A Highly Selective In Vitro JNK3 Inhibitor, FMU200, Restores Mitochondrial Membrane Potential and Reduces Oxidative Stress and Apoptosis in SH-SY5Y Cells
Abstract
:1. Introduction
2. Results
2.1. The Cytotoxic and Neuroprotective Effect of FMU200 in Undifferentiated and RA-Differentiated SH-SY5Y Cells
2.2. Response to H2O2: Apoptosis, ROS Production and MMP
2.3. JNK Inhibition by FMU200
2.4. Anti-Inflammatory Effect
3. Discussion
4. Materials and Methods
4.1. Cell Lines and Reagents
4.2. Cell Culture Methods
4.2.1. SH-SY5Y Cell Line
4.2.2. SH-SY5Y Differentiation Protocol
4.2.3. RAW264.7 Cell Line
4.3. Determination of Cell Viability and Neuroprotection Potential by MTT Assay
4.3.1. MTT Assay and Cytotoxicity of FMU200
4.3.2. Neuroprotection Potential
4.4. Apoptosis Assay by Flow Cytometry
4.5. Mitochondrial Membrane Potential (MMP) Assay
4.6. ROS Production
4.7. Western Blot
4.8. Cytokine Determination in RAW264.7 Cell Line
4.9. Statistical Analysis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Waxman, S. From Neuroscience to Neurology: Neuroscience, Molecular Medicine, and the Therapeutic Transformation of Neurology, 1st ed.; Academic Press: Amsterdam, The Netherlands, 2004; p. 552. ISBN 0-12-738903-2. [Google Scholar]
- Vajda, F.J.E. Neuroprotection and neurodegenerative disease. J. Clin. Neurosci. 2002, 9, 4–8. [Google Scholar] [CrossRef]
- Drug Safety and Availability|FDA. Available online: https://www.fda.gov/drugs/drug-safety-and-availability (accessed on 8 July 2020).
- Liu, P.-P.; Xie, Y.; Meng, X.-Y.; Kang, J.-S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct. Target. Ther. 2019, 4, 29. [Google Scholar] [CrossRef]
- Kondoh, K.; Nishida, E. Regulation of MAP kinases by MAP kinase phosphatases. Biochim. Biophys. Acta 2007, 1773, 1227–1237. [Google Scholar] [CrossRef] [Green Version]
- Tournier, C.; Hess, P.; Yang, D.D.; Xu, J.; Turner, T.K.; Nimnual, A.; Bar-Sagi, D.; Jones, S.N.; Flavell, R.A.; Davis, R.J. Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway. Science 2000, 288, 870–874. [Google Scholar] [CrossRef] [PubMed]
- Zhu, X.; Castellani, R.J.; Takeda, A.; Nunomura, A.; Atwood, C.S.; Perry, G.; Smith, M.A. Differential activation of neuronal ERK, JNK / SAPK and p38 in Alzheimer disease: The ‘two hit’ hypothesis. Mech. Ageing Dev. 2001, 123, 39–46. [Google Scholar] [CrossRef]
- Gourmaud, S.; Paquet, C.; Dumurgier, J.; Pace, C.; Bouras, C.; Gray, F.; Laplanche, J.-L.; Meurs, E.F.; Mouton-Liger, F.; Hugon, J. Increased levels of cerebrospinal fluid JNK3 associated with amyloid pathology: Links to cognitive decline. J. Psychiatry Neurosci. 2015, 40, 151–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takahashi, R.H.; Nagao, T.; Gouras, G.K. Plaque formation and the intraneuronal accumulation of β-amyloid in Alzheimer’s disease. Pathol. Int. 2017, 67, 185–193. [Google Scholar] [CrossRef] [PubMed]
- Chambers, J.W.; LoGrasso, P.V. Mitochondrial c-Jun N-terminal kinase (JNK) signaling initiates physiological changes resulting in amplification of reactive oxygen species generation. J. Biol. Chem. 2011, 286, 16052–16062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef]
