Formononetin Exerts Neuroprotection in Parkinson’s Disease via the Activation of the Nrf2 Signaling Pathway
Abstract
:1. Introduction
2. Results
2.1. Bioinformatic Target Prediction Revealed a Potential Role of FMN in Alleviating PD
2.2. FMN Demonstrated Neuroprotection Effects in C. elegans PD Models
2.2.1. FMN Reduced Dopaminergic Neuron Damage and Alleviated Dyskinesia in the C. elegans PD Model
2.2.2. FMN Had the Potential to Alleviate Oxidative Stress in the C. elegans PD Model
2.3. FMN Demonstrated Neuroprotective Effects in the SH-SY5Y Cell Model of PD
2.4. The Proposed Mechanism for the Anti-PD Effects of FMN Through the Activation of the Nrf2 Signaling Pathway
2.5. The Nrf2 Inhibitor Could Negate the Protective Effects of FMN
2.6. FMN Exhibited Neuroprotective Effects Comparable to the Antioxidant SFN in C. elegans PD Models
3. Discussion
4. Materials and Methods
4.1. Bioinformatic Analysis
4.2. C. elegans Culture and Treatment
4.3. Dopaminergic Neurodegeneration Assay in BZ555 Nematodes
4.4. Thrashing Rate Assay of Nematodes
4.5. Measurement of Reactive Oxygen Species (ROS) Levels in C. elegans
4.6. Cell Culture and Treatment
4.7. Measurement of Cell Viability
4.8. ROS Assay in Cells
4.9. Measurement of Mitochondrial Membrane Potential (MMP) in Cells
4.10. Western Blot
4.11. Statistical Analysis
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Tansey, M.G.; Wallings, R.L.; Houser, M.C.; Herrick, M.K.; Keating, C.E.; Joers, V. Inflammation and immune dysfunction in Parkinson disease. Nat. Rev. Immunol. 2022, 22, 657–673. [Google Scholar] [CrossRef] [PubMed]
- Dickson, D.W. Neuropathology of Parkinson disease. Park. Relat. Disord. 2018, 46 (Suppl. S1), S30–S33. [Google Scholar] [CrossRef] [PubMed]
- Koeglsperger, T.; Rumpf, S.L.; Schließer, P.; Struebing, F.L.; Brendel, M.; Levin, J.; Trenkwalder, C.; Höglinger, G.U.; Herms, J. Neuropathology of incidental Lewy body & prodromal Parkinson’s disease. Mol. Neurodegener. 2023, 18, 32. [Google Scholar] [PubMed]
- Morris, H.R.; Spillantini, M.G.; Sue, C.M.; Williams-Gray, C.H. The pathogenesis of Parkinson’s disease. Lancet 2024, 403, 293–304. [Google Scholar] [CrossRef] [PubMed]
- Ye, H.; Robak, L.A.; Yu, M.; Cykowski, M.; Shulman, J.M. Genetics and Pathogenesis of Parkinson’s Syndrome. Annu. Rev. Pathol. 2023, 18, 95–121. [Google Scholar] [CrossRef]
- GBD 2015 Neurological Disorders Collaborator Group. Global, regional, and national burden of neurological disorders during 1990–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet Neurol. 2015, 16, 877–897. [Google Scholar]
- Dorsey, E.R.; Bloem, B.R. The Parkinson Pandemic-A Call to Action. JAMA Neurol. 2018, 75, 9–10. [Google Scholar] [CrossRef]
- Armstrong, M.J.; Okun, M.S. Diagnosis and Treatment of Parkinson Disease: A Review. JAMA 2020, 323, 548–560. [Google Scholar] [CrossRef]
