Nrf2 Signaling Pathway in Chemoprotection and Doxorubicin Resistance: Potential Application in Drug Discovery
Abstract
:1. Introduction
2. Materials and Methods
3. Nrf2 Signaling Pathway
4. Nrf2 in Protection and Chemoresistance
5. Natural Compounds in Ameliorating Doxorubicin-Mediated Toxicity
6. Nrf2 Modulation
7. Nrf2 in Doxorubicin Resistance
8. Nrf2, Upstream and Downstream Targets
9. Room for Drug Discovery
10. Conclusions and Remarks
Author Contributions
Funding
Conflicts of Interest
Abbreviations
DOX | Doxorubicin |
ROS | reactive oxygen species |
TFAM | mitochondrial transcription factor A |
AMPK | AMP-activated protein kinase |
VASH2 | vasohibin2 |
SpK1 | sphingosine kinase 1 |
HIF-1α | hypoxia inducible factor-1α |
ncRNAs | non-coding RNAs |
lncRNAs | long non-coding RNAs |
miRNAs | microRNAs |
GI | gastrointestinal |
MMP-2 | matrix metalloproteinase-2 |
Nrf2 | nuclear factor erythroid 2-related factor 2 |
Keap1 | kelch-like ECH-associated protein 1 |
Cul3 | cullin3 |
GSK-3β | glycogen synthase-kinase 3β |
βTrCP | β transducing repeat containing E3 ubiquitin-protein ligase |
HO-1 | heme oxygenase-1 |
NQO1 | NAD(p)H dehydrogenase quinone 1 |
ARE | antioxidant response elemento |
PPARγ | peroxisome proliferator-activated receptor gama |
MDA | malondialdehyde |
SOD | superoxide dismutase |
CAT | catálase |
GSH | glutathione |
Pris | pristimerin |
AA | Asiatic acid |
B-LAP | β-LAPachone |
CAR | cardamonin |
COS | chitosan oligosaccharide |
pCA | p-coumaric acid |
LPS | lipopolysaccharide |
Tan IIA | tanshinone IIA |
PUN | punicalagin |
3/-UTR | 3/-untranslated region |
LC-3II | light chain-3II |
ORM1 | orosomucoid 1 |
3D | 3-dimensional |
PN | parthenolide |
P-gp | P-glycoprotein |
HSP70 | heat shock protein 70 |
PI3K | phosphatidylinositide 3-kinase |
Akt | protein kinase-B |
ERK | extracellular signal-regulated kinase |
VP | vialenin P |
MRP1 | multidrug resistance protein 1 |
APG | apigenin |
EMT | epithelial-to-mesenchymal transition |
siRNA | small interfering RNA |
CD44 | cluster of differentiation 44 |
ECM | extracellular matrix |
CSC | cancer stem cell |
SRF | serum response factor |
STAT3 | signal transducer and activator of transcription 3 |
LPA | lisophosphatidate |
ATX | autotaxin |
References
- Prathumsap, N.; Shinlapawittayatorn, K.; Chattipakorn, S.C.; Chattipakorn, N. Effects of doxorubicin on the heart: From molecular mechanisms to intervention strategies. Eur. J. Pharmacol. 2020, 866, 172818. [Google Scholar] [CrossRef]
- Denard, B.; Lee, C.; Ye, J. Doxorubicin blocks proliferation of cancer cells through proteolytic activation of CREB3L1. elife 2012, 1, e00090. [Google Scholar] [CrossRef]
- Lu, M.; Xie, K.; Lu, X.; Lu, L.; Shi, Y.; Tang, Y. Notoginsenoside R1 counteracts mesenchymal stem cell-evoked oncogenesis and doxorubicin resistance in osteosarcoma cells by blocking IL-6 secretion-induced JAK2/STAT3 signaling. Investig. New Drugs 2020. [Google Scholar] [CrossRef]
- Ghandhariyoun, N.; Jaafari, M.R.; Nikoofal-Sahlabadi, S.; Taghdisi, S.M.; Moosavian, S.A. Reducing Doxorubicin resistance in breast cancer by liposomal FOXM1 aptamer: In vitro and in vivo. Life Sci. 2020, 262, 118520. [Google Scholar] [CrossRef] [PubMed]
- Ashrafizadeh, M.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Saleki, H.; Bagherian, M.; Azami, N.; Bejandi, A.K.; Hushmandi, K.; Ang, H.L.; et al. Polychemotherapy with Curcumin and Doxorubicin via Biological Nanoplatforms: Enhancing Antitumor Activity. Pharmaceutics 2020, 12, 1084. [Google Scholar] [CrossRef]
- Poh, H.M.; Chiou, Y.S.; Chong, Q.Y.; Chen, R.M.; Rangappa, K.S.; Ma, L.; Zhu, T.; Kumar, A.P.; Pandey, V.; Lee, S.C.; et al. Inhibition of TFF3 Enhances Sensitivity-and Overcomes Acquired Resistance-to Doxorubicin in Estrogen Receptor-Positive Mammary Carcinoma. Cancers 2019, 11, 1528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, L.; Li, D.; Tang, P.; Zuo, Y. Curcumin increases the sensitivity of K562/DOX cells to doxorubicin by targeting S100 calcium-binding protein A8 and P-glycoprotein. Oncol. Lett. 2020, 19, 83–92. [Google Scholar] [CrossRef] [Green Version]
- Xinyong, C.; Zhiyi, Z.; Lang, H.; Peng, Y.; Xiaocheng, W.; Ping, Z.; Liang, S. The role of toll-like receptors in myocardial toxicity induced by doxorubicin. Immunol. Lett. 2020, 217, 56–64. [Google Scholar] [CrossRef]
- Wei, T.; Xiaojun, X.; Peilong, C. Magnoflorine improves sensitivity to doxorubicin (DOX) of breast cancer cells via inducing apoptosis and autophagy through AKT/mTOR and p38 signaling pathways. Biomed. Pharmacother. 2020, 121, 109139. [Google Scholar] [CrossRef]
- Abd-Rabou, A.A.; Ahmed, H.H.; Shalby, A.B. Selenium Overcomes Doxorubicin Resistance in Their Nano-platforms Against Breast and Colon Cancers. Biol. Trace Elem. Res. 2020, 193, 377–389. [Google Scholar] [CrossRef]
- Zhu, Y.; Xu, J.; Hu, W.; Wang, F.; Zhou, Y.; Xu, W.; Gong, W.; Shao, L. TFAM depletion overcomes hepatocellular carcinoma resistance to doxorubicin and sorafenib through AMPK activation and mitochondrial dysfunction. Gene 2020, 753, 144807. [Google Scholar] [CrossRef]
- Tan, X.; Liao, Z.; Zou, S.; Ma, L.; Wang, A. VASH2 Promotes Cell Proliferation and Resistance to Doxorubicin in Non-Small Cell Lung Cancer via AKT Signaling. Oncol. Res. 2020, 28, 3–11. [Google Scholar] [CrossRef]
- Tanaka, I.; Chakraborty, A.; Saulnier, O.; Benoit-Pilven, C.; Vacher, S.; Labiod, D.; Lam, E.W.F.; Bièche, I.; Delattre, O.; Pouzoulet, F.; et al. ZRANB2 and SYF2-mediated splicing programs converging on ECT2 are involved in breast cancer cell resistance to doxorubicin. Nucleic Acids Res. 2020, 48, 2676–2693. [Google Scholar] [CrossRef] [Green Version]
- Datta, A.; Loo, S.Y.; Huang, B.; Wong, L.; Tan, S.S.; Tan, T.Z.; Lee, S.C.; Thiery, J.P.; Lim, Y.C.; Yong, W.P.; et al. SPHK1 regulates proliferation and survival responses in triple-negative breast cancer. Oncotarget 2014, 5, 5920–5933. [Google Scholar] [CrossRef] [Green Version]
- Ma, Z.; Wang, L.Z.; Cheng, J.T.; Lam, W.S.T.; Ma, X.; Xiang, X.; Wong, A.L.; Goh, B.C.; Gong, Q.; Sethi, G.; et al. Targeting HIF-1-mediated Metastasis for Cancer Therapy. Antioxid. Redox Signal. 2020. [Google Scholar] [CrossRef]
- Ren, X.; Su, C. Sphingosine kinase 1 contributes to doxorubicin resistance and glycolysis in osteosarcoma. Mol. Med. Rep. 2020, 22, 2183–2190. [Google Scholar] [CrossRef]
- Wu, H.; Gu, J.; Zhou, D.; Cheng, W.; Wang, Y.; Wang, Q.; Wang, X. LINC00160 mediated paclitaxel-And doxorubicin-resistance in breast cancer cells by regulating TFF3 via transcription factor C/EBPβ. J. Cell. Mol. Med. 2020, 24, 8589–8602. [Google Scholar] [CrossRef]
- Wang, B.; Xu, L.; Zhang, J.; Cheng, X.; Xu, Q.; Wang, J.; Mao, F. LncRNA NORAD accelerates the progression and doxorubicin resistance of neuroblastoma through up-regulating HDAC8 via sponging miR-144-3p. Biomed. Pharmacother. 2020, 129, 110268. [Google Scholar] [CrossRef]
- Ong, M.S.; Cai, W.; Yuan, Y.; Leong, H.C.; Tan, T.Z.; Mohammad, A.; You, M.L.; Arfuso, F.; Goh, B.C.; Warrier, S.; et al. ‘Lnc’-ing Wnt in female reproductive cancers: Therapeutic potential of long non-coding RNAs in Wnt signalling. Br. J. Pharmacol. 2017, 174, 4684–4700. [Google Scholar] [CrossRef] [Green Version]
- Mishra, S.; Verma, S.S.; Rai, V.; Awasthee, N.; Chava, S.; Hui, K.M.; Kumar, A.P.; Challagundla, K.B.; Sethi, G.; Gupta, S.C. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell. Mol. Life Sci. 2019, 76, 1947–1966. [Google Scholar] [CrossRef]
- Ashrafizadeh, M.; Zarrabi, A.; Hushmandi, K.; Kalantari, M.; Mohammadinejad, R.; Javaheri, T.; Sethi, G. Association of the Epithelial-Mesenchymal Transition (EMT) with Cisplatin Resistance. Int. J. Mol. Sci. 2020, 21, 4002. [Google Scholar] [CrossRef]
- Li, Y.; Li, X.; Lu, Y.; Chaurasiya, B.; Mi, G.; Shi, D.; Chen, D.; Webster, T.J.; Tu, J.; Shen, Y. Co-delivery of Poria cocos extract and doxorubicin as an ‘all-in-one’ nanocarrier to combat breast cancer multidrug resistance during chemotherapy. Nanomedicine 2020, 23, 102095. [Google Scholar] [CrossRef] [PubMed]
- Pfister, C.; Gravis, G.; Fléchon, A.; Soulié, M.; Guy, L.; Laguerre, B.; Mottet, N.; Joly, F.; Allory, Y.; Harter, V.; et al. Randomized Phase III Trial of Dose-dense Methotrexate, Vinblastine, Doxorubicin, and Cisplatin, or Gemcitabine and Cisplatin as Perioperative Chemotherapy for Patients with Muscle-invasive Bladder Cancer. Analysis of the GETUG/AFU V05 VESPER Trial Secondary Endpoints: Chemotherapy Toxicity and Pathological Responses. Eur. Urol. 2020. [Google Scholar] [CrossRef]
- Gadisa, D.A.; Assefa, M.; Wang, S.H.; Yimer, G. Toxicity profile of Doxorubicin-Cyclophosphamide and Doxorubicin-Cyclophosphamide followed by Paclitaxel regimen and its associated factors among women with breast cancer in Ethiopia: A prospective cohort study. J. Oncol. Pharm. Pract. 2020, 26, 1912–1920. [Google Scholar] [CrossRef]
- Harahap, Y.; Ardiningsih, P.; Corintias Winarti, A.; Purwanto, D.J. Analysis of the Doxorubicin and Doxorubicinol in the Plasma of Breast Cancer Patients for Monitoring the Toxicity of Doxorubicin. Drug Des. Dev. Ther. 2020, 14, 3469–3475. [Google Scholar] [CrossRef]
- Li, X.Q.; Liu, Y.K.; Yi, J.; Dong, J.S.; Zhang, P.P.; Wan, L.; Li, K. MicroRNA-143 Increases Oxidative Stress and Myocardial Cell Apoptosis in a Mouse Model of Doxorubicin-Induced Cardiac Toxicity. Med. Sci. Monit. 2020, 26, e920394. [Google Scholar] [CrossRef]