- Zeke, A.; Misheva, M.; Reményi, A.; Bogoyevitch, M.A. JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol. Mol. Biol. Rev. 2016, 80, 793–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roskoski, R. Properties of FDA-approved small molecule protein kinase inhibitors: A 2020 update. Pharmacol. Res. 2020, 152, 104609. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, T.; Zulfiqar, A.; Arguelles, S.; Rasekhian, M.; Nabavi, S.F.; Silva, A.S.; Nabavi, S.M. Map kinase signaling as therapeutic target for neurodegeneration. Pharmacol. Res. 2020, 160, 105090. [Google Scholar] [CrossRef] [PubMed]
- Koch, P.; Gehringer, M.; Laufer, S.A. Inhibitors of c-Jun N-terminal kinases: An update. J. Med. Chem. 2015, 58, 72–95. [Google Scholar] [CrossRef] [PubMed]
- Hepp Rehfeldt, S.C.; Majolo, F.; Goettert, M.I.; Laufer, S. c-Jun N-Terminal Kinase Inhibitors as Potential Leads for New Therapeutics for Alzheimer’s Diseases. Int. J. Mol. Sci. 2020, 21, 9677. [Google Scholar] [CrossRef]
- Manning, G.; Whyte, D.B.; Martinez, R.; Hunter, T.; Sudarsanam, S. The protein kinase complement of the human genome. Science 2002, 298, 1912–1934. [Google Scholar] [CrossRef] [Green Version]
- Modi, V.; Dunbrack, R.L. A Structurally-Validated Multiple Sequence Alignment of 497 Human Protein Kinase Domains. Sci. Rep. 2019, 9, 19790. [Google Scholar] [CrossRef] [Green Version]
- Mehan, S.; Meena, H.; Sharma, D.; Sankhla, R. JNK: A stress-activated protein kinase therapeutic strategies and involvement in Alzheimer’s and various neurodegenerative abnormalities. J. Mol. Neurosci. 2011, 43, 376–390. [Google Scholar] [CrossRef]
- Zhang, T.; Inesta-Vaquera, F.; Niepel, M.; Zhang, J.; Ficarro, S.B.; Machleidt, T.; Xie, T.; Marto, J.A.; Kim, N.; Sim, T.; et al. Discovery of potent and selective covalent inhibitors of JNK. Chem. Biol. 2012, 19, 140–154. [Google Scholar] [CrossRef] [Green Version]
- Muth, F.; El-Gokha, A.; Ansideri, F.; Eitel, M.; Döring, E.; Sievers-Engler, A.; Lange, A.; Boeckler, F.M.; Lämmerhofer, M.; Koch, P.; et al. Tri- and Tetrasubstituted Pyridinylimidazoles as Covalent Inhibitors of c-Jun N-Terminal Kinase 3. J. Med. Chem. 2017, 60, 594–607. [Google Scholar] [CrossRef]
- Guo, T.; Zhang, D.; Zeng, Y.; Huang, T.Y.; Xu, H.; Zhao, Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol. Neurodegener. 2020, 15, 40. [Google Scholar] [CrossRef]
- Yarza, R.; Vela, S.; Solas, M.; Ramirez, M.J. c-Jun N-terminal Kinase (JNK) Signaling as a Therapeutic Target for Alzheimer’s Disease. Front. Pharmacol. 2015, 6, 321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoon, S.O.; Park, D.J.; Ryu, J.C.; Ozer, H.G.; Tep, C.; Shin, Y.J.; Lim, T.H.; Pastorino, L.; Kunwar, A.J.; Walton, J.C.; et al. JNK3 perpetuates metabolic stress induced by Aβ peptides. Neuron 2012, 75, 824–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yue, J.; López, J.M. Understanding MAPK signaling pathways in apoptosis. Int. J. Mol. Sci. 2020, 21, 2346. [Google Scholar] [CrossRef] [Green Version]
- Burkhard, K.; Shapiro, P. Use of inhibitors in the study of MAP kinases. Methods Mol. Biol. 2010, 661, 107–122. [Google Scholar] [CrossRef] [Green Version]