- Cacabelos, R. Parkinson’s Disease: From Pathogenesis to Pharmacogenomics. Int. J. Mol. Sci. 2017, 18, 551. [Google Scholar] [CrossRef]
- Vijiaratnam, N.; Simuni, T.; Bandmann, O.; Morris, H.R.; Foltynie, T. Progress towards therapies for disease modification in Parkinson’s disease. Lancet Neurol. 2021, 20, 559–572. [Google Scholar] [CrossRef]
- Marino, B.L.B.; de Souza, L.R.; Sousa, K.P.A.; Ferreira, J.V.; Padilha, E.C.; da Silva, C.; Taft, C.A.; Hage-Melim, L.I.S. Parkinson’s Disease: A Review from Pathophysiology to Treatment. Mini Rev. Med. Chem. 2020, 20, 754–767. [Google Scholar] [CrossRef] [PubMed]
- Raza, C.; Anjum, R.; Shakeel, N.U.A. Parkinson’s disease: Mechanisms, translational models and management strategies. Life Sci. 2019, 226, 77–90. [Google Scholar] [CrossRef] [PubMed]
- Aliya, S.; Alhammadi, M.; Park, U.; Tiwari, J.N.; Lee, J.H.; Han, Y.K.; Huh, Y.S. The potential role of formononetin in cancer treatment: An updated review. Biomed. Pharmacother. 2023, 168, 115811. [Google Scholar] [CrossRef] [PubMed]
- Machado Dutra, J.; Espitia, P.J.P.; Andrade Batista, R. Formononetin: Biological effects and uses—A review. Food Chem. 2021, 359, 129975. [Google Scholar] [CrossRef] [PubMed]
- Ong, S.K.L.; Shanmugam, M.K.; Fan, L.; Fraser, S.E.; Arfuso, F.; Ahn, K.S.; Sethi, G.; Bishayee, A. Focus on Formononetin: Anticancer Potential and Molecular Targets. Cancers 2019, 11, 611. [Google Scholar] [CrossRef]
- Ma, C.; Xia, R.; Yang, S.; Liu, L.; Zhang, J.; Feng, K.; Shang, Y.; Qu, J.; Li, L.; Chen, N.; et al. Formononetin attenuates atherosclerosis via regulating interaction between KLF4 and SRA in apoE-/- mice. Theranostics 2020, 10, 1090–1106. [Google Scholar] [CrossRef]
- Yu, L.; Zhang, Y.; Chen, Q.; He, Y.; Zhou, H.; Wan, H.; Yang, J. Formononetin protects against inflammation associated with cerebral ischemia-reperfusion injury in rats by targeting the JAK2/STAT3 signaling pathway. Biomed. Pharmacother. 2022, 149, 112836. [Google Scholar] [CrossRef]
- Tian, J.; Wang, X.Q.; Tian, Z. Focusing on Formononetin: Recent Perspectives for its Neuroprotective Potentials. Front. Pharmacol. 2022, 13, 905898. [Google Scholar] [CrossRef]
- Chalorak, P.; Sanguanphun, T.; Limboonreung, T.; Meemon, K. Neurorescue Effects of Frondoside A and Ginsenoside Rg3 in C. elegans Model of Parkinson’s Disease. Molecules 2021, 26, 4843. [Google Scholar] [CrossRef]
- Chang, C.H.; Wei, C.C.; Ho, C.T.; Liao, V.H. N-γ-(L-glutamyl)-L-selenomethionine shows neuroprotective effects against Parkinson’s disease associated with SKN-1/Nrf2 and TRXR-1 in Caenorhabditis elegans. Phytomedicine 2021, 92, 153733. [Google Scholar] [CrossRef]
- He, C.L.; Tang, Y.; Wu, J.M.; Long, T.; Yu, L.; Teng, J.F.; Qiu, W.Q.; Pan, R.; Yu, C.L.; Qin, D.L.; et al. Chlorogenic acid delays the progression of Parkinson’s disease via autophagy induction in Caenorhabditis elegans. Nutr. Neurosci. 2023, 26, 11–24. [Google Scholar] [CrossRef] [PubMed]
