Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson’s Disease
Abstract
:1. Parkinson’s Disease
2. Clinical Studies of Parkinson’s Disease
3. The Role of Endogenous Neurotoxins in Idiopathic Parkinson’s Disease
4. Aminochrome as a Preclinical Model of Idiopathic Parkinson’s Disease
5. KEAP1/Nrf2 Signaling Pathway
6. Activation of the KEAP2/Nrf2 Signaling Pathway by Natural Products Increases the Expression of Antioxidant Enzymes
7. Activation of the KEAP2/Nrf2 Signaling Pathway by Natural Products to Search for Potential New Drugs in the Treatment of Parkinson’s Disease
8. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- LeWitt, P.A. Levodopa therapy for Parkinson’s disease: Pharmacokinetics and pharmacodynamics. Mov. Disord. 2015, 30, 64–72. [Google Scholar] [CrossRef] [PubMed]
- Morales-Martínez, A.; Martínez-Gómez, P.A.; Martinez-Fong, D.; Villegas-Rojas, M.M.; Pérez-Severiano, F.; Del Toro-Colín, M.A.; Delgado-Minjares, K.M.; Blanco-Alvarez, V.M.; Leon-Chavez, B.A.; Aparicio-Trejo, O.E.; et al. Oxidative Stress and Mitochondrial Complex I Dysfunction Correlate with Neurodegeneration in an α-Synucleinopathy Animal Model. Int. J. Mol. Sci. 2022, 23, 11394. [Google Scholar] [CrossRef] [PubMed]
- Ordonez, D.G.; Lee, M.K.; Feany, M.B. α-synuclein Induces Mitochondrial Dysfunction through Spectrin and the Actin Cy-toskeleton. Neuron 2018, 97, 108–124. [Google Scholar] [CrossRef] [PubMed]
- Rocha, E.M.; De Miranda, B.; Sanders, L.H. Alpha-synuclein: Pathology, mitochondrial dysfunction and neuroinflammation in Parkinson’s disease. Neurobiol. Dis. 2018, 109 Pt B, 249–257. [Google Scholar] [CrossRef]
- Nascimento, A.C.; Erustes, A.G.; Reckziegel, P.; Bincoletto, C.; Ureshino, R.P.; Pereira, G.J.S.; Smaili, S.S. α-Synuclein Overexpression Induces Lysosomal Dysfunction and Autophagy Impairment in Human Neuroblastoma SH-SY5Y. Neurochem. Res. 2020, 45, 2749–2761. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, M.; Wong, Y.C.; Ysselstein, D.; Severino, A.; Krainc, D. Synaptic, Mitochondrial, and Lysosomal Dysfunction in Parkinson’s Disease. Trends Neurosci. 2019, 42, 140–149. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Popova, B.; Galka, D.; Häffner, N.; Wang, D.; Schmitt, K.; Valerius, O.; Knop, M.; Braus, G.H. α-Synuclein Decreases the Abundance of Proteasome Subunits and Alters Ubiquitin Conjugates in Yeast. Cells 2021, 10, 2229. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chen, L.; Thiruchelvam, M.J.; Madura, K.; Richfield, E.K. Proteasome dysfunction in aged human alpha-synuclein transgenic mice. Neurobiol. Dis. 2006, 23, 120–126. [Google Scholar] [CrossRef] [PubMed]
- Pérez-Acuña, D.; Shin, S.J.; Rhee, K.H.; Kim, S.J.; Lee, S.J. α-Synuclein propagation leads to synaptic abnormalities in the cortex through microglial synapse phagocytosis. Mol. Brain 2023, 16, 72. [Google Scholar] [CrossRef]
- Brigo, F.; Erro, R.; Marangi, A.; Bhatia, K.; Tinazzi, M. Differentiating drug-induced parkinsonism from Parkinson’s disease: An update on non-motor symptoms and investigations. Park. Relat. Disord. 2014, 20, 808–814. [Google Scholar] [CrossRef]
- McKnight, S.; Hack, N. Toxin-Induced Parkinsonism. Neurol. Clin. 2020, 38, 853–865. [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] [PubMed] [PubMed Central]
- Lees, A.J.; Tolosa, E.; Olanow, C.W. Four pioneers of L-dopa treatment: Arvid Carlsson, Oleh Hornykiewicz, George Cotzias, and Melvin Yahr. Mov. Disord. 2015, 30, 19–36. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, A.; Chandran, K.; Kalivendi, S.V.; Joseph, J.; Antholine, W.E.; Hillard, C.J.; Kanthasamy, A.; Kanthasamy, A.; Kalyanaraman, B. Neuroprotection by a mitochondria-targeted drug in a Parkinson’s disease model. Free Radic. Biol. Med. 2010, 49, 1674–1684. [Google Scholar] [CrossRef] [PubMed]
- Segura-Aguilar, J. Neurotoxins as Preclinical Models for Parkinson’s Disease. Neurotox. Res. 2018, 34, 870–877. [Google Scholar] [CrossRef] [PubMed]
- Segura-Aguilar, J. The importance of choosing a preclinical model that reflects what happens in Parkinson’s disease. Neurochem. Int. 2019, 126, 203–209. [Google Scholar] [CrossRef] [PubMed]
- Klemann, C.J.H.M.; Martens, G.J.M.; Poelmans, G.; Visser, J.E. Validity of the MPTP-Treated Mouse as a Model for Parkinson’s Disease. Mol. Neurobiol. 2016, 53, 1625–1636. [Google Scholar] [CrossRef] [PubMed]
- Bankiewicz, K.S.; Sanchez-Pernaute, R.; Oiwa, Y.; Kohutnicka, M.; Cummins, A.; Eberling, J. Preclinical models of Parkinson’s disease. Curr. Protoc. Neurosci. 2001, 9, Unit9.4. [Google Scholar] [CrossRef] [PubMed]