- Chan, B.Y.H.; Roczkowsky, A.; Cho, W.J.; Poirier, M.; Sergi, C.; Keschrumrus, V.; Churko, J.M.; Granzier, H.; Schulz, R. MMP inhibitors attenuate doxorubicin cardiotoxicity by preventing intracellular and extracellular matrix remodeling. Cardiovasc. Res. 2020. [Google Scholar] [CrossRef]
- Boratto, F.A.; Franco, M.S.; Barros, A.L.B.; Cassali, G.D.; Malachias, A.; Ferreira, L.A.M.; Leite, E.A. Alpha-tocopheryl succinate improves encapsulation, pH-sensitivity, antitumor activity and reduces toxicity of doxorubicin-loaded liposomes. Eur. J. Pharm. Sci. 2020, 144, 105205. [Google Scholar] [CrossRef]
- Khan, T.H.; Ganaie, M.A.; Alharthy, K.M.; Madkhali, H.; Jan, B.L.; Sheikh, I.A. Naringenin prevents doxorubicin-induced toxicity in kidney tissues by regulating the oxidative and inflammatory insult in Wistar rats. Arch. Physiol. Biochem. 2020, 126, 300–307. [Google Scholar] [CrossRef]
- Oh, J.; Lee, B.S.; Lim, G.; Lim, H.; Lee, C.J.; Park, S.; Lee, S.H.; Chung, J.H.; Kang, S.M. Atorvastatin protects cardiomyocyte from doxorubicin toxicity by modulating survivin expression through FOXO1 inhibition. J. Mol. Cell. Cardiol. 2020, 138, 244–255. [Google Scholar] [CrossRef]
- Uruno, A.; Matsumaru, D.; Ryoke, R.; Saito, R.; Kadoguchi, S.; Saigusa, D.; Saito, T.; Saido, T.C.; Kawashima, R.; Yamamoto, M. Nrf2 Suppresses Oxidative Stress and Inflammation in App Knock-In Alzheimer’s Disease Model Mice. Mol. Cell. Biol. 2020, 40. [Google Scholar] [CrossRef]
- Zhou, Y.; Wang, K.; Zhou, Y.; Li, T.; Yang, M.; Wang, R.; Chen, Y.; Cao, M.; Hu, R. HEATR1 deficiency promotes pancreatic cancer proliferation and gemcitabine resistance by up-regulating Nrf2 signaling. Redox Biol. 2020, 29, 101390. [Google Scholar] [CrossRef]
- Hassanein, E.H.; Sayed, A.M.; Hussein, O.E.; Mahmoud, A.M. Coumarins as Modulators of the Keap1/Nrf2/ARE Signaling Pathway. Oxidative Med. Cell. Longev. 2020, 2020, 1675957. [Google Scholar] [CrossRef] [Green Version]
- Rosa, P.L.; Bertini, E.S.; Piemonte, F. The NRF2 Signaling Network Defines Clinical Biomarkers and Therapeutic Opportunity in Friedreich’s Ataxia. Int. J. Mol. Sci. 2020, 21, 916. [Google Scholar] [CrossRef] [Green Version]
- Kohandel, Z.; Farkhondeh, T.; Aschner, M.; Samarghandian, S. Nrf2 a molecular therapeutic target for Astaxanthin. Biomed. Pharmacother. 2021, 137, 111374. [Google Scholar] [CrossRef]
- Uddin, M.S.; Al Mamun, A.; Jakaria, M.; Thangapandiyan, S.; Ahmad, J.; Rahman, M.A.; Mathew, B.; Abdel-Daim, M.M.; Aleya, L. Emerging promise of sulforaphane-mediated Nrf2 signaling cascade against neurological disorders. Sci. Total Environ. 2020, 707, 135624. [Google Scholar] [CrossRef]
- Raghunath, A.; Sundarraj, K.; Arfuso, F.; Sethi, G.; Perumal, E. Dysregulation of Nrf2 in Hepatocellular Carcinoma: Role in Cancer Progression and Chemoresistance. Cancers 2018, 10, 481. [Google Scholar] [CrossRef] [Green Version]
- Kirtonia, A.; Sethi, G.; Garg, M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell. Mol. Life Sci. 2020, 77, 4459–4483. [Google Scholar] [CrossRef]
- Anandhan, A.; Dodson, M.; Schmidlin, C.J.; Liu, P.; Zhang, D.D. Breakdown of an Ironclad Defense System: The Critical Role of NRF2 in Mediating Ferroptosis. Cell. Chem. Biol. 2020, 27, 436–447. [Google Scholar] [CrossRef]
- Taguchi, K.; Yamamoto, M. The KEAP1-NRF2 System as a Molecular Target of Cancer Treatment. Cancers 2020, 13, 46. [Google Scholar] [CrossRef] [PubMed]
- Farkhondeh, T.; Folgado, S.L.; Pourbagher-Shahri, A.M.; Ashrafizadeh, M.; Samarghandian, S. The therapeutic effect of resveratrol: Focusing on the Nrf2 signaling pathway. Biomed. Pharmacother. 2020, 127, 110234. [Google Scholar] [CrossRef]
- Samarghandian, S.; Pourbagher-Shahri, A.M.; Ashrafizadeh, M.; Khan, H.; Forouzanfar, F.; Aramjoo, H.; Farkhondeh, T. A pivotal role of the nrf2 signaling pathway in spinal cord injury: A prospective therapeutics study. CNS Neurol. Disord. Drug Targets Former. Curr. Drug Targets-CNS Neurol. Disord. 2020, 19, 207–219. [Google Scholar] [CrossRef]
- Ahmadi, Z.; Ashrafizadeh, M. Melatonin as a potential modulator of Nrf2. Fundam. Clin. Pharmacol. 2020, 34, 11–19. [Google Scholar] [CrossRef]
- Ashrafizadeh, M.; Fekri, H.S.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Therapeutic and biological activities of berberine: The involvement of Nrf2 signaling pathway. J. Cell. Biochem. 2020, 121, 1575–1585. [Google Scholar] [CrossRef]
- Itoh, K.; Wakabayashi, N.; Katoh, Y.; Ishii, T.; Igarashi, K.; Engel, J.D.; Yamamoto, M. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999, 13, 76–86. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, A.; Kang, M.I.; Okawa, H.; Ohtsuji, M.; Zenke, Y.; Chiba, T.; Igarashi, K.; Yamamoto, M. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell. Biol. 2004, 24, 7130–7139. [Google Scholar] [CrossRef] [Green Version]
- Eggler, A.L.; Liu, G.; Pezzuto, J.M.; van Breemen, R.B.; Mesecar, A.D. Modifying specific cysteines of the electrophile-sensing human Keap1 protein is insufficient to disrupt binding to the Nrf2 domain Neh2. Proc. Natl. Acad. Sci. USA 2005, 102, 10070–10075. [Google Scholar] [CrossRef] [Green Version]
- Shaw, P.; Chattopadhyay, A. Nrf2-ARE signaling in cellular protection: Mechanism of action and the regulatory mechanisms. J. Cell. Physiol. 2020, 235, 3119–3130. [Google Scholar] [CrossRef]
- Cuadrado, A. Structural and functional characterization of Nrf2 degradation by glycogen synthase kinase 3/β-TrCP. Free Radic. Biol. Med. 2015, 88, 147–157. [Google Scholar] [CrossRef]
- Venugopal, R.; Jaiswal, A.K. Nrf2 and Nrf1 in association with Jun proteins regulate antioxidant response element-mediated expression and coordinated induction of genes encoding detoxifying enzymes. Oncogene 1998, 17, 3145–3156. [Google Scholar] [CrossRef] [Green Version]
- Saeedi, B.J.; Liu, K.H.; Owens, J.A.; Hunter-Chang, S.; Camacho, M.C.; Eboka, R.U.; Chandrasekharan, B.; Baker, N.F.; Darby, T.M.; Robinson, B.S.; et al. Gut-Resident Lactobacilli Activate Hepatic Nrf2 and Protect Against Oxidative Liver Injury. Cell. Metab. 2020, 31, 956–968.e955. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.; Wang, W.N.; Matei, N.; Li, X.; Pang, J.W.; Mo, J.; Chen, S.P.; Tang, J.P.; Yan, M.; Zhang, J.H. Ezetimibe Attenuates Oxidative Stress and Neuroinflammation via the AMPK/Nrf2/TXNIP Pathway after MCAO in Rats. Oxidative Med. Cell. Longev. 2020, 2020, 4717258. [Google Scholar] [CrossRef] [PubMed]
- Alcaraz, M.J.; Ferrándiz, M.L. Relevance of Nrf2 and heme oxygenase-1 in articular diseases. Free Radic. Biol. Med. 2020, 157, 83–93. [Google Scholar] [CrossRef]
- Kasai, S.; Shimizu, S.; Tatara, Y.; Mimura, J.; Itoh, K. Regulation of Nrf2 by Mitochondrial Reactive Oxygen Species in Physiology and Pathology. Biomolecules 2020, 10, 320. [Google Scholar] [CrossRef] [Green Version]
- Ahn, K.S.; Sethi, G.; Jain, A.K.; Jaiswal, A.K.; Aggarwal, B.B. Genetic deletion of NAD(P)H:quinone oxidoreductase 1 abrogates activation of nuclear factor-kappaB, IkappaBalpha kinase, c-Jun N-terminal kinase, Akt, p38, and p44/42 mitogen-activated protein kinases and potentiates apoptosis. .J. Biol. Chem. 2006, 281, 19798–19808. [Google Scholar] [CrossRef] [Green Version]
- Soares, M.P.; Seldon, M.P.; Gregoire, I.P.; Vassilevskaia, T.; Berberat, P.O.; Yu, J.; Tsui, T.Y.; Bach, F.H. Heme oxygenase-1 modulates the expression of adhesion molecules associated with endothelial cell activation. J. Immunol. 2004, 172, 3553–3563. [Google Scholar] [CrossRef] [Green Version]
- Li, F.; Shanmugam, M.K.; Chen, L.; Chatterjee, S.; Basha, J.; Kumar, A.P.; Kundu, T.K.; Sethi, G. Garcinol, a polyisoprenylated benzophenone modulates multiple proinflammatory signaling cascades leading to the suppression of growth and survival of head and neck carcinoma. Cancer Prev. Res. 2013, 6, 843–854. [Google Scholar] [CrossRef] [Green Version]
- Shanmugam, M.K.; Ong, T.H.; Kumar, A.P.; Lun, C.K.; Ho, P.C.; Wong, P.T.; Hui, K.M.; Sethi, G. Ursolic acid inhibits the initiation, progression of prostate cancer and prolongs the survival of TRAMP mice by modulating pro-inflammatory pathways. PLoS ONE 2012, 7, e32476. [Google Scholar] [CrossRef] [PubMed]
- Pan, H.; Wang, H.; Wang, X.; Zhu, L.; Mao, L. The absence of Nrf2 enhances NF-κB-dependent inflammation following scratch injury in mouse primary cultured astrocytes. Mediat. Inflamm. 2012, 2012, 217580. [Google Scholar] [CrossRef]
- Abd El-Twab, S.M.; Hussein, O.E.; Hozayen, W.G.; Bin-Jumah, M.; Mahmoud, A.M. Chicoric acid prevents methotrexate-induced kidney injury by suppressing NF-κB/NLRP3 inflammasome activation and up-regulating Nrf2/ARE/HO-1 signaling. Inflamm. Res. 2019, 68, 511–523. [Google Scholar] [CrossRef]
- Mahmoud, A.M.; Hussein, O.E.; Abd El-Twab, S.M.; Hozayen, W.G. Ferulic acid protects against methotrexate nephrotoxicity via activation of Nrf2/ARE/HO-1 signaling and PPARγ, and suppression of NF-κB/NLRP3 inflammasome axis. Food Funct. 2019, 10, 4593–4607. [Google Scholar] [CrossRef]
- Aladaileh, S.H.; Abukhalil, M.H.; Saghir, S.A.M.; Hanieh, H.; Alfwuaires, M.A.; Almaiman, A.A.; Bin-Jumah, M.; Mahmoud, A.M. Galangin Activates Nrf2 Signaling and Attenuates Oxidative Damage, Inflammation, and Apoptosis in a Rat Model of Cyclophosphamide-Induced Hepatotoxicity. Biomolecules 2019, 9, 346. [Google Scholar] [CrossRef] [Green Version]