- Zhu, F.D.; Wang, B.D.; Qin, D.L.; Su, X.H.; Yu, L.; Wu, J.M.; Law, B.Y.; Guo, M.S.; Yu, C.L.; Zhou, X.G.; et al. Carpesii fructus extract exhibits neuroprotective effects in cellular and Caenorhabditis elegans models of Parkinson’s disease. CNS Neurosci. Ther. 2024, 30, e14515. [Google Scholar] [CrossRef] [PubMed]
- Ioghen, O.C.; Ceafalan, L.C.; Popescu, B.O. SH-SY5Y Cell Line In Vitro Models for Parkinson Disease Research-Old Practice for New Trends. J. Integr. Neurosci. 2023, 22, 20. [Google Scholar] [CrossRef] [PubMed]
- Aly, S.H.; Elissawy, A.M.; Fayez, A.M.; Eldahshan, O.A.; Elshanawany, M.A.; Singab, A.N.B. Neuroprotective effects of Sophora secundiflora, Sophora tomentosa leaves and formononetin on scopolamine-induced dementia. Nat. Prod. Res. 2021, 35, 5848–5852. [Google Scholar] [CrossRef] [PubMed]
- Singh, L.; Kaur, H.; Chandra Arya, G.; Bhatti, R. Neuroprotective potential of formononetin, a naturally occurring isoflavone phytoestrogen. Chem. Biol. Drug Des. 2024, 103, e14353. [Google Scholar] [CrossRef]
- Tian, Z.; Liu, S.B.; Wang, Y.C.; Li, X.Q.; Zheng, L.H.; Zhao, M.G. Neuroprotective effects of formononetin against NMDA-induced apoptosis in cortical neurons. Phytother. Res. 2013, 27, 1770–1775. [Google Scholar] [CrossRef]
- Bull, K.; Cook, A.; Hopper, N.A.; Harder, A.; Holden-Dye, L.; Walker, R.J. Effects of the novel anthelmintic emodepside on the locomotion, egg-laying behaviour and development of Caenorhabditis elegans. Int. J. Parasitol. 2007, 37, 627–636. [Google Scholar] [CrossRef]
- Zhang, W.; Li, W.; Li, J.; Chang, X.; Niu, S.; Wu, T.; Kong, L.; Zhang, T.; Tang, M.; Xue, Y. Neurobehavior and neuron damage following prolonged exposure of silver nanoparticles with/without polyvinylpyrrolidone coating in Caenorhabditis elegans. J. Appl. Toxicol. 2021, 41, 2055–2067. [Google Scholar] [CrossRef]
- Hemmati-Dinarvand, M.; Saedi, S.; Valilo, M.; Kalantary-Charvadeh, A.; Alizadeh Sani, M.; Kargar, R.; Safari, H.; Samadi, N. Oxidative stress and Parkinson’s disease: Conflict of oxidant-antioxidant systems. Neurosci. Lett. 2019, 709, 134296. [Google Scholar] [CrossRef]
- Subramaniam, S.R.; Chesselet, M.F. Mitochondrial dysfunction and oxidative stress in Parkinson’s disease. Prog. Neurobiol. 2013, 106–107, 17–32. [Google Scholar] [CrossRef]
- Trist, B.G.; Hare, D.J.; Double, K.L. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell 2019, 18, e13031. [Google Scholar] [CrossRef] [PubMed]
- Mendonça, M.A.A.; Ribeiro, A.R.S.; Lima, A.K.; Bezerra, G.B.; Pinheiro, M.S.; Albuquerque-Júnior, R.L.C.; Gomes, M.Z.; Padilha, F.F.; Thomazzi, S.M.; Novellino, E.; et al. Red Propolis and Its Dyslipidemic Regulator Formononetin: Evaluation of Antioxidant Activity and Gastroprotective Effects in Rat Model of Gastric Ulcer. Nutrients 2020, 12, 2951. [Google Scholar] [CrossRef] [PubMed]