- Athauda, D.; Foltynie, T. The ongoing pursuit of neuroprotective therapies in Parkinson disease. Nat. Rev. Neurol. 2015, 11, 25–40. [Google Scholar] [CrossRef] [PubMed]
- Parkinson Study Group QE3 Investigators; Beal, M.F.; Oakes, D.; Shoulson, I.; Henchcliffe, C.; Galpern, W.R.; Haas, R.; Juncos, J.L.; Nutt, J.G.; Voss, T.S.; et al. Randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson disease: No evidence of benefit. JAMA Neurol. 2014, 71, 543–552. [Google Scholar]
- Snow, B.J.; Rolfe, F.L.; Lockhart, M.M.; Frampton, C.M.; O’Sullivan, J.D.; Fung, V.; Smith, R.A.; Murphy, M.P.; Taylor, K.M.; Protect Study Group. A double-blind, placebo-controlled study to assess the mitochondria-targeted antioxidant MitoQ as a disease-modifying therapy in Parkinson’s disease. Mov. Disord. 2010, 25, 1670–1674. [Google Scholar] [CrossRef] [PubMed]
- Parkinson Study Group SURE-PD3 Investigators; Schwarzschild, M.A.; Ascherio, A.; Casaceli, C.; Curhan, G.C.; Fitzgerald, R.; Kamp, C.; Lungu, C.; Macklin, E.A.; Marek, K.; et al. Effect of Urate-Elevating Inosine on Early Parkinson Disease Progression: The SURE-PD3 Randomized Clinical Trial. JAMA 2021, 326, 926–939. [Google Scholar] [PubMed]
- Gouhier, C.; Chalon, S.; Aubert-Pouessel, A.; Venier-Julienne, M.C.; Jollivet, C.; Benoit, J.P.; Guilloteau, D. Protection of dopaminergic nigrostriatal afferents by GDNF delivered by microspheres in a rodent model of Parkinson’s disease. Synapse 2002, 44, 124–131. [Google Scholar] [CrossRef] [PubMed]
- Rosenblad, C.; Kirik, D.; Devaux, B.; Moffat, B.; Phillips, H.S.; Björklund, A. Protection and regeneration of nigral dopaminergic neurons by neurturin or GDNF in a partial lesion model of Parkinson’s disease after administration into the striatum or the lateral ventricle. Eur. J. Neurosci. 1999, 11, 1554–1566. [Google Scholar] [CrossRef] [PubMed]
- Duarte-Jurado, A.P.; Gopar-Cuevas, Y.; Saucedo-Cardenas, O.; Loera-Arias, M.J.; Montes-de-Oca-Luna, R.; Garcia-Garcia, A.; Rodriguez-Rocha, H. Antioxidant Therapeutics in Parkinson’s Disease: Current Challenges and Opportunities. Antioxidants 2021, 10, 453. [Google Scholar] [CrossRef]
- Williams, A. MPTP parkinsonism. Br. Med. J. 1984, 289, 1401–1402. [Google Scholar] [CrossRef]
- Ni, A.; Ernst, C. Evidence That Substantia Nigra Pars Compacta Dopaminergic Neurons Are Selectively Vulnerable to Oxidative Stress Because They Are Highly Metabolically Active. Front. Cell. Neurosci. 2022, 16, 826193. [Google Scholar] [CrossRef]
- Huenchuguala, S.; Segura-Aguilar, J. Single-neuron neurodegeneration as a degenerative model for Parkinson’s disease. Neural Regen. Res. 2024, 19, 529–535. [Google Scholar] [CrossRef]
- Henderson, M.X.; Cornblath, E.J.; Darwich, A.; Zhang, B.; Brown, H.; Gathagan, R.J.; Sandler, R.M.; Bassett, D.S.; Trojanowski, J.Q.; Lee, V.M.Y. Spread of α-synuclein pathology through the brain connectome is modulated by selective vulnerability and predicted by network analysis. Nat. Neurosci. 2019, 22, 1248–1257. [Google Scholar] [CrossRef]
- Kim, S.; Kwon, S.H.; Kam, T.I.; Panicker, N.; Karuppagounder, S.S.; Lee, S.; Lee, J.H.; Kim, W.R.; Kook, M.; Foss, C.A.; et al. Transneuronal Propagation of Pathologic α-Synuclein from the Gut to the Brain Models Parkinson’s Disease. Neuron 2019, 103, 627–641. [Google Scholar] [CrossRef]
- Okuzumi, A.; Hatano, T.; Matsumoto, G.; Nojiri, S.; Ueno, S.I.; Imamichi-Tatano, Y.; Kimura, H.; Kakuta, S.; Kondo, A.; Fukuhara, T.; et al. Propagative α-synuclein seeds as serum biomarkers for synucleinopathies. Nat. Med. 2023, 29, 1448–1455. [Google Scholar] [CrossRef] [PubMed]
- Bigi, A.; Cascella, R.; Cecchi, C. α-Synuclein oligomers and fibrils: Partners in crime in synucleinopathies. Neural Regen. Res. 2023, 18, 2332–2342. [Google Scholar] [CrossRef] [PubMed]
- Desplats, P.; Lee, H.J.; Bae, E.J.; Patrick, C.; Rockenstein, E.; Crews, L.; Spencer, B.; Masliah, E.; Lee, S.J. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc. Natl. Acad. Sci. USA 2009, 106, 13010–13015. [Google Scholar] [CrossRef]
- Goldstein, D.S. The “Sick-but-not-Dead” phenomenon applied to catecholamine deficiency in neurodegenerative diseases. Semin. Neurol. 2020, 40, 502–514. [Google Scholar] [CrossRef] [PubMed]