- HAS, A.L.; Alotaibi, M.F.; Bin-Jumah, M.; Elgebaly, H.; Mahmoud, A.M. Olea europaea leaf extract up-regulates Nrf2/ARE/HO-1 signaling and attenuates cyclophosphamide-induced oxidative stress, inflammation and apoptosis in rat kidney. Biomed. Pharmacother. 2019, 111, 676–685. [Google Scholar] [CrossRef]
- Ranneh, Y.; Akim, A.M.; Hamid, H.A.; Khazaai, H.; Fadel, A.; Mahmoud, A.M. Stingless bee honey protects against lipopolysaccharide induced-chronic subclinical systemic inflammation and oxidative stress by modulating Nrf2, NF-κB and p38 MAPK. Nutr. Metab. 2019, 16, 15. [Google Scholar] [CrossRef]
- Mahmoud, A.M.; Germoush, M.O.; Al-Anazi, K.M.; Mahmoud, A.H.; Farah, M.A.; Allam, A.A. Commiphora molmol protects against methotrexate-induced nephrotoxicity by up-regulating Nrf2/ARE/HO-1 signaling. Biomed. Pharmacother. 2018, 106, 499–509. [Google Scholar] [CrossRef]
- Manu, K.A.; Shanmugam, M.K.; Li, F.; Chen, L.; Siveen, K.S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. J. Mol. Med. 2014, 92, 267–276. [Google Scholar] [CrossRef] [Green Version]
- Dai, X.; Ahn, K.S.; Wang, L.Z.; Kim, C.; Deivasigamni, A.; Arfuso, F.; Um, J.Y.; Kumar, A.P.; Chang, Y.C.; Kumar, D.; et al. Ascochlorin Enhances the Sensitivity of Doxorubicin Leading to the Reversal of Epithelial-to-Mesenchymal Transition in Hepatocellular Carcinoma. Mol. Cancer Ther. 2016, 15, 2966–2976. [Google Scholar] [CrossRef] [Green Version]
- Manu, K.A.; Shanmugam, M.K.; Ramachandran, L.; Li, F.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Arfuso, F.; Kumar, A.P.; et al. Isorhamnetin augments the anti-tumor effect of capecitabine through the negative regulation of NF-κB signaling cascade in gastric cancer. Cancer lett. 2015, 363, 28–36. [Google Scholar] [CrossRef]
- Pinto, V.; Bergantim, R.; Caires, H.R.; Seca, H.; Guimarães, J.E.; Vasconcelos, M.H. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers 2020, 12, 407. [Google Scholar] [CrossRef] [Green Version]
- Wei, L.; Sun, J.; Zhang, N.; Zheng, Y.; Wang, X.; Lv, L.; Liu, J.; Xu, Y.; Shen, Y.; Yang, M. Noncoding RNAs in gastric cancer: Implications for drug resistance. Mol. Cancer 2020, 19, 62. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Y.Q.; Liu, D.Q.; Chen, S.P.; Chen, N.; Sun, J.; Wang, X.M.; Cao, F.; Tian, Y.K.; Ye, D.W. Nrf2 activation ameliorates mechanical allodynia in paclitaxel-induced neuropathic pain. Acta Pharmacol. Sin. 2020, 41, 1041–1048. [Google Scholar] [CrossRef]
- Zhou, Y.Q.; Liu, D.Q.; Chen, S.P.; Chen, N.; Sun, J.; Wang, X.M.; Li, D.Y.; Tian, Y.K.; Ye, D.W. PPARγ activation mitigates mechanical allodynia in paclitaxel-induced neuropathic pain via induction of Nrf2/HO-1 signaling pathway. Biomed. Pharmacother. 2020, 129, 110356. [Google Scholar] [CrossRef]
- Mohamed, M.E.; Abduldaium, Y.S.; Younis, N.S. Ameliorative Effect of Linalool in Cisplatin-Induced Nephrotoxicity: The Role of HMGB1/TLR4/NF-κB and Nrf2/HO1 Pathways. Biomolecules 2020, 10, 1488. [Google Scholar] [CrossRef] [PubMed]
- Abdel-Wahab, B.A.; Alkahtani, S.A.; Elagab, E.A.M. Tadalafil alleviates cisplatin-induced reproductive toxicity through the activation of the Nrf2/HO-1 pathway and the inhibition of oxidative stress and apoptosis in male rats. Reprod. Toxicol. 2020, 96, 165–174. [Google Scholar] [CrossRef] [PubMed]
- Azouz, A.A.; Abdel-Nassir Abdel-Razek, E.; Abo-Youssef, A.M. Amlodipine alleviates cisplatin-induced nephrotoxicity in rats through gamma-glutamyl transpeptidase (GGT) enzyme inhibition, associated with regulation of Nrf2/HO-1, MAPK/NF-κB, and Bax/Bcl-2 signaling. Saudi Pharm. J. 2020, 28, 1317–1325. [Google Scholar] [CrossRef]
- Lu, Y.; Wu, S.; Xiang, B.; Li, L.; Lin, Y. Curcumin Attenuates Oxaliplatin-Induced Liver Injury and Oxidative Stress by Activating the Nrf2 Pathway. Drug Des. Dev. Ther. 2020, 14, 73–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fang, Y.; Ye, J.; Zhao, B.; Sun, J.; Gu, N.; Chen, X.; Ren, L.; Chen, J.; Cai, X.; Zhang, W.; et al. Formononetin ameliorates oxaliplatin-induced peripheral neuropathy via the KEAP1-NRF2-GSTP1 axis. Redox Biol. 2020, 36, 101677. [Google Scholar] [CrossRef]
- Yu, Y.P.; Cai, L.C.; Wang, X.Y.; Cheng, S.Y.; Zhang, D.M.; Jian, W.G.; Wang, T.D.; Yang, J.K.; Yang, K.B.; Zhang, C. BMP8A promotes survival and drug resistance via Nrf2/TRIM24 signaling pathway in clear cell renal cell carcinoma. Cancer Sci. 2020, 111, 1555–1566. [Google Scholar] [CrossRef] [Green Version]
- Leung, H.W.; Lau, E.Y.T.; Leung, C.O.N.; Lei, M.M.L.; Mok, E.H.K.; Ma, V.W.S.; Cho, W.C.S.; Ng, I.O.L.; Yun, J.P.; Cai, S.H.; et al. NRF2/SHH signaling cascade promotes tumor-initiating cell lineage and drug resistance in hepatocellular carcinoma. Cancer Lett. 2020, 476, 48–56. [Google Scholar] [CrossRef]
- Jiang, G.; Liang, X.; Huang, Y.; Lan, Z.; Zhang, Z.; Su, Z.; Fang, Z.; Lai, Y.; Yao, W.; Liu, T.; et al. p62 promotes proliferation, apoptosis‑resistance and invasion of prostate cancer cells through the Keap1/Nrf2/ARE axis. Oncol. Rep. 2020, 43, 1547–1557. [Google Scholar] [CrossRef] [PubMed]
- Mukhopadhyay, S.; Goswami, D.; Adiseshaiah, P.P.; Burgan, W.; Yi, M.; Guerin, T.M.; Kozlov, S.V.; Nissley, D.V.; McCormick, F. Undermining Glutaminolysis Bolsters Chemotherapy While NRF2 Promotes Chemoresistance in KRAS-Driven Pancreatic Cancers. Cancer Res. 2020, 80, 1630–1643. [Google Scholar] [CrossRef] [Green Version]
- Escoll, M.; Lastra, D.; Pajares, M.; Robledinos-Antón, N.; Rojo, A.I.; Fernández-Ginés, R.; Mendiola, M.; Martínez-Marín, V.; Esteban, I.; López-Larrubia, P.; et al. Transcription factor NRF2 uses the Hippo pathway effector TAZ to induce tumorigenesis in glioblastomas. Redox Biol. 2020, 30, 101425. [Google Scholar] [CrossRef]
- Cucci, M.A.; Grattarola, M.; Dianzani, C.; Damia, G.; Ricci, F.; Roetto, A.; Trotta, F.; Barrera, G.; Pizzimenti, S. Ailanthone increases oxidative stress in CDDP-resistant ovarian and bladder cancer cells by inhibiting of Nrf2 and YAP expression through a post-translational mechanism. Free Radic. Biol. Med. 2020, 150, 125–135. [Google Scholar] [CrossRef]
- Fouzder, C.; Mukhuty, A.; Kundu, R. Kaempferol inhibits Nrf2 signalling pathway via downregulation of Nrf2 mRNA and induces apoptosis in NSCLC cells. Arch. Biochem. Biophys. 2020, 697, 108700. [Google Scholar] [CrossRef] [PubMed]
- Ma, C.S.; Lv, Q.M.; Zhang, K.R.; Tang, Y.B.; Zhang, Y.F.; Shen, Y.; Lei, H.M.; Zhu, L. NRF2-GPX4/SOD2 axis imparts resistance to EGFR-tyrosine kinase inhibitors in non-small-cell lung cancer cells. Acta Pharmacol. Sin. 2020. [Google Scholar] [CrossRef]
- Jiang, C.; Luo, P.; Li, X.; Liu, P.; Li, Y.; Xu, J. Nrf2/ARE is a key pathway for curcumin-mediated protection of TMJ chondrocytes from oxidative stress and inflammation. Cell Stress Chaperones 2020, 25, 395–406. [Google Scholar] [CrossRef]
- Rasheed, M.S.U.; Tripathi, M.K.; Patel, D.K.; Singh, M.P. Resveratrol Regulates Nrf2-Mediated Expression of Antioxidant and Xenobiotic Metabolizing Enzymes in Pesticides-Induced Parkinsonism. Protein Pept. Lett. 2020, 27, 1038–1045. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; Li, Y.; Mu, X. Effect of Quercetin on PC12 Alzheimer’s Disease Cell Model Induced by Aβ (25-35) and Its Mechanism Based on Sirtuin1/Nrf2/HO-1 Pathway. Biomed. Res. Int. 2020, 2020, 8210578. [Google Scholar] [CrossRef]
- Costa, P.M.; Ferreira, P.M.; Bolzani Vda, S.; Furlan, M.; de Freitas Formenton Macedo Dos Santos, V.A.; Corsino, J.; de Moraes, M.O.; Costa-Lotufo, L.V.; Montenegro, R.C.; Pessoa, C. Antiproliferative activity of pristimerin isolated from Maytenus ilicifolia (Celastraceae) in human HL-60 cells. Toxicol. In Vitro 2008, 22, 854–863. [Google Scholar] [CrossRef]
- Tong, L.; Nanjundaiah, S.M.; Venkatesha, S.H.; Astry, B.; Yu, H.; Moudgil, K.D. Pristimerin, a naturally occurring triterpenoid, protects against autoimmune arthritis by modulating the cellular and soluble immune mediators of inflammation and tissue damage. Clin. Immunol. 2014, 155, 220–230. [Google Scholar] [CrossRef]
- El-Agamy, D.S.; El-Harbi, K.M.; Khoshhal, S.; Ahmed, N.; Elkablawy, M.A.; Shaaban, A.A.; Abo-Haded, H.M. Pristimerin protects against doxorubicin-induced cardiotoxicity and fibrosis through modulation of Nrf2 and MAPK/NF-kB signaling pathways. Cancer Manag. Res. 2019, 11, 47–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aktaş, I.; Özmen, Ö.; Tutun, H.; Yalçın, A.; Türk, A. Artemisinin attenuates doxorubicin induced cardiotoxicity and hepatotoxicity in rats. Biotech. Histochem. 2020, 95, 121–128. [Google Scholar] [CrossRef] [PubMed]
- Prospero, A.G.; Fidelis-de-Oliveira, P.; Soares, G.A.; Miranda, M.F.; Pinto, L.A.; Dos Santos, D.C.; Silva, V.D.S.; Zufelato, N.; Bakuzis, A.F.; Miranda, J.R. AC biosusceptometry and magnetic nanoparticles to assess doxorubicin-induced kidney injury in rats. Nanomedicine 2020, 15, 511–525. [Google Scholar] [CrossRef]
- Oh, S.-K.; Rha, C.; Kadir, A.; Ng, T. Use of Asiatic Acid or Asiaticoside for Treatment of Cancer. U.S. Patent Application No. 10/362,720, 20 May 2004. [Google Scholar]
- Han, Y.; Jiang, Y.; Li, Y.; Wang, M.; Fan, T.; Liu, M.; Ke, Q.; Xu, H.; Yi, Z. An aligned porous electrospun fibrous scaffold with embedded asiatic acid for accelerating diabetic wound healing. J. Mater. Chem. B 2019, 7, 6125–6138. [Google Scholar] [CrossRef]