- Sugimoto, M.; Ko, R.; Goshima, H.; Koike, A.; Shibano, M.; Fujimori, K. Formononetin attenuates H2O2-induced cell death through decreasing ROS level by PI3K/Akt-Nrf2-activated antioxidant gene expression and suppressing MAPK-regulated apoptosis in neuronal SH-SY5Y cells. Neurotoxicology 2021, 85, 186–200. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Sha, X.; Wu, D.; Wu, B.; Pan, X.; Pan, L.L.; Gu, Y.; Dong, X. Formononetin alleviates acute pancreatitis by reducing oxidative stress and modulating intestinal barrier. Chin. Med. 2023, 18, 78. [Google Scholar] [CrossRef]
- de Oliveira Caland, R.B.; Cadavid, C.O.M.; Carmona, L.; Peña, L.; de Paula Oliveira, R. Pasteurized Orange Juice Rich in Carotenoids Protects Caenorhabditis elegans against Oxidative Stress and β-Amyloid Toxicity through Direct and Indirect Mechanisms. Oxid. Med. Cell Longev. 2019, 2019, 5046280. [Google Scholar] [CrossRef]
- Elkhedir, A.E.; Iqbal, A.; Zogona, D.; Mohammed, H.H.; Murtaza, A.; Xu, X. Apigenin glycosides from green pepper enhance longevity and stress resistance in Caenorhabditis elegans. Nutr. Res. 2022, 102, 23–34. [Google Scholar] [CrossRef]
- Nguyen, V.T.; Park, A.R.; Duraisamy, K.; Vo, D.D.; Kim, J.C. Elucidation of the nematicidal mode of action of grammicin on Caenorhabditis elegans. Pestic. Biochem. Physiol. 2022, 188, 105244. [Google Scholar] [CrossRef]
- Amo, T.; Oji, Y.; Saiki, S.; Hattori, N. Metabolomic analysis data of MPP(+)-exposed SH-SY5Y cells using CE-TOFMS. Data Brief. 2021, 34, 106707. [Google Scholar] [CrossRef]
- Enogieru, A.B.; Haylett, W.; Hiss, D.; Ekpo, O. Inhibition of γH2AX, COX-2 and regulation of antioxidant enzymes in MPP(+)-exposed SH-SY5Y cells pre-treated with rutin. Metab. Brain Dis. 2021, 36, 2119–2130. [Google Scholar] [CrossRef]
- Eruslanov, E.; Kusmartsev, S. Identification of ROS using oxidized DCFDA and flow-cytometry. Methods Mol. Biol. 2010, 594, 57–72. [Google Scholar]
- Angelova, P.R.; Abramov, A.Y. Role of mitochondrial ROS in the brain: From physiology to neurodegeneration. FEBS Lett. 2018, 592, 692–702. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [PubMed]
- Marcondes, N.A.; Terra, S.R.; Lasta, C.S.; Hlavac, N.R.C.; Dalmolin, M.L.; Lacerda, L.A.; Faulhaber, G.A.M.; González, F.H.D. Comparison of JC-1 and MitoTracker probes for mitochondrial viability assessment in stored canine platelet concentrates: A flow cytometry study. Cytom. A 2019, 95, 214–218. [Google Scholar] [CrossRef] [PubMed]
- Perelman, A.; Wachtel, C.; Cohen, M.; Haupt, S.; Shapiro, H.; Tzur, A. JC-1: Alternative excitation wavelengths facilitate mitochondrial membrane potential cytometry. Cell Death Dis. 2012, 3, e430. [Google Scholar] [CrossRef]
- Hybertson, B.M.; Gao, B.; Bose, S.K.; McCord, J.M. Oxidative stress in health and disease: The therapeutic potential of Nrf2 activation. Mol. Asp. Med. 2011, 32, 234–246. [Google Scholar] [CrossRef]
- Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef]