- Grünblatt, E.; Mandel, S.; Jacob-Hirsch, J.; Zeligson, S.; Amariglo, N.; Rechavi, G.; Li, J.; Ravid, R.; Roggendorf, W.; Riederer, P.; et al. Gene expression profiling of parkinsonian substantia nigra pars compacta; alterations in ubiquitin-proteasome heat shock protein iron and oxidative stress regulated proteins cell adhesion/cellular matrix and vesicle trafficking genes. J. Neural Transm. 2004, 111, 1543–1573. [Google Scholar] [CrossRef] [PubMed]
- Masato, A.; Plotegher, N.; Terrin, F.; Sandre, M.; Faustini, G.; Thor, A.; Adams, S.; Berti, G.; Cogo, S.; De Lazzari, F.; et al. DOPAL initiates αSynuclein-dependent impaired proteostasis and degeneration of neuronal projections in Parkinson’s disease. NPJ Park. Dis. 2023, 9, 42. [Google Scholar] [CrossRef]
- Jinsmaa, Y.; Sullivan, P.; Gross, D.; Cooney, A.; Sharabi, Y.; Goldstein, D.S. Divalent metal ions enhance DOPAL-induced oli-gomerization of alpha-synuclein. Neurosci. Lett. 2014, 569, 27–32. [Google Scholar] [CrossRef]
- Jinsmaa, Y.; Sullivan, P.; Sharabi, Y.; Goldstein, D.S. DOPAL is transmissible to and oligomerizes alpha-synuclein in human glial cells. Auton. Neurosci. 2016, 194, 46–51. [Google Scholar] [CrossRef]
- Bisaglia, M.; Mammi, S.; Bubacco, L. Kinetic and structural analysis of the early oxidation products of dopamine: Analysis of the interactions with alpha-synuclein. J. Biol. Chem. 2007, 282, 15597–15605. [Google Scholar] [CrossRef]
- Segura-Aguilar, J.; Metodiewa, D.; Welch, C.J. Metabolic activation of dopamine o-quinones to o-semiquinones by NADPH cytochrome P450 reductase may play an important role in oxidative stress and apoptotic effects. Biochim. Biophys. Acta 1998, 1381, 1–6. [Google Scholar] [CrossRef]
- Segura-Aguilar, J.; Lind, C. On the mechanism of the Mn3(+)-induced neurotoxicity of dopamine: Prevention of quinone-derived oxygen toxicity by DT diaphorase and superoxide dismutase. Chem. Biol. Interact. 1989, 72, 309–324. [Google Scholar] [CrossRef] [PubMed]
- Lozano, J.; Muñoz, P.; Nore, B.F.; Ledoux, S.; Segura-Aguilar, J. Stable expression of short interfering RNA for DT-diaphorase induces neurotoxicity. Chem. Res. Toxicol. 2010, 23, 1492–1496. [Google Scholar] [CrossRef] [PubMed]
- Paris, I.; Perez-Pastene, C.; Cardenas, S.; Iturriaga-Vasquez, P.; Muñoz, P.; Couve, E.; Caviedes, P.; Segura-Aguilar, J. Aminochrome induces disruption of actin, alpha-, and beta-tubulin cytoskeleton networks in substantia-nigra-derived cell line. Neurotox. Res. 2010, 18, 82–92. [Google Scholar] [CrossRef] [PubMed]
- Muñoz, P.; Cardenas, S.; Huenchuguala, S.; Briceño, A.; Couve, E.; Paris, I.; Segura-Aguilar, J. DT-Diaphorase Prevents Aminochrome-Induced Alpha-Synuclein Oligomer Formation and Neurotoxicity. Toxicol. Sci. 2015, 145, 37–47. [Google Scholar] [CrossRef] [PubMed]
- Aguirre, P.; Urrutia, P.; Tapia, V.; Villa, M.; Paris, I.; Segura-Aguilar, J.; Núñez, M.T. The dopamine metabolite aminochrome inhibits mitochondrial complex I and modifies the expression of iron transporters DMT1 and FPN1. Biometals 2012, 25, 795–803. [Google Scholar] [CrossRef]
- Meléndez, C.; Muñoz, P.; Segura-Aguilar, J. DT-Diaphorase Prevents Aminochrome-Induced Lysosome Dysfunction in SH-SY5Y Cells. Neurotox. Res. 2019, 35, 255–259. [Google Scholar] [CrossRef]
- Herrera, A.; Muñoz, P.; Paris, I.; Díaz-Veliz, G.; Mora, S.; Inzunza, J.; Hultenby, K.; Cardenas, C.; Jaña, F.; Raisman-Vozari, R.; et al. Aminochrome induces dopaminergic neuronal dysfunction: A new animal model for Parkinson’s disease. Cell. Mol. Life Sci. 2016, 73, 3583–3597. [Google Scholar] [CrossRef]
- Arriagada, C.; Paris, I.; Sanchez de las Matas, M.J.; Martinez-Alvarado, P.; Cardenas, S.; Castañeda, P.; Graumann, R.; Perez-Pastene, C.; Olea-Azar, C.; Couve, E.; et al. On the neurotoxicity mechanism of leukoaminochrome o-semiquinone radical derived from dopamine oxidation: Mitochondria damage, necrosis, and hydroxyl radical formation. Neurobiol. Dis. 2004, 16, 468–477. [Google Scholar] [CrossRef]
- Muñoz, P.; Huenchuguala, S.; Paris, I.; Segura-Aguilar, J. Dopamine oxidation and autophagy. Park. Dis. 2012, 2012, 920953. [Google Scholar] [CrossRef]
- Zafar, K.S.; Inayat-Hussain, S.H.; Siegel, D.; Bao, A.; Shieh, B.; Ross, D. Overexpression of NQO1 protects human SK-N-MC neuroblastoma cells against dopamine-induced cell death. Toxicol. Lett. 2006, 166, 261–267. [Google Scholar] [CrossRef]
- de Araújo, F.M.; Ferreira, R.S.; Souza, C.S.; Dos Santos, C.C.; Rodrigues, T.L.R.S.; e Silva, J.H.C.; Gasparotto, J.; Gelain, D.P.; El-Bachá, R.S.; Costa, M.d.F.D.; et al. Aminochrome decreases NGF, GDNF and induces neuroinflammation in organotypic midbrain slice cultures. Neurotoxicology 2018, 66, 98–106. [Google Scholar] [CrossRef] [PubMed]