- Hu, W.Y.; Li, X.X.; Diao, Y.F.; Qi, J.J.; Wang, D.L.; Zhang, J.B.; Sun, B.X.; Liang, S. Asiatic acid protects oocytes against in vitro aging-induced deterioration and improves subsequent embryonic development in pigs. Aging 2020, 12. [Google Scholar] [CrossRef]
- Lian, G.Y.; Wang, Q.M.; Tang, P.M.; Zhou, S.; Huang, X.R.; Lan, H.Y. Combination of Asiatic Acid and Naringenin Modulates NK Cell Anti-cancer Immunity by Rebalancing Smad3/Smad7 Signaling. Mol. Ther. 2018, 26, 2255–2266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kamble, S.M.; Patil, C.R. Asiatic Acid Ameliorates Doxorubicin-Induced Cardiac and Hepato-Renal Toxicities with Nrf2 Transcriptional Factor Activation in Rats. Cardiovasc. Toxicol. 2018, 18, 131–141. [Google Scholar] [CrossRef]
- Song, C.; Heping, H.; Shen, Y.; Jin, S.; Li, D.; Zhang, A.; Ren, X.; Wang, K.; Zhang, L.; Wang, J.; et al. AMPK/p38/Nrf2 activation as a protective feedback to restrain oxidative stress and inflammation in microglia stimulated with sodium fluoride. Chemosphere 2020, 244, 125495. [Google Scholar] [CrossRef]
- Lee, E.H.; Baek, S.Y.; Park, J.Y.; Kim, Y.W. Rifampicin activates AMPK and alleviates oxidative stress in the liver as mediated with Nrf2 signaling. Chem. Biol. Interact. 2020, 315, 108889. [Google Scholar] [CrossRef]
- Nazari Soltan Ahmad, S.; Sanajou, D.; Kalantary-Charvadeh, A.; Hosseini, V.; Roshangar, L.; Khojastehfard, M.; Haiaty, S.; Mesgari-Abbasi, M. β-LAPachone ameliorates doxorubicin-induced cardiotoxicity via regulating autophagy and Nrf2 signalling pathways in mice. Basic Clin. Pharmacol. Toxicol. 2020, 126, 364–373. [Google Scholar] [CrossRef]
- Wu, Z.; Zai, W.; Chen, W.; Han, Y.; Jin, X.; Liu, H. Curdione Ameliorated Doxorubicin-Induced Cardiotoxicity Through Suppressing Oxidative Stress and Activating Nrf2/HO-1 Pathway. J. Cardiovasc. Pharmacol. 2019, 74, 118–127. [Google Scholar] [CrossRef]
- Yoo, O.K.; Choi, W.J.; Keum, Y.S. Cardamonin Inhibits Oxazolone-Induced Atopic Dermatitis by the Induction of NRF2 and the Inhibition of Th2 Cytokine Production. Antioxidants 2020, 9, 834. [Google Scholar] [CrossRef] [PubMed]
- Tan, Y.; Wan, H.H.; Sun, M.M.; Zhang, W.J.; Dong, M.; Ge, W.; Ren, J.; Peng, H. Cardamonin protects against lipopolysaccharide-induced myocardial contractile dysfunction in mice through Nrf2-regulated mechanism. Acta Pharmacol. Sin. 2020. [Google Scholar] [CrossRef]
- Qi, W.; Boliang, W.; Xiaoxi, T.; Guoqiang, F.; Jianbo, X.; Gang, W. Cardamonin protects against doxorubicin-induced cardiotoxicity in mice by restraining oxidative stress and inflammation associated with Nrf2 signaling. Biomed. Pharmacother. 2020, 122, 109547. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Meng, Q.; Li, Q.; Liu, J.; Zhou, M.; Jin, Z.; Zhao, K. Chitosan derivatives and their application in biomedicine. Int. J. Mol. Sci. 2020, 21, 487. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Liu, H.; Xu, Q.-S.; Du, Y.-G.; Xu, J. Chitosan oligosaccharides block LPS-induced O-GlcNAcylation of NF-κB and endothelial inflammatory response. Carbohydr. Polym. 2014, 99, 568–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernandes, J.C.; Sereno, J.; Garrido, P.; Parada, B.; Cunha, M.F.; Reis, F.; Pintado, M.E.; Santos-Silva, A. Inhibition of bladder tumor growth by chitooligosaccharides in an experimental carcinogenesis model. Marine Drugs 2012, 10, 2661–2675. [Google Scholar] [CrossRef] [Green Version]
- Muanprasat, C.; Chatsudthipong, V. Chitosan oligosaccharide: Biological activities and potential therapeutic applications. Pharmacol. Ther. 2017, 170, 80–97. [Google Scholar] [CrossRef]
- Zou, P.; Yang, X.; Wang, J.; Li, Y.; Yu, H.; Zhang, Y.; Liu, G. Advances in characterisation and biological activities of chitosan and chitosan oligosaccharides. Food Chem. 2016, 190, 1174–1181. [Google Scholar] [CrossRef]
- Fang, I.-M.; Yang, C.-M.; Yang, C.-H. Chitosan oligosaccharides prevented retinal ischemia and reperfusion injury via reduced oxidative stress and inflammation in rats. Exp. Eye Res. 2015, 130, 38–50. [Google Scholar] [CrossRef]
- Zhang, Y.; Ahmad, K.A.; Khan, F.U.; Yan, S.; Ihsan, A.U.; Ding, Q. Chitosan oligosaccharides prevent doxorubicin-induced oxidative stress and cardiac apoptosis through activating p38 and JNK MAPK mediated Nrf2/ARE pathway. Chem. Biol. Interact. 2019, 305, 54–65. [Google Scholar] [CrossRef]
- Wang, L.F.; Su, S.W.; Wang, L.; Zhang, G.Q.; Zhang, R.; Niu, Y.J.; Guo, Y.S.; Li, C.Y.; Jiang, W.B.; Liu, Y.; et al. Tert-butylhydroquinone ameliorates doxorubicin-induced cardiotoxicity by activating Nrf2 and inducing the expression of its target genes. Am. J. Transl. Res. 2015, 7, 1724–1735. [Google Scholar]
- Hajra, S.; Basu, A.; Singha Roy, S.; Patra, A.R.; Bhattacharya, S. Attenuation of doxorubicin-induced cardiotoxicity and genotoxicity by an indole-based natural compound 3,3′-diindolylmethane (DIM) through activation of Nrf2/ARE signaling pathways and inhibiting apoptosis. Free Radic. Res. 2017, 51, 812–827. [Google Scholar] [CrossRef]
- Kanski, J.; Aksenova, M.; Stoyanova, A.; Butterfield, D.A. Ferulic acid antioxidant protection against hydroxyl and peroxyl radical oxidation in synaptosomal and neuronal cell culture systems in vitro: Structure-activity studies. J. Nutr. Biochem. 2002, 13, 273–281. [Google Scholar] [CrossRef]
- Kheiry, M.; Dianat, M.; Badavi, M.; Mard, S.A.; Bayati, V. p-Coumaric Acid Attenuates Lipopolysaccharide-Induced Lung Inflammation in Rats by Scavenging ROS Production: An In Vivo and In Vitro Study. Inflammation 2019, 42, 1939–1950. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.J.; Deng, J.S.; Huang, C.C.; Li, P.Y.; Liang, Y.C.; Chou, C.Y.; Huang, G.J. p-Coumaric-Acid-Containing Adenostemma lavenia Ameliorates Acute Lung Injury by Activating AMPK/Nrf2/HO-1 Signaling and Improving the Anti-oxidant Response. Am. J. Chin. Med. 2019, 47, 1483–1506. [Google Scholar] [CrossRef] [PubMed]
- Sunitha, M.C.; Dhanyakrishnan, R.; PrakashKumar, B.; Nevin, K.G. p-Coumaric acid mediated protection of H9c2 cells from Doxorubicin-induced cardiotoxicity: Involvement of augmented Nrf2 and autophagy. Biomed. Pharmacother. 2018, 102, 823–832. [Google Scholar] [CrossRef]
- Elsherbiny, N.M.; El-Sherbiny, M. Thymoquinone attenuates Doxorubicin-induced nephrotoxicity in rats: Role of Nrf2 and NOX4. Chem. Biol. Interact. 2014, 223, 102–108. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.; Liu, P. Tanshinone II-A: New perspectives for old remedies. Expert Opin. Ther. Pat. 2013, 23, 149–153. [Google Scholar] [CrossRef] [PubMed]
- Feng, F.; Cheng, P.; Zhang, H.; Li, N.; Qi, Y.; Wang, H.; Wang, Y.; Wang, W. The Protective Role of Tanshinone IIA in Silicosis Rat Model via TGF-β1/Smad Signaling Suppression, NOX4 Inhibition and Nrf2/ARE Signaling Activation. Drug Des. Dev. Ther. 2019, 13, 4275–4290. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Liu, L.; Zhang, X.; Jiang, X.; Wang, L. Tanshinone IIA prevents rifampicin-induced liver injury by regulating BSEP/NTCP expression via epigenetic activation of NRF2. Liver Int. 2020, 40, 141–154. [Google Scholar] [CrossRef]
- An, L.; Peng, L.Y.; Sun, N.Y.; Yang, Y.L.; Zhang, X.W.; Li, B.; Liu, B.L.; Li, P.; Chen, J. Tanshinone IIA Activates Nuclear Factor-Erythroid 2-Related Factor 2 to Restrain Pulmonary Fibrosis via Regulation of Redox Homeostasis and Glutaminolysis. Antioxid. Redox Signal. 2019, 30, 1831–1848. [Google Scholar] [CrossRef] [PubMed]
- Guo, Z.; Yan, M.; Chen, L.; Fang, P.; Li, Z.; Wan, Z.; Cao, S.; Hou, Z.; Wei, S.; Li, W.; et al. Nrf2-dependent antioxidant response mediated the protective effect of tanshinone IIA on doxorubicin-induced cardiotoxicity. Exp. Ther. Med. 2018, 16, 3333–3344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feng, X.; Yang, Q.; Wang, C.; Tong, W.; Xu, W. Punicalagin Exerts Protective Effects against Ankylosing Spondylitis by Regulating NF-κB-TH17/JAK2/STAT3 Signaling and Oxidative Stress. Biomed. Res. Int. 2020, 2020, 4918239. [Google Scholar] [CrossRef]
- Wang, W.; Bai, J.; Zhang, W.; Ge, G.; Wang, Q.; Liang, X.; Li, N.; Gu, Y.; Li, M.; Xu, W.; et al. Protective Effects of Punicalagin on Osteoporosis by Inhibiting Osteoclastogenesis and Inflammation via the NF-κB and MAPK Pathways. Front. Pharmacol. 2020, 11, 696. [Google Scholar] [CrossRef] [PubMed]
- Ye, M.; Zhang, L.; Yan, Y.; Lin, H. Punicalagin protects H9c2 cardiomyocytes from doxorubicin-induced toxicity through activation of Nrf2/HO-1 signaling. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [Green Version]
- Arnold, J.; Engelmann, J.C.; Schneider, N.; Bosserhoff, A.K.; Kuphal, S. miR-488-5p and its role in melanoma. Exp. Mol. Pathol. 2020, 112, 104348. [Google Scholar] [CrossRef]
- Kolenda, T.; Guglas, K.; Kopczyńska, M.; Sobocińska, J.; Teresiak, A.; Bliźniak, R.; Lamperska, K. Good or not good: Role of miR-18a in cancer biology. Rep. Pract. Oncol. Radiother. 2020, 25, 808–819. [Google Scholar] [CrossRef]
- Ashrafizadeh, M.; Hushmandi, K.; Hashemi, M.; Akbari, M.E.; Kubatka, P.; Raei, M.; Koklesova, L.; Shahinozzaman, M.; Mohammadinejad, R.; Najafi, M.; et al. Role of microRNA/Epithelial-to-Mesenchymal Transition Axis in the Metastasis of Bladder Cancer. Biomolecules 2020, 10, 1159. [Google Scholar] [CrossRef]
- Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.A.; Sethi, G. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules 2019, 9, 735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dastmalchi, N.; Hosseinpourfeizi, M.A.; Khojasteh, S.M.B.; Baradaran, B.; Safaralizadeh, R. Tumor suppressive activity of miR-424-5p in breast cancer cells through targeting PD-L1 and modulating PTEN/PI3K/AKT/mTOR signaling pathway. Life Sci. 2020, 259, 118239. [Google Scholar] [CrossRef] [PubMed]
- Aslan, C.; Maralbashi, S.; Kahroba, H.; Asadi, M.; Soltani-Zangbar, M.S.; Javadian, M.; Shanehbandi, D.; Baradaran, B.; Darabi, M.; Kazemi, T. Docosahexaenoic acid (DHA) inhibits pro-angiogenic effects of breast cancer cells via down-regulating cellular and exosomal expression of angiogenic genes and microRNAs. Life Sci. 2020, 258, 118094. [Google Scholar] [CrossRef]
- Khalili, N.; Nouri-Vaskeh, M.; Hasanpour Segherlou, Z.; Baghbanzadeh, A.; Halimi, M.; Rezaee, H.; Baradaran, B. Diagnostic, prognostic, and therapeutic significance of miR-139-5p in cancers. Life Sci. 2020, 256, 117865. [Google Scholar] [CrossRef]
- Azarbarzin, S.; Hosseinpour-Feizi, M.A.; Banan Khojasteh, S.M.; Baradaran, B.; Safaralizadeh, R. MicroRNA -383-5p restrains the proliferation and migration of breast cancer cells and promotes apoptosis via inhibition of PD-L1. Life Sci. 2021, 267, 118939. [Google Scholar] [CrossRef] [PubMed]
- Godínez-Rubí, M.; Ortuño-Sahagún, D. miR-615 Fine-Tunes Growth and Development and Has a Role in Cancer and in Neural Repair. Cells 2020, 9, 1566. [Google Scholar] [CrossRef]
- Ferrari, E.; Gandellini, P. Unveiling the ups and downs of miR-205 in physiology and cancer: Transcriptional and post-transcriptional mechanisms. Cell Death Dis. 2020, 11, 980. [Google Scholar] [CrossRef]
- Mu, Z.; Zhang, H.; Lei, P. Piceatannol inhibits pyroptosis and suppresses oxLDL-induced lipid storage in macrophages by regulating miR-200a/Nrf2/GSDMD axis. Biosci. Rep. 2020, 40. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Ye, L.; Zhang, K.; Gao, L.; Xiao, J.; Zhang, Y. Upregulation of microRNA-200a in bone marrow mesenchymal stem cells enhances the repair of spinal cord injury in rats by reducing oxidative stress and regulating Keap1/Nrf2 pathway. Artif. Organs 2020, 44, 744–752. [Google Scholar] [CrossRef]
- Zhao, L.; Qi, Y.; Xu, L.; Tao, X.; Han, X.; Yin, L.; Peng, J. MicroRNA-140-5p aggravates doxorubicin-induced cardiotoxicity by promoting myocardial oxidative stress via targeting Nrf2 and Sirt2. Redox Biol. 2018, 15, 284–296. [Google Scholar] [CrossRef]
- Jiang, Z.; Wu, J.; Ma, F.; Jiang, J.; Xu, L.; Du, L.; Huang, W.; Wang, Z.; Jia, Y.; Lu, L.; et al. MicroRNA-200a improves diabetic endothelial dysfunction by targeting KEAP1/NRF2. J. Endocrinol. 2020, 245, 129–140. [Google Scholar] [CrossRef]
- Ma, Y.; Pan, C.; Tang, X.; Zhang, M.; Shi, H.; Wang, T.; Zhang, Y. MicroRNA-200a represses myocardial infarction-related cell death and inflammation by targeting the Keap1/Nrf2 and β-catenin pathways. Hellenic. J. Cardiol. 2020. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Liu, H.; Wang, Z.; Hu, Z.; Li, L. miR-200a Attenuated Doxorubicin-Induced Cardiotoxicity through Upregulation of Nrf2 in Mice. Oxidative Med. Cell. Longev. 2019, 2019, 1512326. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Wang, W.; Niu, T.; Wang, H.; Li, B.; Shao, L.; Lai, Y.; Li, H.; Janicki, J.S.; Wang, X.L.; et al. Nrf2 deficiency exaggerates doxorubicin-induced cardiotoxicity and cardiac dysfunction. Oxidative Med. Cell. Longev. 2014, 2014, 748524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, S.S.; Vats, S.; Chia, A.Y.; Tan, T.Z.; Deng, S.; Ong, M.S.; Arfuso, F.; Yap, C.T.; Goh, B.C.; Sethi, G.; et al. Dual role of autophagy in hallmarks of cancer. Oncogene 2018, 37, 1142–1158. [Google Scholar] [CrossRef]
- Patra, S.; Mishra, S.R.; Behera, B.P.; Mahapatra, K.K.; Panigrahi, D.P.; Bhol, C.S.; Praharaj, P.P.; Sethi, G.; Patra, S.K.; Bhutia, S.K. Autophagy-modulating phytochemicals in cancer therapeutics: Current evidences and future perspectives. Semin. Cancer Biol. 2020. [Google Scholar] [CrossRef]
- Ren, H.; Shao, Y.; Wu, C.; Ma, X.; Lv, C.; Wang, Q. Metformin alleviates oxidative stress and enhances autophagy in diabetic kidney disease via AMPK/SIRT1-FoxO1 pathway. Mol. Cell. Endocrinol. 2020, 500, 110628. [Google Scholar] [CrossRef]
- Liang, D.; Zhuo, Y.; Guo, Z.; He, L.; Wang, X.; He, Y.; Li, L.; Dai, H. SIRT1/PGC-1 pathway activation triggers autophagy/mitophagy and attenuates oxidative damage in intestinal epithelial cells. Biochimie 2020, 170, 10–20. [Google Scholar] [CrossRef]
- Renu, K.; Valsala Gopalakrishnan, A. Deciphering the molecular mechanism during doxorubicin-mediated oxidative stress, apoptosis through Nrf2 and PGC-1α in a rat testicular milieu. Reprod. Biol. 2019, 19, 22–37. [Google Scholar] [CrossRef]
- Zhou, B.; Liu, J.; Kang, R.; Klionsky, D.J.; Kroemer, G.; Tang, D. Ferroptosis is a type of autophagy-dependent cell death. Semin. Cancer Biol. 2020, 66, 89–100. [Google Scholar] [CrossRef]
- Liu, Q.; Zhao, M.; Chen, W.; Xu, K.; Huang, F.; Qu, J.; Xu, Z.; Wang, X.; Wang, Y.; Zhu, Y.; et al. Mainstream cigarette smoke induces autophagy and promotes apoptosis in oral mucosal epithelial cells. Arch. Oral Biol. 2020, 111, 104646. [Google Scholar] [CrossRef]
- Deng, S.; Shanmugam, M.K.; Kumar, A.P.; Yap, C.T.; Sethi, G.; Bishayee, A. Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer 2019, 125, 1228–1246. [Google Scholar] [CrossRef] [PubMed]
- Chihara, D.; Westin, J.R.; Oki, Y.; Ahmed, M.A.; Do, B.; Fayad, L.E.; Hagemeister, F.B.; Romaguera, J.E.; Fanale, M.A.; Lee, H.J.; et al. Management strategies and outcomes for very elderly patients with diffuse large B-cell lymphoma. Cancer 2016, 122, 3145–3151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGuckin, M.M.; Giesy, S.L.; Davis, A.N.; Abyeta, M.A.; Horst, E.A.; Saed Samii, S.; Zang, Y.; Butler, W.R.; Baumgard, L.H.; McFadden, J.W.; et al. The acute phase protein orosomucoid 1 is upregulated in early lactation but does not trigger appetite-suppressing STAT3 signaling via the leptin receptor. J. Dairy Sci. 2020, 103, 4765–4776. [Google Scholar] [CrossRef] [PubMed]
- Fandiño-Vaquero, R.; Fernández-Trasancos, A.; Alvarez, E.; Ahmad, S.; Batista-Oliveira, A.L.; Adrio, B.; Fernández, A.L.; González-Juanatey, J.R.; Eiras, S. Orosomucoid secretion levels by epicardial adipose tissue as possible indicator of endothelial dysfunction in diabetes mellitus or inflammation in coronary artery disease. Atherosclerosis 2014, 235, 281–288. [Google Scholar] [CrossRef]
- Ren, F.; Chen, Y.; Wang, Y.; Yan, Y.; Zhao, J.; Ding, M.; Zhang, J.; Jiang, Y.; Zhai, Y.; Duan, Z. Comparative serum proteomic analysis of patients with acute-on-chronic liver failure: Alpha-1-acid glycoprotein maybe a candidate marker for prognosis of hepatitis B virus infection. J. Viral. Hepat. 2010, 17, 816–824. [Google Scholar] [CrossRef]
- Zhu, H.Z.; Zhou, W.J.; Wan, Y.F.; Ge, K.; Lu, J.; Jia, C.K. Downregulation of orosomucoid 2 acts as a prognostic factor associated with cancer-promoting pathways in liver cancer. World J. Gastroenterol. 2020, 26, 804–817. [Google Scholar] [CrossRef]
- Cheng, X.; Liu, D.; Xing, R.; Song, H.; Tian, X.; Yan, C.; Han, Y. Orosomucoid 1 Attenuates Doxorubicin-Induced Oxidative Stress and Apoptosis in Cardiomyocytes via Nrf2 Signaling. Biomed. Res. Int. 2020, 2020, 5923572. [Google Scholar] [CrossRef]
- Tomlinson, L.; Lu, Z.Q.; Bentley, R.A.; Colley, H.E.; Murdoch, C.; Webb, S.D.; Cross, M.J.; Copple, I.M.; Sharma, P. Attenuation of doxorubicin-induced cardiotoxicity in a human in vitro cardiac model by the induction of the NRF-2 pathway. Biomed. Pharmacother. 2019, 112, 108637. [Google Scholar] [CrossRef]
- Wang, H.; Brown, J.; Martin, M. Glycogen synthase kinase 3: A point of convergence for the host inflammatory response. Cytokine 2011, 53, 130–140. [Google Scholar] [CrossRef] [Green Version]
- Cohen, P.; Frame, S. The renaissance of GSK3. Nat. Rev. Mol. Cell. Biol. 2001, 2, 769–776. [Google Scholar] [CrossRef]
- Bitar, M.S.; Al-Mulla, F. A defect in Nrf2 signaling constitutes a mechanism for cellular stress hypersensitivity in a genetic rat model of type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E1119–E1129. [Google Scholar] [CrossRef]
- Tomobe, K.; Shinozuka, T.; Kuroiwa, M.; Nomura, Y. Age-related changes of Nrf2 and phosphorylated GSK-3β in a mouse model of accelerated aging (SAMP8). Arch. Gerontol. Geriatr. 2012, 54, e1–e7. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Bao, H.; Ge, Y.; Tang, W.; Cheng, D.; Luo, K.; Gong, G.; Gong, R. Therapeutic targeting of GSK3β enhances the Nrf2 antioxidant response and confers hepatic cytoprotection in hepatitis C. Gut 2015, 64, 168–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rojo, A.I.; Rada, P.; Egea, J.; Rosa, A.O.; López, M.G.; Cuadrado, A. Functional interference between glycogen synthase kinase-3 beta and the transcription factor Nrf2 in protection against kainate-induced hippocampal cell death. Mol. Cell. Neurosci. 2008, 39, 125–132. [Google Scholar] [CrossRef] [PubMed]
- Espada, S.; Rojo, A.I.; Salinas, M.; Cuadrado, A. The muscarinic M1 receptor activates Nrf2 through a signaling cascade that involves protein kinase C and inhibition of GSK-3beta: Connecting neurotransmission with neuroprotection. J. Neurochem. 2009, 110, 1107–1119. [Google Scholar] [CrossRef] [PubMed]