- Buendia, I.; Michalska, P.; Navarro, E.; Gameiro, I.; Egea, J.; León, R. Nrf2-ARE pathway: An emerging target against oxidative stress and neuroinflammation in neurodegenerative diseases. Pharmacol. Ther. 2016, 157, 84–104. [Google Scholar] [CrossRef]
- Luo, X.; Weng, X.; Bao, X.; Bai, X.; Lv, Y.; Zhang, S.; Chen, Y.; Zhao, C.; Zeng, M.; Huang, J.; et al. A novel anti-atherosclerotic mechanism of quercetin: Competitive binding to KEAP1 via Arg483 to inhibit macrophage pyroptosis. Redox Biol. 2022, 57, 102511. [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]
- Houghton, C.A.; Fassett, R.G.; Coombes, J.S. Sulforaphane and Other Nutrigenomic Nrf2 Activators: Can the Clinician’s Expectation Be Matched by the Reality? Oxid. Med. Cell Longev. 2016, 2016, 7857186. [Google Scholar] [CrossRef]
- Russo, M.; Spagnuolo, C.; Russo, G.L.; Skalicka-Woźniak, K.; Daglia, M.; Sobarzo-Sánchez, E.; Nabavi, S.F.; Nabavi, S.M. Nrf2 targeting by sulforaphane: A potential therapy for cancer treatment. Crit. Rev. Food Sci. Nutr. 2018, 58, 1391–1405. [Google Scholar] [CrossRef] [PubMed]
- Uddin, M.S.; Mamun, A.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] [PubMed]
- Fan, M.; Li, Z.; Hu, M.; Zhao, H.; Wang, T.; Jia, Y.; Yang, R.; Wang, S.; Song, J.; Liu, Y.; et al. Formononetin attenuates Aβ(25-35)-induced adhesion molecules in HBMECs via Nrf2 activation. Brain Res. Bull. 2022, 183, 162–171. [Google Scholar] [CrossRef] [PubMed]
- Fei, H.X.; Zhang, Y.B.; Liu, T.; Zhang, X.J.; Wu, S.L. Neuroprotective effect of formononetin in ameliorating learning and memory impairment in mouse model of Alzheimer’s disease. Biosci. Biotechnol. Biochem. 2018, 82, 57–64. [Google Scholar] [CrossRef]
- Xiao, H.; Qin, X.; Wan, J.; Li, R. Pharmacological Targets and the Biological Mechanisms of Formononetin for Alzheimer’s Disease: A Network Analysis. Med. Sci. Monit. 2019, 25, 4273–4277. [Google Scholar] [CrossRef]
- Luo, J.; Cai, Y.; Wei, D.; Cao, L.; He, Q.; Wu, Y. Formononetin alleviates cerebral ischemia-reperfusion injury in rats by targeting the PARP-1/PARG/Iduna signaling pathway. Brain Res. 2024, 1829, 148845. [Google Scholar] [CrossRef]
- Wu, Q.L.; Cheng, Y.Q.; Liu, A.J.; Zhang, W.D. Formononetin recovered injured nerve functions by enhancing synaptic plasticity in ischemic stroke rats. Biochem. Biophys. Res. Commun. 2020, 525, 67–72. [Google Scholar] [CrossRef]
- Wang, X.S.; Guan, S.Y.; Liu, A.; Yue, J.; Hu, L.N.; Zhang, K.; Yang, L.K.; Lu, L.; Tian, Z.; Zhao, M.G.; et al. Anxiolytic effects of Formononetin in an inflammatory pain mouse model. Mol. Brain 2019, 12, 36. [Google Scholar] [CrossRef]
- Li, Z.; Dong, X.; Zhang, J.; Zeng, G.; Zhao, H.; Liu, Y.; Qiu, R.; Mo, L.; Ye, Y. Formononetin protects TBI rats against neurological lesions and the underlying mechanism. J. Neurol. Sci. 2014, 338, 112–117. [Google Scholar] [CrossRef]