- Xiong, R.; Siegel, D.; Ross, D. Quinone-induced protein handling changes: Implications for major protein handling systems in quinone-mediated toxicity. Toxicol. Appl. Pharmacol. 2014, 280, 285–295. [Google Scholar] [CrossRef] [PubMed]
- Du, X.Y.; Xie, X.X.; Liu, R.T. The Role of α-Synuclein Oligomers in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 8645. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cuevas, C.; Huenchuguala, S.; Muñoz, P.; Villa, M.; Paris, I.; Mannervik, B.; Segura-Aguilar, J. Glutathione transferase-M2-2 secreted from glioblastoma cell protects SH-SY5Y cells from aminochrome neurotoxicity. Neurotox. Res. 2015, 27, 217–228. [Google Scholar] [CrossRef] [PubMed]
- Segura-Aguilar, J.; Muñoz, P.; Inzunza, J.; Varshney, M.; Nalvarte, I.; Mannervik, B. Neuroprotection against Aminochrome Neurotoxicity: Glutathione Transferase M2-2 and DT-Diaphorase. Antioxidants 2022, 11, 296. [Google Scholar] [CrossRef] [PubMed]
- Segura-Aguilar, J.; Mannervik, B.; Inzunza, J.; Varshney, M.; Nalvarte, I.; Muñoz, P. Astrocytes protect dopaminergic neurons against aminochrome neurotoxicity. Neural Regen. Res. 2022, 17, 1861–1866. [Google Scholar] [CrossRef]
- Zecca, L.; Fariello, R.; Riederer, P.; Sulzer, D.; Gatti, A.; Tampellini, D. The absolute concentration of nigral neuromelanin, assayed by a new sensitive method, increases throughout the life and is dramatically decreased in Parkinson’s disease. FEBS Lett. 2002, 510, 216–220. [Google Scholar] [CrossRef]
- Zucca, F.A.; Segura-Aguilar, J.; Ferrari, E.; Muñoz, P.; Paris, I.; Sulzer, D.; Sarna, T.; Casella, L.; Zecca, L. Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson’s disease. Prog. Neurobiol. 2017, 155, 96–119. [Google Scholar] [CrossRef]
- Reisman, S.A.; Yeager, R.L.; Yamamoto, M.; Klaassen, C.D. Increased Nrf2 activation in livers from Keap1-knockdown mice increases expression of cytoprotective genes that detoxify electrophiles more than those that detoxify reactive oxygen species. Toxicol. Sci. 2009, 108, 35–47. [Google Scholar] [CrossRef]
- Lin, X.; Bai, D.; Wei, Z.; Zhang, Y.; Huang, Y.; Deng, H.; Huang, X. Curcumin attenuates oxidative stress in RAW264.7 cells by increasing the activity of antioxidant enzymes and activating the Nrf2-Keap1 pathway. PLoS ONE 2019, 14, e0216711. [Google Scholar] [CrossRef]
- Silva-Adaya, D.; Garza-Lombó, C.; Gonsebatt, M.E. Xenobiotic transport and metabolism in the human brain. Neurotoxicology 2021, 86, 125–138. [Google Scholar] [CrossRef] [PubMed]
- Baird, L.; Yamamoto, M. The Molecular Mechanisms Regulating the KEAP1-NRF2 Pathway. Mol. Cell. Biol. 2020, 40, e00099-20. [Google Scholar] [CrossRef] [PubMed]
- Tonelli, C.; Chio, I.I.C.; Tuveson, D.A. Transcriptional Regulation by Nrf2. Antioxid. Redox Signal. 2018, 29, 1727–1745. [Google Scholar] [CrossRef] [PubMed]
- Zhang, D.D.; Hannink, M. Distinct cysteine residues in Keap1 are required for Keap1-dependent ubiquitination of Nrf2 and for stabilization of Nrf2 by chemopreventive agents and oxidative stress. Mol. Cell. Biol. 2003, 23, 8137–8151. [Google Scholar] [CrossRef]
- 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]
- He, X.; Ma, Q. NRF2 cysteine residues are critical for oxidant/electrophile-sensing, Kelch-like ECH-associated protein-1-dependent ubiquitination-proteasomal degradation, and transcription activation. Mol. Pharmacol. 2009, 76, 1265–1278. [Google Scholar] [CrossRef]
- Wang, Y.; Li, J.; Gu, J.; He, W.; Ma, B.; Fan, H. Hyperoside, a natural flavonoid compound, attenuates Triptolide-induced testicular damage by activating the Keap1-Nrf2 and SIRT1-PGC1α signalling pathway. J. Pharm. Pharmacol. 2022, 74, 985–995. [Google Scholar] [CrossRef]
- Chen, Y.; Ye, L.; Li, W.; Li, D.; Li, F. Hyperoside protects human kidney 2 cells against oxidative damage induced by oxalic acid. Mol. Med. Rep. 2018, 18, 486–494. [Google Scholar] [CrossRef]
- Zhou, Q.; Chen, B.; Wang, X.; Wu, L.; Yang, Y.; Cheng, X.; Hu, Z.; Cai, X.; Yang, J.; Sun, X.; et al. Sulforaphane protects against rotenone-induced neurotoxicity in vivo: Involvement of the mTOR, Nrf2, and autophagy pathways. Sci. Rep. 2016, 6, 32206. [Google Scholar] [CrossRef]
- Kleszczyński, K.; Ernst, I.M.; Wagner, A.E.; Kruse, N.; Zillikens, D.; Rimbach, G.; Fischer, T.W. Sulforaphane and phenylethyl isothiocyanate protect human skin against UVR-induced oxidative stress and apoptosis: Role of Nrf2-dependent gene expression and antioxidant enzymes. Pharmacol. Res. 2013, 78, 28–40. [Google Scholar] [CrossRef]