- Correa, F.; Mallard, C.; Nilsson, M.; Sandberg, M. Activated microglia decrease histone acetylation and Nrf2-inducible anti-oxidant defence in astrocytes: Restoring effects of inhibitors of HDACs, p38 MAPK and GSK3β. Neurobiol. Dis. 2011, 44, 142–151. [Google Scholar] [CrossRef] [Green Version]
- Zhou, S.; Wang, P.; Qiao, Y.; Ge, Y.; Wang, Y.; Quan, S.; Yao, R.; Zhuang, S.; Wang, L.J.; Du, Y.; et al. Genetic and Pharmacologic Targeting of Glycogen Synthase Kinase 3β Reinforces the Nrf2 Antioxidant Defense against Podocytopathy. J. Am. Soc. Nephrol. 2016, 27, 2289–2308. [Google Scholar] [CrossRef] [Green Version]
- Nordgren, K.K.S.; Wallace, K.B. Disruption of the Keap1/Nrf2-Antioxidant Response System After Chronic Doxorubicin Exposure In Vivo. Cardiovasc. Toxicol. 2020, 20, 557–570. [Google Scholar] [CrossRef]
- Wu, W.Y.; Cui, Y.K.; Hong, Y.X.; Li, Y.D.; Wu, Y.; Li, G.; Li, G.R.; Wang, Y. Doxorubicin cardiomyopathy is ameliorated by acacetin via Sirt1-mediated activation of AMPK/Nrf2 signal molecules. J. Cell. Mol. Med. 2020, 24, 12141–12153. [Google Scholar] [CrossRef]
- Chen, M.; Samuel, V.P.; Wu, Y.; Dang, M.; Lin, Y.; Sriramaneni, R.; Sah, S.K.; Chinnaboina, G.K.; Zhang, G. Nrf2/HO-1 Mediated Protective Activity of Genistein Against Doxorubicin-Induced Cardiac Toxicity. J. Environ. Pathol. Toxicol. Oncol. 2019, 38, 143–152. [Google Scholar] [CrossRef]
- Singh, A.; Venkannagari, S.; Oh, K.H.; Zhang, Y.Q.; Rohde, J.M.; Liu, L.; Nimmagadda, S.; Sudini, K.; Brimacombe, K.R.; Gajghate, S.; et al. Small Molecule Inhibitor of NRF2 Selectively Intervenes Therapeutic Resistance in KEAP1-Deficient NSCLC Tumors. ACS Chem. Biol. 2016, 11, 3214–3225. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Kong, L.; Yang, Q.; Duan, A.; Ju, X.; Cai, B.; Chen, L.; An, T.; Li, Y. Parthenolide inhibits ubiquitin-specific peptidase 7 (USP7), Wnt signaling, and colorectal cancer cell growth. J. Biol. Chem. 2020, 295, 3576–3589. [Google Scholar] [CrossRef] [PubMed]
- Sztiller-Sikorska, M.; Czyz, M. Parthenolide as Cooperating Agent for Anti-Cancer Treatment of Various Malignancies. Pharmaceuticals 2020, 13, 194. [Google Scholar] [CrossRef]
- Jin, X.; Yang, Q.; Cai, N.; Zhang, Z. A cocktail of betulinic acid, parthenolide, honokiol and ginsenoside Rh2 in liposome systems for lung cancer treatment. Nanomedicine 2020, 15, 41–54. [Google Scholar] [CrossRef] [PubMed]
- Carlisi, D.; De Blasio, A.; Drago-Ferrante, R.; Di Fiore, R.; Buttitta, G.; Morreale, M.; Scerri, C.; Vento, R.; Tesoriere, G. Parthenolide prevents resistance of MDA-MB231 cells to doxorubicin and mitoxantrone: The role of Nrf2. Cell Death Discov. 2017, 3, 17078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, B.; Huang, J.; Xiang, T.; Yin, X.; Luo, X.; Huang, J.; Luo, F.; Li, H.; Li, H.; Ren, G. Chrysin inhibits metastatic potential of human triple-negative breast cancer cells by modulating matrix metalloproteinase-10, epithelial to mesenchymal transition, and PI3K/Akt signaling pathway. J. Appl. Toxicol. 2014, 34, 105–112. [Google Scholar] [CrossRef] [PubMed]
- Woo, K.J.; Jeong, Y.J.; Park, J.W.; Kwon, T.K. Chrysin-induced apoptosis is mediated through caspase activation and Akt inactivation in U937 leukemia cells. Biochem. Biophys. Res. Commun. 2004, 325, 1215–1222. [Google Scholar] [CrossRef]
- Wang, J.; Wang, H.; Sun, K.; Wang, X.; Pan, H.; Zhu, J.; Ji, X.; Li, X. Chrysin suppresses proliferation, migration, and invasion in glioblastoma cell lines via mediating the ERK/Nrf2 signaling pathway. Drug Des. Dev. Ther. 2018, 12, 721–733. [Google Scholar] [CrossRef] [Green Version]
- Xia, Y.; Lian, S.; Khoi, P.N.; Yoon, H.J.; Joo, Y.E.; Chay, K.O.; Kim, K.K.; Do Jung, Y. Chrysin inhibits tumor promoter-induced MMP-9 expression by blocking AP-1 via suppression of ERK and JNK pathways in gastric cancer cells. PLoS ONE 2015, 10, e0124007. [Google Scholar] [CrossRef] [Green Version]
- Gao, A.M.; Ke, Z.P.; Shi, F.; Sun, G.C.; Chen, H. Chrysin enhances sensitivity of BEL-7402/ADM cells to doxorubicin by suppressing PI3K/Akt/Nrf2 and ERK/Nrf2 pathway. Chem. Biol. Interact. 2013, 206, 100–108. [Google Scholar] [CrossRef]
- Gao, H.L.; Xia, Y.Z.; Zhang, Y.L.; Yang, L.; Kong, L.Y. Vielanin P enhances the cytotoxicity of doxorubicin via the inhibition of PI3K/Nrf2-stimulated MRP1 expression in MCF-7 and K562 DOX-resistant cell lines. Phytomedicine 2019, 58, 152885. [Google Scholar] [CrossRef] [PubMed]
- Qi, Y.; Ding, Z.; Yao, Y.; Ren, F.; Yin, M.; Yang, S.; Chen, A. Apigenin induces apoptosis and counteracts cisplatin-induced chemoresistance via Mcl-1 in ovarian cancer cells. Exp. Ther. Med. 2020, 20, 1329–1336. [Google Scholar] [CrossRef]
- Korga-Plewko, A.; Michalczyk, M.; Adamczuk, G.; Humeniuk, E.; Ostrowska-Lesko, M.; Jozefczyk, A.; Iwan, M.; Wojcik, M.; Dudka, J. Apigenin and Hesperidin Downregulate DNA Repair Genes in MCF-7 Breast Cancer Cells and Augment Doxorubicin Toxicity. Molecules 2020, 25, 4421. [Google Scholar] [CrossRef] [PubMed]
- Şirin, N.; Elmas, L.; Seçme, M.; Dodurga, Y. Investigation of possible effects of apigenin, sorafenib and combined applications on apoptosis and cell cycle in hepatocellular cancer cells. Gene 2020, 737, 144428. [Google Scholar] [CrossRef]
- Gao, A.M.; Zhang, X.Y.; Ke, Z.P. Apigenin sensitizes BEL-7402/ADM cells to doxorubicin through inhibiting miR-101/Nrf2 pathway. Oncotarget 2017, 8, 82085–82091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohan, C.D.; Srinivasa, V.; Rangappa, S.; Mervin, L.; Mohan, S.; Paricharak, S.; Baday, S.; Li, F.; Shanmugam, M.K.; Chinnathambi, A.; et al. Trisubstituted-Imidazoles Induce Apoptosis in Human Breast Cancer Cells by Targeting the Oncogenic PI3K/Akt/mTOR Signaling Pathway. PLoS ONE 2016, 11, e0153155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ong, P.S.; Wang, L.Z.; Dai, X.; Tseng, S.H.; Loo, S.J.; Sethi, G. Judicious Toggling of mTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front. Pharmacol. 2016, 7, 395. [Google Scholar] [CrossRef] [PubMed]
- Gao, A.M.; Ke, Z.P.; Wang, J.N.; Yang, J.Y.; Chen, S.Y.; Chen, H. Apigenin sensitizes doxorubicin-resistant hepatocellular carcinoma BEL-7402/ADM cells to doxorubicin via inhibiting PI3K/Akt/Nrf2 pathway. Carcinogenesis 2013, 34, 1806–1814. [Google Scholar] [CrossRef]
- Wei, Y.; Liu, D.; Jin, X.; Gao, P.; Wang, Q.; Zhang, J.; Zhang, N. PA-MSHA inhibits the growth of doxorubicin-resistant MCF-7/ADR human breast cancer cells by downregulating Nrf2/p62. Cancer Med. 2016, 5, 3520–3531. [Google Scholar] [CrossRef]
- Kim, D.; Choi, B.H.; Ryoo, I.G.; Kwak, M.K. High NRF2 level mediates cancer stem cell-like properties of aldehyde dehydrogenase (ALDH)-high ovarian cancer cells: Inhibitory role of all-trans retinoic acid in ALDH/NRF2 signaling. Cell Death Dis. 2018, 9, 896. [Google Scholar] [CrossRef]
- Yang, H.Y.; Zhao, L.; Yang, Z.; Zhao, Q.; Qiang, L.; Ha, J.; Li, Z.Y.; You, Q.D.; Guo, Q.L. Oroxylin a reverses multi-drug resistance of human hepatoma BEL7402/5¯FU cells via downregulation of P¯glycoprotein expression by inhibiting NF-κB signaling pathway. Mol. Carcinog. 2012, 51, 185–195. [Google Scholar] [CrossRef] [PubMed]
- Koh, H.; Sun, H.N.; Xing, Z.; Liu, R.; Chandimali, N.; Kwon, T.; Lee, D.S. Wogonin Influences Osteosarcoma Stem Cell Stemness Through ROS-dependent Signaling. In Vivo 2020, 34, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
- Yang, D.; Guo, Q.; Liang, Y.; Zhao, Y.; Tian, X.; Ye, Y.; Tian, J.; Wu, T.; Lu, N. Wogonin induces cellular senescence in breast cancer via suppressing TXNRD2 expression. Arch. Toxicol. 2020, 94, 3433–3447. [Google Scholar] [CrossRef] [PubMed]
- Zhong, Y.; Zhang, F.; Sun, Z.; Zhou, W.; Li, Z.Y.; You, Q.D.; Guo, Q.L.; Hu, R. Drug resistance associates with activation of Nrf2 in MCF-7/DOX cells, and wogonin reverses it by down-regulating Nrf2-mediated cellular defense response. Mol. Carcinog. 2013, 52, 824–834. [Google Scholar] [CrossRef]
- Ohnuma, T.; Matsumoto, T.; Itoi, A.; Kawana, A.; Nishiyama, T.; Ogura, K.; Hiratsuka, A. Enhanced sensitivity of A549 cells to the cytotoxic action of anticancer drugs via suppression of Nrf2 by procyanidins from Cinnamomi Cortex extract. Biochem. Biophys. Res. Commun. 2011, 413, 623–629. [Google Scholar] [CrossRef]
- Selvi, R.B.; Swaminathan, A.; Chatterjee, S.; Shanmugam, M.K.; Li, F.; Ramakrishnan, G.B.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; et al. Inhibition of p300 lysine acetyltransferase activity by luteolin reduces tumor growth in head and neck squamous cell carcinoma (HNSCC) xenograft mouse model. Oncotarget 2015, 6, 43806–43818. [Google Scholar] [CrossRef] [Green Version]
- Masraksa, W.; Tanasawet, S.; Hutamekalin, P.; Wongtawatchai, T.; Sukketsiri, W. Luteolin attenuates migration and invasion of lung cancer cells via suppressing focal adhesion kinase and non-receptor tyrosine kinase signaling pathway. Nutr. Res. Pract. 2020, 14, 127–133. [Google Scholar] [CrossRef]
- Cao, D.; Zhu, G.Y.; Lu, Y.; Yang, A.; Chen, D.; Huang, H.J.; Peng, S.X.; Chen, L.W.; Li, Y.W. Luteolin suppresses epithelial-mesenchymal transition and migration of triple-negative breast cancer cells by inhibiting YAP/TAZ activity. Biomed. Pharmacother. 2020, 129, 110462. [Google Scholar] [CrossRef] [PubMed]
- Monti, E.; Marras, E.; Prini, P.; Gariboldi, M.B. Luteolin impairs hypoxia adaptation and progression in human breast and colon cancer cells. Eur. J. Pharmacol. 2020, 881, 173210. [Google Scholar] [CrossRef]