- Li, Z.; Wang, Y.; Zeng, G.; Zheng, X.; Wang, W.; Ling, Y.; Tang, H.; Zhang, J. Increased miR-155 and heme oxygenase-1 expression is involved in the protective effects of formononetin in traumatic brain injury in rats. Am. J. Transl. Res. 2017, 9, 5653–5661. [Google Scholar]
- Li, Z.; Zeng, G.; Zheng, X.; Wang, W.; Ling, Y.; Tang, H.; Zhang, J. Neuroprotective effect of formononetin against TBI in rats via suppressing inflammatory reaction in cortical neurons. Biomed. Pharmacother. 2018, 106, 349–354. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Wang, J. Formononetin: A Pathway to Protect Neurons. Front. Integr. Neurosci. 2022, 16, 908378. [Google Scholar] [CrossRef] [PubMed]
- Tian, S.; Zhang, J.; Yuan, S.; Wang, Q.; Lv, C.; Wang, J.; Fang, J.; Fu, L.; Yang, J.; Zu, X.; et al. Exploring pharmacological active ingredients of traditional Chinese medicine by pharmacotranscriptomic map in ITCM. Brief. Bioinform. 2023, 24, bbad027. [Google Scholar] [CrossRef] [PubMed]
- Anjaneyulu, J.; Vidyashankar, R.; Godbole, A. Differential effect of Ayurvedic nootropics on C. elegans models of Parkinson’s disease. J. Ayurveda Integr. Med. 2020, 11, 440–447. [Google Scholar] [CrossRef]
- Wang, X.; Liu, Y.; Kang, N.; Xu, G. Wide identification of chemical constituents in fermented licorice and explore its efficacy of anti-neurodegeneration by combining quasi-targeted metabolomics and in-depth bioinformatics. Front. Neurosci. 2023, 17, 1156037. [Google Scholar] [CrossRef]
- Sorenson, R.C.; Bisgaier, C.L.; Aviram, M.; Hsu, C.; Billecke, S.; La Du, B.N. Human serum Paraoxonase/Arylesterase’s retained hydrophobic N-terminal leader sequence associates with HDLs by binding phospholipids: Apolipoprotein A-I stabilizes activity. Arterioscler. Thromb. Vasc. Biol. 1999, 19, 2214–2225. [Google Scholar] [CrossRef]
- Newton-Vinson, P.; Hubalek, F.; Edmondson, D.E. High-level expression of human liver monoamine oxidase B in Pichia pastoris. Protein Expr. Purif. 2000, 20, 334–345. [Google Scholar] [CrossRef]
- Okada, H.; Zhang, W.; Peterhoff, C.; Hwang, J.C.; Nixon, R.A.; Ryu, S.H.; Kim, T.W. Proteomic identification of sorting nexin 6 as a negative regulator of BACE1-mediated APP processing. FASEB J. 2010, 24, 2783–2794. [Google Scholar] [CrossRef]
- Campbell, P.; Morton, P.E.; Takeichi, T.; Salam, A.; Roberts, N.; Proudfoot, L.E.; Mellerio, J.E.; Aminu, K.; Wellington, C.; Patil, S.N.; et al. Epithelial inflammation resulting from an inherited loss-of-function mutation in EGFR. J. Investig. Dermatol. 2014, 134, 2570–2578. [Google Scholar] [CrossRef]
- Paez, J.G.; Jänne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J.; et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef]
- Mingari, M.C.; Gerosa, F.; Carra, G.; Accolla, R.S.; Moretta, A.; Zubler, R.H.; Waldmann, T.A.; Moretta, L. Human interleukin-2 promotes proliferation of activated B cells via surface receptors similar to those of activated T cells. Nature 1984, 312, 641–643. [Google Scholar] [CrossRef] [PubMed]