- Matana, A.; Ziros, P.G.; Chartoumpekis, D.V.; Renaud, C.O.; Polašek, O.; Hayward, C.; Zemunik, T.; Sykiotis, G.P. Rare and common genetic variations in the Keap1/Nrf2 antioxidant response pathway impact thyroglobulin gene expression and circulating levels, respectively. Biochem. Pharmacol. 2020, 173, 113605. [Google Scholar] [CrossRef] [PubMed]
- Chikara, S.; Nagaprashantha, L.D.; Singhal, J.; Horne, D.; Awasthi, S.; Singhal, S.S. Oxidative stress and dietary phytochemicals: Role in cancer chemoprevention and treatment. Cancer Lett. 2018, 413, 122–134. [Google Scholar] [CrossRef] [PubMed]
- Shen, C.; Cheng, W.; Yu, P.; Wang, L.; Zhou, L.; Zeng, L.; Yang, Q. Resveratrol pretreatment attenuates injury and promotes proliferation of neural stem cells following oxygen-glucose deprivation/reoxygenation by upregulating the expression of Nrf2, HO-1 and NQO1 in vitro. Mol. Med. Rep. 2016, 14, 3646–3654. [Google Scholar] [CrossRef] [PubMed]
- Xu, J.; Zhou, L.; Weng, Q.; Xiao, L.; Li, Q. Curcumin analogues attenuate Aβ25-35-induced oxidative stress in PC12 cells via Keap1/Nrf2/HO-1 signaling pathways. Chem. Biol. Interact. 2019, 305, 171–179. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.P.; Chang, H.C.; Lu, L.S.; Liu, D.Z.; Wang, T.J. Activation of kelch-like ECH-associated protein 1/nuclear factor erythroid 2-related factor 2/antioxidant response element pathway by curcumin enhances the anti-oxidative capacity of corneal endothelial cells. Biomed. Pharmacother. 2021, 141, 111834. [Google Scholar] [CrossRef]
- Xie, Y.; Zhao, Q.Y.; Li, H.Y.; Zhou, X.; Liu, Y.; Zhang, H. Curcumin ameliorates cognitive deficits heavy ion irradiation-induced learning and memory deficits through enhancing of Nrf2 antioxidant signaling pathways. Pharmacol. Biochem. Behav. 2014, 126, 181–186. [Google Scholar] [CrossRef]
- Tanigawa, S.; Fujii, M.; Hou, D.X. Action of Nrf2 and Keap1 in ARE-mediated NQO1 expression by quercetin. Free Radic. Biol. Med. 2007, 42, 1690–1703. [Google Scholar] [CrossRef]
- Gum, S.I.; Shin, H.M.; Cho, M.K. The small GTPases regulate HMC05-induced NQO-1 expression with an antioxidant effect in smooth muscle cells. Biol. Pharm. Bull. 2014, 37, 1626–1632. [Google Scholar] [CrossRef]
- Heyninck, K.; Sabbe, L.; Chirumamilla, C.S.; Szarc Vel Szic, K.; Vander Veken, P.; Lemmens, K.J.A.; Lahtela-Kakkonen, M.; Naulaerts, S.; Op de Beeck, K.; Laukens, K.; et al. Withaferin A induces heme oxygenase (HO-1) expression in endothelial cells via activation of the Keap1/Nrf2 pathway. Biochem. Pharmacol. 2016, 109, 48–61. [Google Scholar] [CrossRef]
- Sun, Y.; He, L.; Wang, W.; Xie, Z.; Zhang, X.; Wang, P.; Wang, L.; Yan, C.; Liu, Z.; Zhao, J.; et al. Activation of Atg7-dependent autophagy by a novel inhibitor of the Keap1-Nrf2 protein-protein interaction from Penthorum chinense Pursh. attenuates 6-hydroxydopamine-induced ferroptosis in zebrafish and dopaminergic neurons. Food Funct. 2022, 13, 7885–7900. [Google Scholar] [CrossRef]
- Hassanein, E.H.M.; Shalkami, A.S.; Khalaf, M.M.; Mohamed, W.R.; Hemeida, R.A.M. The impact of Keap1/Nrf2, P38MAPK/NF-κB and Bax/Bcl2/caspase-3 signaling pathways in the protective effects of berberine against methotrexate-induced nephrotoxicity. Biomed. Pharmacother. 2019, 109, 47–56. [Google Scholar] [CrossRef] [PubMed]
- Pistol, G.C.; Marin, D.E.; Bulgaru, V.C.; Anghel, A.C.; Sărăcilă, M.; Vlassa, M.; Filip, M.; Taranu, I. Grape seed meal by-product is able to counteract oxidative stress induced by lipopolysaccharide and dextran sulphate in IPEC cells and piglets after weaning. PLoS ONE 2023, 18, e0283607. [Google Scholar] [CrossRef] [PubMed]
- Shen, Y.; Teng, L.; Qu, Y.; Huang, Y.; Peng, Y.; Tang, M.; Fu, Q. Hederagenin Suppresses Inflammation and Cartilage Degradation to Ameliorate the Progression of Osteoarthritis: An In vivo and In vitro Study. Inflammation 2023, 46, 655–678. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Zheng, B.; Liu, P.; Zhang, J.; Chu, X.; Dong, C.; Shi, J.; Liang, Y.; Chu, L.; Liu, Y.; et al. Exploration of the hepatoprotective effect and mechanism of magnesium isoglycyrrhizinate in mice with arsenic trioxide-induced acute liver injury. Mol. Med. Rep. 2021, 23, 438. [Google Scholar] [CrossRef] [PubMed]
- Dou, C.; Shang, Z.; Qiao, J.; Wang, Y.; Li, H. Clostridium butyricum Protects IPEC-J2 Cells from ETEC K88-Induced Oxidative Damage by Activating the Nrf2/ARE Signaling Pathway. Oxidative Med. Cell. Longev. 2021, 2021, 4464002. [Google Scholar] [CrossRef]