- Cuenda, A.; Rouse, J.; Doza, Y.N.; Meier, R.; Cohen, P.; Gallagher, T.F.; Young, P.R.; Lee, J.C. SB 203580 is a specific inhibitor of a MAP kinase homologue which is stimulated by cellular stresses and interleukin-1. FEBS Lett. 1995, 364, 229–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bennett, B.L.; Sasaki, D.T.; Murray, B.W.; O’Leary, E.C.; Sakata, S.T.; Xu, W.; Leisten, J.C.; Motiwala, A.; Pierce, S.; Satoh, Y.; et al. SP600125, an anthrapyrazolone inhibitor of Jun N-terminal kinase. Proc. Natl. Acad. Sci. USA 2001, 98, 13681–13686. [Google Scholar] [CrossRef] [Green Version]
- Moon, D.O.; Kim, M.O.; Kang, C.H.; Lee, J.D.; Choi, Y.H.; Kim, G.Y. JNK inhibitor SP600125 promotes the formation of polymerized tubulin, leading to G2/M phase arrest, endoreduplication, and delayed apoptosis. Exp. Mol. Med. 2009, 41, 665–677. [Google Scholar] [CrossRef] [Green Version]
- Inoue, C.; Sobue, S.; Aoyama, Y.; Mizutani, N.; Kawamoto, Y.; Nishizawa, Y.; Ichihara, M.; Abe, A.; Hayakawa, F.; Suzuki, M.; et al. BCL2 inhibitor ABT-199 and JNK inhibitor SP600125 exhibit synergistic cytotoxicity against imatinib-resistant Ph+ ALL cells. Biochem. Biophys. Rep. 2018, 15, 69–75. [Google Scholar] [CrossRef]
- Yang, C.; Pei, W.; Zhao, J.; Cheng, Y.; Zheng, X.; Rong, J. Bornyl caffeate induces apoptosis in human breast cancer MCF-7 cells via the ROS- and JNK-mediated pathways. Acta Pharmacol. Sin. 2014, 35, 113–123. [Google Scholar] [CrossRef] [Green Version]
- Tang, C.; Liang, J.; Qian, J.; Jin, L.; Du, M.; Li, M.; Li, D. Opposing role of JNK-p38 kinase and ERK1/2 in hydrogen peroxide-induced oxidative damage of human trophoblast-like JEG-3 cells. Int. J. Clin. Exp. Pathol. 2014, 7, 959–968. [Google Scholar]
- Xia, M.; Huang, R.; Witt, K.L.; Southall, N.; Fostel, J.; Cho, M.-H.; Jadhav, A.; Smith, C.S.; Inglese, J.; Portier, C.J.; et al. Compound cytotoxicity profiling using quantitative high-throughput screening. Environ. Health Perspect. 2008, 116, 284–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Soto-Otero, R.; Méndez-Alvarez, E.; Hermida-Ameijeiras, A.; Muñoz-Patiño, A.M.; Labandeira-Garcia, J.L. Autoxidation and neurotoxicity of 6-hydroxydopamine in the presence of some antioxidants: Potential implication in relation to the pathogenesis of Parkinson’s disease. J. Neurochem. 2000, 74, 1605–1612. [Google Scholar] [CrossRef]
- Halliwell, B.; Whiteman, M. Measuring reactive species and oxidative damage in vivo and in cell culture: How should you do it and what do the results mean? Br. J. Pharmacol. 2004, 142, 231–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halliwell, B. Oxidative stress in cell culture: An under-appreciated problem? FEBS Lett. 2003, 540, 3–6. [Google Scholar] [CrossRef] [Green Version]
- Goettert, M.; Luik, S.; Graeser, R.; Laufer, S.A. A direct ELISA assay for quantitative determination of the inhibitory potency of small molecules inhibitors for JNK3. J. Pharm. Biomed. Anal. 2011, 55, 236–240. [Google Scholar] [CrossRef] [PubMed]
- Allan, S.M.; Rothwell, N.J. Cytokines and acute neurodegeneration. Nat. Rev. Neurosci. 2001, 2, 734–744. [Google Scholar] [CrossRef] [PubMed]
- Afridi, R.; Lee, W.-H.; Suk, K. Microglia gone awry: Linking immunometabolism to neurodegeneration. Front. Cell. Neurosci. 2020, 14, 246. [Google Scholar] [CrossRef] [PubMed]