- Tang, X.; Wang, H.; Fan, L.; Wu, X.; Xin, A.; Ren, H.; Wang, X.J. Luteolin inhibits Nrf2 leading to negative regulation of the Nrf2/ARE pathway and sensitization of human lung carcinoma A549 cells to therapeutic drugs. Free Radic. Biol. Med. 2011, 50, 1599–1609. [Google Scholar] [CrossRef]
- Sabzichi, M.; Hamishehkar, H.; Ramezani, F.; Sharifi, S.; Tabasinezhad, M.; Pirouzpanah, M.; Ghanbari, P.; Samadi, N. Luteolin-loaded phytosomes sensitize human breast carcinoma MDA-MB 231 cells to doxorubicin by suppressing Nrf2 mediated signalling. Asian Pac. J. Cancer Prev. 2014, 15, 5311–5316. [Google Scholar] [CrossRef] [Green Version]
- Chen, F.; Liu, Y.; Wang, S.; Guo, X.; Shi, P.; Wang, W.; Xu, B. Triptolide, a Chinese herbal extract, enhances drug sensitivity of resistant myeloid leukemia cell lines through downregulation of HIF-1α and Nrf2. Pharmacogenomics 2013, 14, 1305–1317. [Google Scholar] [CrossRef] [PubMed]
- Shanmugam, M.K.; Warrier, S.; Kumar, A.P.; Sethi, G.; Arfuso, F. Potential Role of Natural Compounds as Anti-Angiogenic Agents in Cancer. Curr. Vasc. Pharmacol. 2017, 15, 503–519. [Google Scholar] [CrossRef]
- Kashyap, D.; Tuli, H.S.; Yerer, M.B.; Sharma, A.; Sak, K.; Srivastava, S.; Pandey, A.; Garg, V.K.; Sethi, G.; Bishayee, A. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef]
- Sadeghi, M.R.; Jeddi, F.; Soozangar, N.; Somi, M.H.; Shirmohamadi, M.; Khaze, V.; Samadi, N. Nrf2/P-glycoprotein axis is associated with clinicopathological characteristics in colorectal cancer. Biomed. Pharmacother. 2018, 104, 458–464. [Google Scholar] [CrossRef] [PubMed]
- Xia, X.; Wang, Q.; Ye, T.; Liu, Y.; Liu, D.; Song, S.; Zheng, C. NRF2/ABCB1-mediated efflux and PARP1-mediated dampening of DNA damage contribute to doxorubicin resistance in chronic hypoxic HepG2 cells. Fundam. Clin. Pharmacol. 2020, 34, 41–50. [Google Scholar] [CrossRef] [Green Version]
- Ryoo, I.G.; Kim, G.; Choi, B.H.; Lee, S.H.; Kwak, M.K. Involvement of NRF2 Signaling in Doxorubicin Resistance of Cancer Stem Cell-Enriched Colonospheres. Biomol. Ther. 2016, 24, 482–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.; Zhang, Q.; Liu, Y.; Zhang, X.; Shan, W.; Ye, S.; Zhou, X.; Ge, Y.; Wang, X.; Ren, L. Nanoparticle-based co-delivery of siRNA and paclitaxel for dual-targeting of glioblastoma. Nanomedicine 2020, 15, 1391–1409. [Google Scholar] [CrossRef] [PubMed]
- Wu, B.; Yuan, Y.; Han, X.; Wang, Q.; Shang, H.; Liang, X.; Jing, H.; Cheng, W. Structure of LINC00511-siRNA-conjugated nanobubbles and improvement of cisplatin sensitivity on triple negative breast cancer. FASEB J. 2020, 34, 9713–9726. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Wu, Z.; Ding, W.; Xiao, C.; Zhang, Y.; Gao, S.; Gao, Y.; Cai, W. SREBP1 siRNA enhance the docetaxel effect based on a bone-cancer dual-targeting biomimetic nanosystem against bone metastatic castration-resistant prostate cancer. Theranostics 2020, 10, 1619–1632. [Google Scholar] [CrossRef] [PubMed]
- Li, P.C.; Tu, M.J.; Ho, P.Y.; Jilek, J.L.; Duan, Z.; Zhang, Q.Y.; Yu, A.X.; Yu, A.M. Bioengineered NRF2-siRNA Is Effective to Interfere with NRF2 Pathways and Improve Chemosensitivity of Human Cancer Cells. Drug Metab. Dispos. 2018, 46, 2–10. [Google Scholar] [CrossRef] [Green Version]
- Shao, J.; Glorieux, C.; Liao, J.; Chen, P.; Lu, W.; Liang, Z.; Wen, S.; Hu, Y.; Huang, P. Impact of Nrf2 on tumour growth and drug sensitivity in oncogenic K-ras-transformed cells in vitro and in vivo. Free Radic. Res. 2018, 52, 661–671. [Google Scholar] [CrossRef]
- Yan, Y.; Zuo, X.; Wei, D. Concise Review: Emerging Role of CD44 in Cancer Stem Cells: A Promising Biomarker and Therapeutic Target. Stem. Cells Transl. Med. 2015, 4, 1033–1043. [Google Scholar] [CrossRef]
- Zöller, M. CD44: Can a cancer-initiating cell profit from an abundantly expressed molecule? Nat. Rev. Cancer 2011, 11, 254–267. [Google Scholar] [CrossRef] [PubMed]
- Ryoo, I.G.; Choi, B.H.; Ku, S.K.; Kwak, M.K. High CD44 expression mediates p62-associated NFE2L2/NRF2 activation in breast cancer stem cell-like cells: Implications for cancer stem cell resistance. Redox Biol. 2018, 17, 246–258. [Google Scholar] [CrossRef] [PubMed]
- Probst, B.L.; McCauley, L.; Trevino, I.; Wigley, W.C.; Ferguson, D.A. Cancer Cell Growth Is Differentially Affected by Constitutive Activation of NRF2 by KEAP1 Deletion and Pharmacological Activation of NRF2 by the Synthetic Triterpenoid, RTA 405. PLoS ONE 2015, 10, e0135257. [Google Scholar] [CrossRef] [Green Version]
- Goto, S.; Kawabata, T.; Li, T.S. Enhanced Expression of ABCB1 and Nrf2 in CD133-Positive Cancer Stem Cells Associates with Doxorubicin Resistance. Stem. Cells Int. 2020, 2020, 8868849. [Google Scholar] [CrossRef]
- Song, Z.; Liu, Z.; Sun, J.; Sun, F.L.; Li, C.Z.; Sun, J.Z.; Xu, L.Y. The MRTF-A/B function as oncogenes in pancreatic cancer. Oncol. Rep. 2016, 35, 127–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xing, W.J.; Liao, X.H.; Wang, N.; Zhao, D.W.; Zheng, L.; Zheng, D.L.; Dong, J.; Zhang, T.C. MRTF-A and STAT3 promote MDA-MB-231 cell migration via hypermethylating BRSM1. IUBMB Life 2015, 67, 202–217. [Google Scholar] [CrossRef]
- Xu, Y.; Luo, Y.; Wang, Z.Y.; Li, X.; Zheng, P.; Zhang, T.C. MRTF-A can activate Nrf2 to increase the resistance to doxorubicin. Oncotarget 2017, 8, 8436–8446. [Google Scholar] [CrossRef] [Green Version]
- Brindley, D.N.; Lin, F.-T.; Tigyi, G.J. Role of the autotaxin–lysophosphatidate axis in cancer resistance to chemotherapy and radiotherapy. Biochim. Biophys. Acta BBA Mol. Cell Biol. Lipids 2013, 1831, 74–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kehlen, A.; Lauterbach, R.; Santos, A.; Thiele, K.; Kabisch, U.; Weber, E.; Riemann, D.; Langner, J. IL-1β-and IL-4-induced down-regulation of autotaxin mRNA and PC-1 in fibroblast-like synoviocytes of patients with rheumatoid arthritis (RA). Clin. Exp. Immunol. 2001, 123, 147–154. [Google Scholar] [CrossRef] [PubMed]
- Nikitopoulou, I.; Oikonomou, N.; Karouzakis, E.; Sevastou, I.; Nikolaidou-Katsaridou, N.; Zhao, Z.; Mersinias, V.; Armaka, M.; Xu, Y.; Masu, M. Autotaxin expression from synovial fibroblasts is essential for the pathogenesis of modeled arthritis. J. Exp. Med. 2012, 209, 925–933. [Google Scholar] [CrossRef] [Green Version]
- Tager, A.M.; LaCamera, P.; Shea, B.S.; Campanella, G.S.; Selman, M.; Zhao, Z.; Polosukhin, V.; Wain, J.; Karimi-Shah, B.A.; Kim, N.D. The lysophosphatidic acid receptor LPA 1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascular leak. Nat. Med. 2008, 14, 45–54. [Google Scholar] [CrossRef] [PubMed]
- Hozumi, H.; Hokari, R.; Kurihara, C.; Narimatsu, K.; Sato, H.; Sato, S.; Ueda, T.; Higashiyama, M.; Okada, Y.; Watanabe, C. Involvement of autotaxin/lysophospholipase D expression in intestinal vessels in aggravation of intestinal damage through lymphocyte migration. Lab. Investig. 2013, 93, 508–519. [Google Scholar] [CrossRef] [PubMed]
- Euer, N.; Schwirzke, M.; Evtimova, V.; Burtscher, H.; Jarsch, M.; Tarin, D.; Weidle, U.H. Identification of genes associated with metastasis of mammary carcinoma in metastatic versus non-metastatic cell lines. Anticancer Res. 2002, 22, 733–740. [Google Scholar] [PubMed]
- Lin, S.; Wang, D.; Iyer, S.; Ghaleb, A.M.; Shim, H.; Yang, V.W.; Chun, J.; Yun, C.C. The absence of LPA2 attenuates tumor formation in an experimental model of colitis-associated cancer. Gastroenterology 2009, 136, 1711–1720. [Google Scholar] [CrossRef] [Green Version]
- Cooper, A.B.; Wu, J.; Lu, D.; Maluccio, M.A. Is autotaxin (ENPP2) the link between hepatitis C and hepatocellular cancer? J. Gastrointest. Surg. 2007, 11, 1628–1634; discussion 1634–1625. [Google Scholar] [CrossRef] [PubMed]
- Venkatraman, G.; Benesch, M.G.; Tang, X.; Dewald, J.; McMullen, T.P.; Brindley, D.N. Lysophosphatidate signaling stabilizes Nrf2 and increases the expression of genes involved in drug resistance and oxidative stress responses: Implications for cancer treatment. FASEB J. 2015, 29, 772–785. [Google Scholar] [CrossRef] [Green Version]
- Loignon, M.; Miao, W.; Hu, L.; Bier, A.; Bismar, T.A.; Scrivens, P.J.; Mann, K.; Basik, M.; Bouchard, A.; Fiset, P.O.; et al. Cul3 overexpression depletes Nrf2 in breast cancer and is associated with sensitivity to carcinogens, to oxidative stress, and to chemotherapy. Mol. Cancer Ther. 2009, 8, 2432–2440. [Google Scholar] [CrossRef] [Green Version]
- Kang, H.J.; Yi, Y.W.; Hong, Y.B.; Kim, H.J.; Jang, Y.J.; Seong, Y.S.; Bae, I. HER2 confers drug resistance of human breast cancer cells through activation of NRF2 by direct interaction. Sci. Rep. 2014, 4, 7201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manandhar, S.; Lee, S.; Kwak, M.K. Effect of stable inhibition of NRF2 on doxorubicin sensitivity in human ovarian carcinoma OV90 cells. Arch. Pharm. Res. 2010, 33, 717–726. [Google Scholar] [CrossRef]
- Shim, G.S.; Manandhar, S.; Shin, D.H.; Kim, T.H.; Kwak, M.K. Acquisition of doxorubicin resistance in ovarian carcinoma cells accompanies activation of the NRF2 pathway. Free Radic. Biol. Med. 2009, 47, 1619–1631. [Google Scholar] [CrossRef] [PubMed]