- Chen, H.; Lou, Y.; Lin, S.; Tan, X.; Zheng, Y.; Yu, H.; Jiang, R.; Wei, Y.; Huang, H.; Qi, X.; et al. Formononetin, a bioactive isoflavonoid constituent from Astragalus membranaceus (Fisch.) Bunge, ameliorates type 1 diabetes mellitus via activation of Keap1/Nrf2 signaling pathway: An integrated study supported by network pharmacology and experimental validation. J. Ethnopharmacol. 2024, 322, 117576. [Google Scholar] [CrossRef]
- Hao, Y.; Miao, J.; Liu, W.; Peng, L.; Chen, Y.; Zhong, Q. Formononetin protects against cisplatin-induced acute kidney injury through activation of the PPARα/Nrf2/HO-1/NQO1 pathway. Int. J. Mol. Med. 2021, 47, 511–522. [Google Scholar] [CrossRef]
- Cai, Y.; Zhu, J.; Zhu, L.; Hong, L.; Zhang, J.; Kong, L.; Chen, C.; Luo, J. Physalin H ameliorates LPS-induced acute lung injury via KEAP1/NRF2 axis. Int. Immunopharmacol. 2024, 131, 111789. [Google Scholar] [CrossRef]
- Ding, X.; Zhao, H.; Qiao, C. Icariin protects podocytes from NLRP3 activation by Sesn2-induced mitophagy through the Keap1-Nrf2/HO-1 axis in diabetic nephropathy. Phytomedicine 2022, 99, 154005. [Google Scholar] [CrossRef]
- Yang, Y.; Chen, S.; Tao, L.; Gan, S.; Luo, H.; Xu, Y.; Shen, X. Inhibitory Effects of Oxymatrine on Transdifferentiation of Neonatal Rat Cardiac Fibroblasts to Myofibroblasts Induced by Aldosterone via Keap1/Nrf2 Signaling Pathways In Vitro. Med. Sci. Monit. 2019, 25, 5375–5388. [Google Scholar] [CrossRef]
- Ma, B.X.; Meng, X.S.; Tong, J.; Ge, L.L.; Zhou, G.; Wang, Y.W. Protective effects of Coptis chinensis inflorescence extract and linarin against carbon tetrachloride-induced damage in HepG2 cells through the MAPK/Keap1-Nrf2 pathway. Food Funct. 2018, 9, 2353–2361. [Google Scholar] [CrossRef] [PubMed]
- Kim, N.H.; Oh, M.K.; Park, H.J.; Kim, I.S. Auranofin, a gold(I)-containing antirheumatic compound, activates Keap1/Nrf2 signaling via Rac1/iNOS signal and mitogen-activated protein kinase activation. J. Pharmacol. Sci. 2010, 113, 246–254. [Google Scholar] [CrossRef] [PubMed]
- Tossetta, G.; Marzioni, D. Natural and synthetic compounds in Ovarian Cancer: A focus on NRF2/KEAP1 pathway. Pharmacol. Res. 2022, 183, 106365. [Google Scholar] [CrossRef] [PubMed]
- Tossetta, G.; Marzioni, D. Targeting the NRF2/KEAP1 pathway in cervical and endometrial cancers. Eur. J. Pharmacol. 2023, 941, 175503. [Google Scholar] [CrossRef]
- Wu, A.; Yang, Z.; Huang, Y.; Yuan, H.; Lin, C.; Wang, T.; Zhao, Z.; Zhou, Y.; Zhu, C. Natural phenylethanoid glycosides isolated from Callicarpa kwangtungensis suppressed lipopolysaccharide-mediated inflammatory response via activating Keap1/Nrf2/HO-1 pathway in RAW 264.7 macrophages cell. J. Ethnopharmacol. 2020, 258, 112857. [Google Scholar] [CrossRef]
- Jia, Y.N.; Lu, H.P.; Peng, Y.L.; Zhang, B.S.; Gong, X.B.; Su, J.; Zhou, Y.; Pan, M.H.; Xu, L. Oxyresveratrol prevents lipopolysaccharide/d-galactosamine-induced acute liver injury in mice. Int. Immunopharmacol. 2018, 56, 105–112. [Google Scholar] [CrossRef]
- Li, J.; Gao, Y.; Cui, L.; He, H.; Zheng, J.; Mo, S.; Zhou, X.; Chu, S.; Sun, X.; Chen, N.; et al. Combination of monoammonium glycyrrhizinate and cysteine hydrochloride protects mice against acetaminophen-induced liver injury via Keap1/Nrf2/ARE pathway. J. Pharm. Pharmacol. 2022, 74, 730–739. [Google Scholar] [CrossRef]
- Liao, Q.; Chen, W.; Tong, Z.; Xue, M.; Gu, T.; Yuan, Y.; Song, Z.; Tao, Z. Shufeng Jiedu capsules protect rats against LPS-induced acute lung injury via activating NRF2-associated antioxidant pathway. Histol. Histopathol. 2021, 36, 317–324. [Google Scholar] [CrossRef]
- Zhang, Z.; Peng, L.; Fu, Y.; Wang, W.; Wang, P.; Zhou, F. Ginnalin A Binds to the Subpockets of Keap1 Kelch Domain To Activate the Nrf2-Regulated Antioxidant Defense System in SH-SY5Y Cells. ACS Chem. Neurosci. 2021, 12, 872–882. [Google Scholar] [CrossRef]
- Mo, M.; Li, S.; Dong, Z.; Li, C.; Sun, Y.; Li, A.; Zhao, Z. S-allylmercaptocysteine ameliorates lipopolysaccharide-induced acute lung injury in mice by inhibiting inflammation and oxidative stress via nuclear factor kappa B and Keap1/Nrf2 pathways. Int. Immunopharmacol. 2020, 81, 106273. [Google Scholar] [CrossRef]
- Qu, L.; Ji, L.; Wang, C.; Luo, H.; Li, S.; Peng, W.; Yin, F.; Lu, D.; Liu, X.; Kong, L.; et al. Synthesis and evaluation of multi-target-directed ligands with BACE-1 inhibitory and Nrf2 agonist activities as potential agents against Alzheimer’s disease. Eur. J. Med. Chem. 2021, 219, 113441. [Google Scholar] [CrossRef] [PubMed]