- Yu, T.; Li, Y.J.; Bian, A.H.; Zuo, H.B.; Zhu, T.W.; Ji, S.X.; Kong, F.; Yin, D.Q.; Wang, C.B.; Wang, Z.F.; et al. The regulatory role of activating transcription factor 2 in inflammation. Mediat. Inflamm. 2014, 2014, 950472. [Google Scholar] [CrossRef] [Green Version]
- Allan, S.M. The role of pro- and antiinflammatory cytokines in neurodegeneration. Ann. N. Y. Acad. Sci. 2000, 917, 84–93. [Google Scholar] [CrossRef]
- Azam, S.; Haque, M.E.; Kim, I.-S.; Choi, D.-K. Microglial Turnover in Ageing-Related Neurodegeneration: Therapeutic Avenue to Intervene in Disease Progression. Cells 2021, 10, 150. [Google Scholar] [CrossRef]
- Stenvinkel, P.; Ketteler, M.; Johnson, R.J.; Lindholm, B.; Pecoits-Filho, R.; Riella, M.; Heimbürger, O.; Cederholm, T.; Girndt, M. IL-10, IL-6, and TNF-alpha: Central factors in the altered cytokine network of uremia--the good, the bad, and the ugly. Kidney Int. 2005, 67, 1216–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Påhlman, S.; Ruusala, A.I.; Abrahamsson, L.; Mattsson, M.E.; Esscher, T. Retinoic acid-induced differentiation of cultured human neuroblastoma cells: A comparison with phorbolester-induced differentiation. Cell Differ. 1984, 14, 135–144. [Google Scholar] [CrossRef]
- Xie, H.; Hu, L.; Li, G. SH-SY5Y human neuroblastoma cell line: In vitro cell model of dopaminergic neurons in Parkinson’s disease. Chin. Med. J. 2010, 123, 1086–1092. [Google Scholar] [PubMed]
- Lopes, F.M.; Schröder, R.; da Frota, M.L.C.; Zanotto-Filho, A.; Müller, C.B.; Pires, A.S.; Meurer, R.T.; Colpo, G.D.; Gelain, D.P.; Kapczinski, F.; et al. Comparison between proliferative and neuron-like SH-SY5Y cells as an in vitro model for Parkinson disease studies. Brain Res. 2010, 1337, 85–94. [Google Scholar] [CrossRef] [PubMed]
- Glinka, Y.; Gassen, M.; Youdim, M.B. Mechanism of 6-hydroxydopamine neurotoxicity. J. Neural Transm. Suppl. 1997, 50, 55–66. [Google Scholar] [CrossRef]
- Cohen, G.; Heikkila, R.E. The generation of hydrogen peroxide, superoxide radical, and hydroxyl radical by 6-hydroxydopamine, dialuric acid, and related cytotoxic agents. J. Biol. Chem. 1974, 249, 2447–2452. [Google Scholar] [CrossRef]
- Glinka, Y.; Tipton, K.F.; Youdim, M.B. Mechanism of inhibition of mitochondrial respiratory complex I by 6-hydroxydopamine and its prevention by desferrioxamine. Eur. J. Pharmacol. 1998, 351, 121–129. [Google Scholar] [CrossRef]
- Glinka, Y.Y.; Youdim, M.B. Inhibition of mitochondrial complexes I and IV by 6-hydroxydopamine. Eur. J. Pharmacol. 1995, 292, 329–332. [Google Scholar] [CrossRef]
- Glinka, Y.; Tipton, K.F.; Youdim, M.B. Nature of inhibition of mitochondrial respiratory complex I by 6-Hydroxydopamine. J. Neurochem. 1996, 66, 2004–2010. [Google Scholar] [CrossRef]
- Sharma, L.K.; Lu, J.; Bai, Y. Mitochondrial respiratory complex I: Structure, function and implication in human diseases. Curr. Med. Chem. 2009, 16, 1266–1277. [Google Scholar] [CrossRef] [Green Version]
- Tysnes, O.-B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef] [PubMed]
- Norat, P.; Soldozy, S.; Sokolowski, J.D.; Gorick, C.M.; Kumar, J.S.; Chae, Y.; Yağmurlu, K.; Prada, F.; Walker, M.; Levitt, M.R.; et al. Mitochondrial dysfunction in neurological disorders: Exploring mitochondrial transplantation. NPJ Regen. Med. 2020, 5, 22. [Google Scholar] [CrossRef] [PubMed]