- Choi, B.H.; Ryu, D.Y.; Ryoo, I.G.; Kwak, M.K. NFE2L2/NRF2 silencing-inducible miR-206 targets c-MET/EGFR and suppresses BCRP/ABCG2 in cancer cells. Oncotarget 2017, 8, 107188–107205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Toxicity | Signaling Network | Compound | Nrf2 Expression | Outcomes | Refs |
---|---|---|---|---|---|
Cardiotoxicity | MiRNA-140-5p/Nrf2 | – | Down-regulation | Deteriorating DOX-mediated cardiotoxicity Reducing expressions of NQO1 and HO-1 Enhancing oxidative stress level | [140] |
Cardiotoxicity Hepatotoxicity Renotoxicity | – | Asiatic acid | Up-regulation | Reducing necrosis, congestion and hyaline degeneration in heart Decreasing leukocyte inflammation, necrosis, apoptosis and fatty change in liver Decreasing necrosis and inflammation in kidney Mediating these protective effects via Nrf2 induction | [98] |
Cardiotoxicity | Nrf2/HO-1 Nrf2/NQO1 Nrf2/GCL | Pristimerin | Up-regulation | Increasing expressions of Nrf2 and its downstream targets HO-1, NQO1 and GCL Reducing oxidative stress and fibrosis | [91] |
Cardiotoxicity | Nrf2/HO-1 Nrf2/NQO1 | Tert-butylhydroquinone | Up-regulation | Ameliorating cardiotoxicity via induction of Nrf2 and its downstream targets | [113] |
Cardiotoxicity | Nrf2/HO-1 | b-LAPachone | Up-regulation | Triggering nuclear translocation of Nrf2 Enhancing expressions of HO-1 and antioxidant enzymes such as SOD, CAT and GPx | [101] |
Cardiotoxicity | Nrf2/HO-1 Nrf2/NQO1 | Cardamonin | Up-regulation | Protecting cells against inflammation and oxidative stress Reducing oxidative stress, apoptosis, and inflammation Inducing Nrf2 signaling and its downstream targets HO-1 and NQO1 | [105] |
Cardiotoxicity | Nrf2/HO-1 | Curdione | Up-regulation | Alleviating oxidative stress Preventing ROS overgeneration and mediating mitochondrial dysfunction Triggering Nrf2/HO-1 axis as an antioxidant axis | [102] |
Cardiotoxicity | MAPK/Nrf2/ARE | Chitosan oligosaccharide | Up-regulation | Decreasing oxidative stress and apoptosis Stimulating MAPK and subsequent induction of Nrf2/ARE axis Reinforcing antioxidant defense system | [112] |
Cardiotoxicity | MiRNA-200a/Nrf2 | – | Up-regulation | Improving cardiomyocyte contractile function Reducing levels of cardiac troponin I Ameliorating oxidative stress, inflammation and apoptosis Inducing Nrf2 signaling | [143] |
Cardiotoxicity | Nrf2/ARE | 3,3′-diindolylmethane | Up-regulation | Suppressing apoptosis Improving histopathological profile Enhancing expressions of HO-1, NQO1 and GST Reducing Bax and caspase-3 expression | [114] |
Cardiotoxicity | Sirt1/AMPK/Nrf2 | Acacetin | Up-regulation | Alleviation of cardiomyopathy Enhancing cell viability Preventing ROS overgeneration Activation of Sirt1/AMPK to induce Nrf2 signaling Triggering cell defense system | [170] |
Cardiotoxicity | Nrf2/HO-1 | Genistein | Up-regulation | Inducing Nrf2/HO-1 axis Reducing ROS levels by its scavenging feature Reducing lipid peroxidation and DNA damage | [171] |
Cardiotoxicity | Nrf2/LC-3II/autophagy | – | Down-regulation | Reducing oxidative stress Activating autophagy as a protective mechanism via LC-3II up-regulation Nrf2 inhibition aggravates DOX-mediated cardiotoxicity via impairing autophagy and enhancing oxidative stress | [144] |
Cardiotoxicity | – | p-coumaric acid | Up-regulation | Enhancing cell survival Inhibiting apoptosis and oxidative stress Providing nuclear translocation of Nrf2 | [118] |
Cardiotoxicity | Nrf2/NQO1 | Tanshinone IIA | Up-regulation | Enhancing cell viability and morphological profile Reducing oxidative parameters Up-regulation of NQO1 | [124] |
Cardiotoxicity | ORM1/Nrf2 | – | Up-regulation | ORM1 is correlated with a decrease in oxidative stress and apoptosis Up-regulation of Nrf2 and its downstream target HO-1 | [158] |
Testicular toxicity | – | – | Down-regulation | Inducing apoptosis and oxidative stress in testis Reducing Nrf2 expression | [149] |
Nephrotoxicity | – | Thymoquinone | Up-regulation | Reducing malondialdehyde and lipid peroxidation levels Enhancing SOD and GST levels Preventing necrosis and oxidative stressActivation of Nrf2 and improving antioxidant defense system | [119] |
Cancer Type | Signaling Network | Compound/Agent | Nrf2 Expression | Remarks | Refs |
---|---|---|---|---|---|
Breast cancer | P62/Nrf2 | – | Up-regulation | Reducing oxidative stress Mediating DOX resistance Promoting colony formation and migration capacities Improving cancer stem cell features | [216] |
Breast cancer | Cul3/Nrf2 | – | Down-regulation | Association of Cul3 with Nrf2 depletion Inducing oxidative stress Increasing DOX sensitivity | [231] |
Breast cancer | PI3K/Nrf2/MRP1 | Vielanin P | Down-regulation | Inhibiting PI3K/Nrf2 axis Suppressing MRP1 expression Promoting DOX sensitivity | [182] |
Breast cancer | Nrf2/HSP70 | Parthenolide | Down-regulation | Reducing expressions of Nrf2 and HSP70 Enhancing DOX sensitivity of breast cancer cells | [176] |
Breast cancer | Nrf2/HO-1 Nrf2/MDR1 | Luteolin | Down-regulation | Enhancing number of cancer cells undergoing cell death Increasing cytotoxicity of DOX Down-regulation of Nrf2 and subsequent inhibition of its downstream targets HO-1 and MDR1 | [202] |
Breast cancer | Nrf2/HO-1 Nrf2/NQO1 | Wogonin | Down-regulation | Impairing cellular defense systemNrf2 signaling inhibition Down-regulation of HO-1 and NQO1 Increasing DOX cytotoxicity towards cancer cells | [195] |
Breast cancer | HER2/Nrf2 | – | Up-regulation | Conferring drug resistance Enhancing activities of antioxidant enzymes such as GSTA2, GSTP1 and HO-1 | [232] |
Breast cancer | Nrf2/p62 | Pseudomonas aeruginosa mannose-sensitive hemagglutinin | Down-regulation | Inhibiting Nrf2 signaling and its downstream target p62 Increasing DOX sensitivity Impairing cancer growth | [190] |
Hepatocellular carcinoma | MiRNA-101/Nrf2 | Apigenin | Down-regulation | Enhancing miRNA-101 expression Inhibiting Nrf2 signaling by binding to 3/-UTR Enhancing DOX sensitivity | [186] |
Hepatocellular carcinoma | PI3K/Akt/Nrf2 | Apigenin | Down-regulation | Reducing mRNA and protein levels of Nrf2 via PI3K/Akt inhibition Reducing cell proliferation Inducing apoptosis Promoting DOX sensitivity | [189] |
Liver cancer | Nrf2/ABCB1 | – | Up-regulation | Nrf2 overexpression occurs in hypoxic conditions Reducing apoptosis and DNA damage Inducing DOX resistance ABCB1 up-regulation | [207] |
Different cancers | MRTF-A/Nrf2 | – | Up-regulation | Reducing apoptosis Triggering DOX resistance | [221] |
Different cancers | – | – | Down-regulation | SiRNA is a powerful in Nrf2 down-regulation Inhibiting activities of ABCC3, ABCC4 and ABCG2 Enhancing DOX sensitivity | [212] |
Different cancers | PI3K/Akt/Nrf2 ERK/Nrf2 | Chrysin | Down-regulation | Suppressing PI3K/Akt/Nrf2 and ERK/Nrf2 signaling pathwaysNrf2 down-regulation and inhibiting its downstream targets HO-1 Enhancing DOX sensitivity | [181] |
Ovarian cancer | ALDH/Nrf2 | All-trans retinoic acid | Down-regulation | Promoting cancer stem features Enhancing colony formation capacity Mediating DOX resistance Suppressing ALDH/Nrf2 signaling by retinoic acid in reducing DOX resistance | [191] |
Ovarian cancer | – | – | Up-regulation | Overexpression of Nrf2 in DOX-resistant cancer cells Reducing tumor growth following Nrf2 down-regulation | [233] |
Ovarian cancer | – | – | Up-regulation | Obtaining DOX resistance via Nrf2 signaling and reducing cell death | [234] |
Ovarian cancer | Nrf2/miRNA-206/c-MET/EGFR | – | Up-regulation | Reducing miRNA-206 expression Inducing expressions of c-MET and EGFR expressions Triggering DOX resistance | [235] |
Colorectal cancer | Nrf2/P-gp | – | Up-regulation | Enhancing P-gp expressions Reducing cell death Inducing DOX resistance | [208] |
Lung cancer | – | ML385 | Down-regulation | ML385 functions as an inhibitor of Nrf2 signaling Promoting DOX sensitivity | [172] |
Myeloid leukemia | Nrf2/HO-1 Nrf2/NQO1 | Tritolide | Down-regulation | Enhancing drug sensitivity Apoptosis induction Suppressing Nrf2 and its downstream targets | [203] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mirzaei, S.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Saleki, H.; Azami, N.; Hamzehlou, S.; Farahani, M.V.; Hushmandi, K.; Ashrafizadeh, M.; et al. Nrf2 Signaling Pathway in Chemoprotection and Doxorubicin Resistance: Potential Application in Drug Discovery. Antioxidants 2021, 10, 349. https://doi.org/10.3390/antiox10030349
Mirzaei S, Zarrabi A, Hashemi F, Zabolian A, Saleki H, Azami N, Hamzehlou S, Farahani MV, Hushmandi K, Ashrafizadeh M, et al. Nrf2 Signaling Pathway in Chemoprotection and Doxorubicin Resistance: Potential Application in Drug Discovery. Antioxidants. 2021; 10(3):349. https://doi.org/10.3390/antiox10030349
Chicago/Turabian StyleMirzaei, Sepideh, Ali Zarrabi, Farid Hashemi, Amirhossein Zabolian, Hossein Saleki, Negar Azami, Soodeh Hamzehlou, Mahdi Vasheghani Farahani, Kiavash Hushmandi, Milad Ashrafizadeh, and et al. 2021. "Nrf2 Signaling Pathway in Chemoprotection and Doxorubicin Resistance: Potential Application in Drug Discovery" Antioxidants 10, no. 3: 349. https://doi.org/10.3390/antiox10030349
APA StyleMirzaei, S., Zarrabi, A., Hashemi, F., Zabolian, A., Saleki, H., Azami, N., Hamzehlou, S., Farahani, M. V., Hushmandi, K., Ashrafizadeh, M., Khan, H., & Kumar, A. P. (2021). Nrf2 Signaling Pathway in Chemoprotection and Doxorubicin Resistance: Potential Application in Drug Discovery. Antioxidants, 10(3), 349. https://doi.org/10.3390/antiox10030349