- Satoh, T.; Rezaie, T.; Seki, M.; Sunico, C.R.; Tabuchi, T.; Kitagawa, T.; Yanagitai, M.; Senzaki, M.; Kosegawa, C.; Taira, H.; et al. Dual neuroprotective pathways of a pro-electrophilic compound via HSF-1-activated heat-shock proteins and Nrf2-activated phase 2 antioxidant response enzymes. J. Neurochem. 2011, 119, 569–578. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.; Zheng, B.; Li, J.; Shi, J.; Chu, L.; Han, X.; Chu, X.; Zhang, X.; Zhang, J. Crocin ameliorates arsenic trioxide-induced cardiotoxicity via Keap1-Nrf2/HO-1 pathway: Reducing oxidative stress, inflammation, and apoptosis. Biomed. Pharmacother. 2020, 131, 110713. [Google Scholar] [CrossRef] [PubMed]
- Shang, H.; Yang, D.; Zhang, W.; Li, T.; Ren, X.; Wang, X.; Zhao, W. Time course of Keap1-Nrf2 pathway expression after experimental intracerebral haemorrhage: Correlation with brain oedema and neurological deficit. Free Radic. Res. 2013, 47, 368–375. [Google Scholar] [CrossRef] [PubMed]
- Bolton, J.L.; Dunlap, T.L.; Hajirahimkhan, A.; Mbachu, O.; Chen, S.N.; Chadwick, L.; Nikolic, D.; van Breemen, R.B.; Pauli, G.F.; Dietz, B.M. The Multiple Biological Targets of Hops and Bioactive Compounds. Chem. Res. Toxicol. 2019, 32, 222–233. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.Y.; Fu, R.H.; Chou, R.H.; Chen, J.H.; Wu, C.R.; Chang, S.W.; Tsai, C.W. Inhibition of JNK by pi class of glutathione S-transferase through PKA/CREB pathway is associated with carnosic acid protection against 6-hydroxydopamine-induced apoptosis. Food Chem. Toxicol. 2017, 103, 194–202. [Google Scholar] [CrossRef]
- Wang, Z.; Ji, C.; Wu, L.; Qiu, J.; Li, Q.; Shao, Z.; Chen, G. Tert-butylhydroquinone alleviates early brain injury and cognitive dysfunction after experimental subarachnoid hemorrhage: Role of Keap1/Nrf2/ARE pathway. PLoS ONE 2014, 9, e97685. [Google Scholar] [CrossRef]
- Pan, P.K.; Qiao, L.Y.; Wen, X.N. Safranal prevents rotenone-induced oxidative stress and apoptosis in an in vitro model of Parkinson’s disease through regulating Keap1/Nrf2 signaling pathway. Cell. Mol. Biol. (Noisy-le-grand) 2016, 62, 11–17. [Google Scholar] [CrossRef]
- Saw, C.L.; Yang, A.Y.; Guo, Y.; Kong, A.N. Astaxanthin and omega-3 fatty acids individually and in combination protect against oxidative stress via the Nrf2-ARE pathway. Food Chem. Toxicol. 2013, 62, 869–875. [Google Scholar] [CrossRef] [PubMed]
- Kuo, W.H.; Wang, C.J.; Young, S.C.; Sun, Y.C.; Chen, Y.J.; Chou, F.P. Differential induction of the expression of GST subunits by geniposide in rat hepatocytes. Pharmacology 2004, 70, 15–22. [Google Scholar] [CrossRef] [PubMed]
- Segura-Aguilar, J.; Mannervik, B. A Preclinical Model for Parkinson’s Disease Based on Transcriptional Gene Activation via KEAP1/NRF2 to Develop New Antioxidant Therapies. Antioxidants 2023, 12, 673. [Google Scholar] [CrossRef] [PubMed]
- Huenchuguala, S.; Segura-Aguilar, J. Targets to Search for New Pharmacological Treatment in Idiopathic Parkinson’s Disease According to the Single-Neuron Degeneration Model. Biomolecules 2024, 14, 673. [Google Scholar] [CrossRef] [PubMed]
- Kwon, I.H.; Choi, H.S.; Shin, K.S.; Lee, B.K.; Lee, C.K.; Hwang, B.Y.; Lim, S.C.; Lee, M.K. Effects of berberine on 6-hydroxydopamine-induced neurotoxicity in PC12 cells and a rat model of Parkinson’s disease. Neurosci. Lett. 2010, 486, 29–33. [Google Scholar] [CrossRef] [PubMed]
- Khuwaja, G.; Khan, M.M.; Ishrat, T.; Ahmad, A.; Raza, S.S.; Ashafaq, M.; Javed, H.; Khan, M.B.; Khan, A.; Vaibhav, K.; et al. Neuroprotective effects of curcumin on 6-hydroxydopamine-induced Parkinsonism in rats: Behavioral, neurochemical and immunohistochemical studies. Brain Res. 2011, 1368, 254–263. [Google Scholar] [CrossRef] [PubMed]
- Ren, R.; Shi, C.; Cao, J.; Sun, Y.; Zhao, X.; Guo, Y.; Wang, C.; Lei, H.; Jiang, H.; Ablat, N.; et al. Neuroprotective Effects of a Standardized Flavonoid Extract of Safflower against Neurotoxin-Induced Cellular and Animal Models of Parkinson’s Disease. Sci. Rep. 2016, 6, 22135. [Google Scholar] [CrossRef] [PubMed]
- Ablat, N.; Lv, D.; Ren, R.; Xiaokaiti, Y.; Ma, X.; Zhao, X.; Sun, Y.; Lei, H.; Xu, J.; Ma, Y.; et al. Neuroprotective Effects of a Standardized Flavonoid Extract from Safflower against a Rotenone-Induced Rat Model of Parkinson’s Disease. Molecules 2016, 21, 1107. [Google Scholar] [CrossRef]