- Manczak, M.; Park, B.S.; Jung, Y.; Reddy, P.H. Differential expression of oxidative phosphorylation genes in patients with Alzheimer’s disease: Implications for early mitochondrial dysfunction and oxidative damage. Neuromol. Med. 2004, 5, 147–162. [Google Scholar] [CrossRef]
- Wang, W.; Zhao, F.; Ma, X.; Perry, G.; Zhu, X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: Recent advances. Mol. Neurodegener. 2020, 15, 30. [Google Scholar] [CrossRef]
- Du, H.; ShiDu Yan, S. Unlocking the Door to Neuronal Woes in Alzheimer’s Disease: Aβ and Mitochondrial Permeability Transition Pore. Pharmaceuticals 2010, 3, 1936–1948. [Google Scholar] [CrossRef] [Green Version]
- Parks, J.K.; Smith, T.S.; Trimmer, P.A.; Bennett, J.P.; Parker, W.D. Neurotoxic Abeta peptides increase oxidative stress in vivo through NMDA-receptor and nitric-oxide-synthase mechanisms, and inhibit complex IV activity and induce a mitochondrial permeability transition in vitro. J. Neurochem. 2001, 76, 1050–1056. [Google Scholar] [CrossRef]
- Hanawa, N.; Shinohara, M.; Saberi, B.; Gaarde, W.A.; Han, D.; Kaplowitz, N. Role of JNK translocation to mitochondria leading to inhibition of mitochondria bioenergetics in acetaminophen-induced liver injury. J. Biol. Chem. 2008, 283, 13565–13577. [Google Scholar] [CrossRef] [Green Version]
- Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef] [Green Version]
- Schroeter, H.; Boyd, C.S.; Ahmed, R.; Spencer, J.P.E.; Duncan, R.F.; Rice-Evans, C.; Cadenas, E. c-Jun N-terminal kinase (JNK)-mediated modulation of brain mitochondria function: New target proteins for JNK signalling in mitochondrion-dependent apoptosis. Biochem. J. 2003, 372, 359–369. [Google Scholar] [CrossRef] [Green Version]
- Melino, G.; Thiele, C.J.; Knight, R.A.; Piacentini, M. Retinoids and the control of growth/death decisions in human neuroblastoma cell lines. J. Neurooncol. 1997, 31, 65–83. [Google Scholar] [CrossRef]
- Cheung, Y.-T.; Lau, W.K.-W.; Yu, M.-S.; Lai, C.S.-W.; Yeung, S.-C.; So, K.-F.; Chang, R.C.-C. Effects of all-trans-retinoic acid on human SH-SY5Y neuroblastoma as in vitro model in neurotoxicity research. Neurotoxicology 2009, 30, 127–135. [Google Scholar] [CrossRef] [PubMed]
- Shipley, M.M.; Mangold, C.A.; Szpara, M.L. Differentiation of the SH-SY5Y Human Neuroblastoma Cell Line. J. Vis. Exp. 2016, 53193. [Google Scholar] [CrossRef] [PubMed]
- Presgraves, S.P.; Ahmed, T.; Borwege, S.; Joyce, J.N. Terminally differentiated SH-SY5Y cells provide a model system for studying neuroprotective effects of dopamine agonists. Neurotox. Res. 2004, 5, 579–598. [Google Scholar] [CrossRef]
- Fornari, F.A.; Randolph, J.K.; Yalowich, J.C.; Ritke, M.K.; Gewirtz, D.A. Interference by doxorubicin with DNA unwinding in MCF-7 breast tumor cells. Mol. Pharmacol. 1994, 45, 649–656. [Google Scholar] [PubMed]
- Chen, X.; Ji, Z.L.; Chen, Y.Z. TTD: Therapeutic target database. Nucleic Acids Res. 2002, 30, 412–415. [Google Scholar] [CrossRef] [Green Version]
- López-Carballo, G.; Moreno, L.; Masiá, S.; Pérez, P.; Barettino, D. Activation of the phosphatidylinositol 3-kinase/Akt signaling pathway by retinoic acid is required for neural differentiation of SH-SY5Y human neuroblastoma cells. J. Biol. Chem. 2002, 277, 25297–25304. [Google Scholar] [CrossRef] [Green Version]