- Laâbi, Y.; Gras, M.P.; Carbonnel, F.; Brouet, J.C.; Berger, R.; Larsen, C.J.; Tsapis, A. A new gene, BCM, on chromosome 16 is fused to the interleukin 2 gene by a t(4;16)(q26;p13) translocation in a malignant T cell lymphoma. EMBO J. 1992, 11, 3897–3904. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; He, H.Y.; Zhang, X.J. Increased expression of intranuclear AChE involved in apoptosis of SK-N-SH cells. Neurosci. Res. 2002, 42, 261–268. [Google Scholar] [CrossRef] [PubMed]
- Choi, K.H.; Chen, C.J.; Kriegler, M.; Roninson, I.B. An altered pattern of cross-resistance in multidrug-resistant human cells results from spontaneous mutations in the mdr1 (P-glycoprotein) gene. Cell 1988, 53, 519–529. [Google Scholar] [CrossRef] [PubMed]
- Son, S.Y.; Ma, J.; Kondou, Y.; Yoshimura, M.; Yamashita, E.; Tsukihara, T. Structure of human monoamine oxidase A at 2.2-A resolution: The control of opening the entry for substrates/inhibitors. Proc. Natl. Acad. Sci. USA 2008, 105, 5739–5744. [Google Scholar] [CrossRef]
- Brunner, H.G.; Nelen, M.; Breakefield, X.O.; Ropers, H.H.; van Oost, B.A. Abnormal behavior associated with a point mutation in the structural gene for monoamine oxidase A. Science 1993, 262, 578–580. [Google Scholar] [CrossRef]
- Molli, P.R.; Singh, R.R.; Lee, S.W.; Kumar, R. MTA1-mediated transcriptional repression of BRCA1 tumor suppressor gene. Oncogene 2008, 27, 1971–1980. [Google Scholar] [CrossRef]
- Quaynor, S.D.; Stradtman, E.W., Jr.; Kim, H.G.; Shen, Y.; Chorich, L.P.; Schreihofer, D.A.; Layman, L.C. Delayed puberty and estrogen resistance in a woman with estrogen receptor α variant. N. Engl. J. Med. 2013, 369, 164–171. [Google Scholar] [CrossRef]
- Bartke, T.; Pohl, C.; Pyrowolakis, G.; Jentsch, S. Dual role of BRUCE as an antiapoptotic IAP and a chimeric E2/E3 ubiquitin ligase. Mol. Cell. 2004, 14, 801–811. [Google Scholar] [CrossRef]
- Strauss, K.M.; Martins, L.M.; Plun-Favreau, H.; Marx, F.P.; Kautzmann, S.; Berg, D.; Gasser, T.; Wszolek, Z.; Müller, T.; Bornemann, A.; et al. Loss of function mutations in the gene encoding Omi/HtrA2 in Parkinson’s disease. Hum. Mol. Genet. 2005, 14, 2099–2111. [Google Scholar] [CrossRef]
- Lai, X.; Wichers, H.J.; Soler-Lopez, M.; Dijkstra, B.W. Structure of Human Tyrosinase Related Protein 1 Reveals a Binuclear Zinc Active Site Important for Melanogenesis. Angew. Chem. Int. Ed. Engl. 2017, 56, 9812–9815. [Google Scholar] [CrossRef] [PubMed]
- Tsai, C.H.; Tsai, F.J.; Wu, J.Y.; Lin, S.P.; Chang, J.G.; Yang, C.F.; Lee, C.C. Insertion/deletion mutations of type I oculocutaneous albinism in chinese patients from Taiwan. Hum. Mutat. 1999, 14, 542. [Google Scholar] [CrossRef]
- Tan, T.H.; Edgerton, S.A.; Kumari, R.; McAlister, M.S.; Roe, S.M.; Nagl, S.; Pearl, L.H.; Selkirk, M.E.; Bianco, A.E.; Totty, N.F.; et al. Macrophage migration inhibitory factor of the parasitic nematode Trichinella spiralis. Biochem. J. 2001, 357, 373–383. [Google Scholar] [CrossRef] [PubMed]