- Ahmad, M.; Saleem, S.; Ahmad, A.S.; Yousuf, S.; Ansari, M.A.; Khan, M.B.; Ishrat, T.; Chaturvedi, R.K.; Agrawal, A.K.; Islam, F. Ginkgo biloba affords dose-dependent protection against 6-hydroxydopamine-induced parkinsonism in rats: Neurobehavioural, neurochemical and immunohistochemical evidences. J. Neurochem. 2005, 93, 94–104. [Google Scholar] [CrossRef]
- Hritcu, L.; Foyet, H.S.; Stefan, M.; Mihasan, M.; Asongalem, A.E.; Kamtchouing, P. Neuroprotective effect of the methanolic extract of Hibiscus asper leaves in 6-hydroxydopamine-lesioned rat model of Parkinson’s disease. J. Ethnopharmacol. 2011, 137, 585–591. [Google Scholar] [CrossRef]
- Rabiei, Z.; Solati, K.; Amini-Khoei, H. Phytotherapy in treatment of Parkinson’s disease: A review. Pharm. Biol. 2019, 57, 355–362. [Google Scholar] [CrossRef]
- Sun, C.P.; Zhou, J.J.; Yu, Z.L.; Huo, X.K.; Zhang, J.; Morisseau, C.; Hammock, B.D.; Ma, X.C. Kurarinone alleviated Parkinson’s disease via stabilization of epoxyeicosatrienoic acids in animal model. Proc. Natl. Acad. Sci. USA 2022, 119, e2118818119. [Google Scholar] [CrossRef]
Compound | Model | Increased Enzymes | Methodology (Nrf2) | Reference (N°) |
---|---|---|---|---|
Hyperoside | Testicular injury Renal cells | CAT, Mn-SOD, HO-1, NQO1 NQO1 | Western blot Western blot | [67] [68] |
Sulforaphane | Rotenone animal Keratinocytes | HO-1, NQO1, HO-1, NQO1, γGCS | Western blot mRNA level | [69] [70] |
Resveratrol | PC12 cells Neural stem cells | HO-1 HO-1, NQO1 | Western blot Western blot | [72] [73] |
Curcumin | PC12 cells Corneal endothelial cells Kunming mice | HO-1 SOD-1, HO-1 HO-1,NQO1, γGCS | Western blot Nuclear/cytosol fractionation kit Western blot | [74] [75] [76] |
Quercetin | Human HepG2 cells | NQO1 | Western blot | [77] |
Epigallocatechin gallate | Mammary epithelial cells | HO-1, SOD-1 | Western blot | [72] |
Withania somnifera | Primary human umbilical vein endothelial cells | HO-1 | Western blot | [79] |
Thonningianin A | SH-SY5Y cells | HO-1 | Western blot Molecular docking | [80] |
Berberine | Male rats | SOD-1 | q-RT-PCR | [81] |
Byproduct grape seed meal | IPEC-1 cells | CAT, SOD-1, GPx, eNOS, iNO | q-RT-PCR | [84] |
Hederagenin | Extracellular matrix | HO-1, NQO1 | Western blot | [83] |
Isoglycyrrhizinate | KunMing mice | CAT, SOD-1 | Western blot | [84] |
Clostridium butyricum | IPEC-J2) cells | SOD-1, GPx | siRNA | [85] |
Formononetin | C57BL/6J mice HK-2 cells | HO-1, NQO1 HO-1, NQO1 | Nrf2 knockout Western blot | [86] [87] |
Physalin H | RAW264.7 cells | HO-1, NQO1 | qRT-PCR, western blot | [88] |
Icarin | Diabetes mellitus rats | HO-1 | Western blot | [89] |
Oxymatrine | Primary cardiac fibroblasts | HO-1 | qRT-PCR, siRNA | [90] |
Coptis chinensis | HepG2 cells | GST, NQO1, HO-1 | Western blot | [91] |
Withaferin A | Endothelial cells | HO-1 | Western blot, siRNA | [79] |
Callicarpa kwangtungensis | RAW 264.7 macrophages | HO-1, NQO1 | Western blot | [95] |
Oxyresveratrol | Hepatocytes | HO-1, NOQ1 | Western blot | [96] |
Monoammonium glycyrrhizinate | Hepatic injury | HO-1, NOQ1 | Western blot | [97] |
Shufeng Jiedu | LPS-induced acute lung injury | HO-1, NQO1 | Western blot | [98] |
Ginnalin A | SH-SY5Y cells | HO-1, NQO1 | Western blot, qRT-PCR | [99] |
S-allylmercaptocysteine | LPS-induced acute lung injury | HO-1, NQO1 | Western blot | [100] |
Crocin | Male rats | HO-1, NQO1 | Western blot | [103] |
Chalcone xanthohumol | PC12 cells | NQO1 | Nrf2 knockdown | [101] |
Safranal | Rotenone | HO-1,GST, NQO1 | Western blot | [108] |
Astaxanthin | Increase | HO-1, NQO1, GSTM2 | qRT-PCR | [109] |
Docosahexaenoic acid | Increase | HO-1, NQO1, GSTM2 | qRT-PCR | [109] |
Eicosapentaenoic acid | Increase | HO-1, NQO1, GSTM2 | qRT-PCR | [109] |
Thonningianin A | SH-SY5Y cells | HO-1 | Immunofluorescence | [80] |
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
Huenchuguala, S.; Segura-Aguilar, J. Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson’s Disease. Antioxidants 2024, 13, 1125. https://doi.org/10.3390/antiox13091125
Huenchuguala S, Segura-Aguilar J. Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson’s Disease. Antioxidants. 2024; 13(9):1125. https://doi.org/10.3390/antiox13091125
Chicago/Turabian StyleHuenchuguala, Sandro, and Juan Segura-Aguilar. 2024. "Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson’s Disease" Antioxidants 13, no. 9: 1125. https://doi.org/10.3390/antiox13091125
APA StyleHuenchuguala, S., & Segura-Aguilar, J. (2024). Natural Compounds That Activate the KEAP1/Nrf2 Signaling Pathway as Potential New Drugs in the Treatment of Idiopathic Parkinson’s Disease. Antioxidants, 13(9), 1125. https://doi.org/10.3390/antiox13091125