- Itano, Y.; Ito, A.; Uehara, T.; Nomura, Y. Regulation of Bcl-2 protein expression in human neuroblastoma SH-SY5Y cells: Positive and negative effects of protein kinases C and A, respectively. J. Neurochem. 1996, 67, 131–137. [Google Scholar] [CrossRef]
- Vroegop, S.M.; Decker, D.E.; Buxser, S.E. Localization of damage induced by reactive oxygen species in cultured cells. Free Radic. Biol. Med. 1995, 18, 141–151. [Google Scholar] [CrossRef]
- Lee, Y.M.; He, W.; Liou, Y.-C. The redox language in neurodegenerative diseases: Oxidative post-translational modifications by hydrogen peroxide. Cell Death Dis. 2021, 12, 58. [Google Scholar] [CrossRef]
- Starkov, A.A. The role of mitochondria in reactive oxygen species metabolism and signaling. Ann. N. Y. Acad. Sci. 2008, 1147, 37–52. [Google Scholar] [CrossRef] [Green Version]
- Hamanaka, R.B.; Chandel, N.S. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem. Sci. 2010, 35, 505–513. [Google Scholar] [CrossRef] [Green Version]
- Whittemore, E.R.; Loo, D.T.; Watt, J.A.; Cotman, C.W. A detailed analysis of hydrogen peroxide-induced cell death in primary neuronal culture. Neuroscience 1995, 67, 921–932. [Google Scholar] [CrossRef]
- Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Butterfield, D.A.; Castegna, A.; Lauderback, C.M.; Drake, J. Evidence that amyloid beta-peptide-induced lipid peroxidation and its sequelae in Alzheimer’s disease brain contribute to neuronal death. Neurobiol. Aging 2002, 23, 655–664. [Google Scholar] [CrossRef]
- Dexter, D.T.; Carter, C.J.; Wells, F.R.; Javoy-Agid, F.; Agid, Y.; Lees, A.; Jenner, P.; Marsden, C.D. Basal lipid peroxidation in substantia nigra is increased in Parkinson’s disease. J. Neurochem. 1989, 52, 381–389. [Google Scholar] [CrossRef]
- Pedersen, W.A.; Fu, W.; Keller, J.N.; Markesbery, W.R.; Appel, S.; Smith, R.G.; Kasarskis, E.; Mattson, M.P. Protein modification by the lipid peroxidation product 4-hydroxynonenal in the spinal cords of amyotrophic lateral sclerosis patients. Ann. Neurol. 1998, 44, 819–824. [Google Scholar] [CrossRef]
- Zabel, M.; Nackenoff, A.; Kirsch, W.M.; Harrison, F.E.; Perry, G.; Schrag, M. Markers of oxidative damage to lipids, nucleic acids and proteins and antioxidant enzymes activities in Alzheimer’s disease brain: A meta-analysis in human pathological specimens. Free Radic. Biol. Med. 2018, 115, 351–360. [Google Scholar] [CrossRef]
- Andersen, J.K. Oxidative stress in neurodegeneration: Cause or consequence? Nat. Med. 2004, 10, S18–S25. [Google Scholar] [CrossRef]
- Polidori, M.C.; Nelles, G. Antioxidant clinical trials in mild cognitive impairment and Alzheimer’s disease—Challenges and perspectives. Curr. Pharm. Des. 2014, 20, 3083–3092. [Google Scholar] [CrossRef]
- Galasko, D.R.; Peskind, E.; Clark, C.M.; Quinn, J.F.; Ringman, J.M.; Jicha, G.A.; Cotman, C.; Cottrell, B.; Montine, T.J.; Thomas, R.G.; et al. Alzheimer’s Disease Cooperative Study Antioxidants for Alzheimer disease: A randomized clinical trial with cerebrospinal fluid biomarker measures. Arch. Neurol. 2012, 69, 836–841. [Google Scholar] [CrossRef] [Green Version]