- Donn, R.P.; Shelley, E.; Ollier, W.E.; Thomson, W. A novel 5′-flanking region polymorphism of macrophage migration inhibitory factor is associated with systemic-onset juvenile idiopathic arthritis. Arthritis Rheumatol. 2001, 44, 1782–1785. [Google Scholar] [CrossRef]
- Franco, R.; Cordomí, A.; Llinas Del Torrent, C.; Lillo, A.; Serrano-Marín, J.; Navarro, G.; Pardo, L. Structure and function of adenosine receptor heteromers. Cell. Mol. Life Sci. 2021, 78, 3957–3968. [Google Scholar] [CrossRef]
- Pedata, F.; Pugliese, A.M.; Coppi, E.; Dettori, I.; Maraula, G.; Cellai, L.; Melani, A. Adenosine A2A receptors modulate acute injury and neuroinflammation in brain ischemia. Mediat. Inflamm. 2014, 2014, 805198. [Google Scholar] [CrossRef]
- Oka, Y.; Hamada, M.; Nakazawa, Y.; Muramatsu, H.; Okuno, Y.; Higasa, K.; Shimada, M.; Takeshima, H.; Hanada, K.; Hirano, T.; et al. Digenic mutations in ALDH2 and ADH5 impair formaldehyde clearance and cause a multisystem disorder, AMeD syndrome. Sci. Adv. 2020, 6, eabd7197. [Google Scholar] [CrossRef]
- Takeshita, F.; Leifer, C.A.; Gursel, I.; Ishii, K.J.; Takeshita, S.; Gursel, M.; Klinman, D.M. Cutting edge: Role of Toll-like receptor 9 in CpG DNA-induced activation of human cells. J. Immunol. 2001, 167, 3555–3558. [Google Scholar] [CrossRef]
- Sohl, C.D.; Guengerich, F.P. Kinetic analysis of the three-step steroid aromatase reaction of human cytochrome P450 19A1. J. Biol. Chem. 2010, 285, 17734–17743. [Google Scholar] [CrossRef]
- Bouchoucha, N.; Samara-Boustani, D.; Pandey, A.V.; Bony-Trifunovic, H.; Hofer, G.; Aigrain, Y.; Polak, M.; Flück, C.E. Characterization of a novel CYP19A1 (aromatase) R192H mutation causing virilization of a 46,XX newborn, undervirilization of the 46,XY brother, but no virilization of the mother during pregnancies. Mol. Cell. Endocrinol. 2014, 390, 8–17. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Wang, X.; Kang, N.; Liu, Y.; Xu, G. Formononetin Exerts Neuroprotection in Parkinson’s Disease via the Activation of the Nrf2 Signaling Pathway. Molecules 2024, 29, 5364. https://doi.org/10.3390/molecules29225364
Wang X, Kang N, Liu Y, Xu G. Formononetin Exerts Neuroprotection in Parkinson’s Disease via the Activation of the Nrf2 Signaling Pathway. Molecules. 2024; 29(22):5364. https://doi.org/10.3390/molecules29225364
Chicago/Turabian StyleWang, Xiaotong, Nianxin Kang, Ying Liu, and Guojie Xu. 2024. "Formononetin Exerts Neuroprotection in Parkinson’s Disease via the Activation of the Nrf2 Signaling Pathway" Molecules 29, no. 22: 5364. https://doi.org/10.3390/molecules29225364
APA StyleWang, X., Kang, N., Liu, Y., & Xu, G. (2024). Formononetin Exerts Neuroprotection in Parkinson’s Disease via the Activation of the Nrf2 Signaling Pathway. Molecules, 29(22), 5364. https://doi.org/10.3390/molecules29225364