186/1471-2407-10-444.
    1. Matsushima H., Mori T., Ito F., Yamamoto T., Akiyama M., Kokabu T., Yoriki K., Umemura S., Akashi K., Kitawaki J. Anti-Tumor Effect of Estrogen-Related Receptor Alpha Knockdown on Uterine Endometrial Cancer. Oncotarget. 2016;7:34131–34148. doi: 10.18632/oncotarget.9151.
    1. Ariazi E.A., Clark G.M., Mertz J.E. Estrogen-Related Receptor α and Estrogen-Related Receptor γ Associate with Unfavorable and Favorable Biomarkers, Respectively, in Human Breast Cancer. Cancer Res. 2002;62:6510–6518.
    1. Fujimoto J., Alam S.M., Jahan I., Sato E., Sakaguchi H., Tamaya T. Clinical Implication of Estrogen-Related Receptor (ERR) Expression in Ovarian Cancers. J. Steroid Biochem. Mol. Biol. 2007;104:301–304. doi: 10.1016/j.jsbmb.2007.03.016.
    1. Suzuki S., Takagi K., Miki Y., Onodera Y., Akahira J.I., Ebata A., Ishida T., Watanabe M., Sasano H., Suzuki T. Estrogen-Related Receptor α in Human Breast Carcinoma as a Potent Prognostic Factor. Cancer Sci. 2012;103:136–143. doi: 10.1111/j.1349-7006.2011.02119.x.
    1. Remmele W., Stegner H.E. Recommendation for Uniform Definition of an Immunoreactive Score (IRS) for Immunohistochemical Estrogen Receptor Detection (ER-ICA) in Breast Cancer Tissue. Pathologe. 1987;8:138–140.

Source: PubMed

3
구독하다