- Kamenecka, T.; Jiang, R.; Song, X.; Duckett, D.; Chen, W.; Ling, Y.Y.; Habel, J.; Laughlin, J.D.; Chambers, J.; Figuera-Losada, M.; et al. Synthesis, biological evaluation, X-ray structure, and pharmacokinetics of aminopyrimidine c-jun-N-terminal kinase (JNK) inhibitors. J. Med. Chem. 2010, 53, 419–431. [Google Scholar] [CrossRef] [Green Version]
- Turrens, J.F. Mitochondrial formation of reactive oxygen species. J. Physiol. 2003, 552, 335–344. [Google Scholar] [CrossRef]
- Korshunov, S.S.; Skulachev, V.P.; Starkov, A.A. High protonic potential actuates a mechanism of production of reactive oxygen species in mitochondria. FEBS Lett. 1997, 416, 15–18. [Google Scholar] [CrossRef] [Green Version]
- Marchi, S.; Giorgi, C.; Suski, J.M.; Agnoletto, C.; Bononi, A.; Bonora, M.; De Marchi, E.; Missiroli, S.; Patergnani, S.; Poletti, F.; et al. Mitochondria-ros crosstalk in the control of cell death and aging. J. Signal Transduct. 2012, 2012, 329635. [Google Scholar] [CrossRef] [Green Version]
- Kim, I.; Rodriguez-Enriquez, S.; Lemasters, J.J. Selective degradation of mitochondria by mitophagy. Arch. Biochem. Biophys. 2007, 462, 245–253. [Google Scholar] [CrossRef] [Green Version]
- Connolly, N.M.C.; Theurey, P.; Adam-Vizi, V.; Bazan, N.G.; Bernardi, P.; Bolaños, J.P.; Culmsee, C.; Dawson, V.L.; Deshmukh, M.; Duchen, M.R.; et al. Guidelines on experimental methods to assess mitochondrial dysfunction in cellular models of neurodegenerative diseases. Cell Death Differ. 2018, 25, 542–572. [Google Scholar] [CrossRef] [Green Version]
- Heslop, K.A.; Rovini, A.; Hunt, E.G.; Fang, D.; Morris, M.E.; Christie, C.F.; Gooz, M.B.; DeHart, D.N.; Dang, Y.; Lemasters, J.J.; et al. JNK activation and translocation to mitochondria mediates mitochondrial dysfunction and cell death induced by VDAC opening and sorafenib in hepatocarcinoma cells. Biochem. Pharmacol. 2020, 171, 113728. [Google Scholar] [CrossRef]
- Lucero, M.; Suarez, A.E.; Chambers, J.W. Phosphoregulation on mitochondria: Integration of cell and organelle responses. CNS Neurosci. Ther. 2019, 25, 837–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shieh, J.-M.; Huang, T.-F.; Hung, C.-F.; Chou, K.-H.; Tsai, Y.-J.; Wu, W.-B. Activation of c-Jun N-terminal kinase is essential for mitochondrial membrane potential change and apoptosis induced by doxycycline in melanoma cells. Br. J. Pharmacol. 2010, 160, 1171–1184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Guo, S.-H.; Shang, X.-J.; Yu, L.-S.; Zhu, J.-W.; Zhao, A.; Zhou, Y.-F.; An, G.-H.; Zhang, Q.; Ma, B. Triptolide induces Sertoli cell apoptosis in mice via ROS/JNK-dependent activation of the mitochondrial pathway and inhibition of Nrf2-mediated antioxidant response. Acta Pharmacol. Sin. 2018, 39, 311–327. [Google Scholar] [CrossRef] [Green Version]
- Chauhan, D.; Li, G.; Hideshima, T.; Podar, K.; Mitsiades, C.; Mitsiades, N.; Munshi, N.; Kharbanda, S.; Anderson, K.C. JNK-dependent release of mitochondrial protein, Smac, during apoptosis in multiple myeloma (MM) cells. J. Biol. Chem. 2003, 278, 17593–17596. [Google Scholar] [CrossRef] [Green Version]
- Che, X.-F.; Moriya, S.; Zheng, C.-L.; Abe, A.; Tomoda, A.; Miyazawa, K. 2-Aminophenoxazine-3-one-induced apoptosis via generation of reactive oxygen species followed by c-jun N-terminal kinase activation in the human glioblastoma cell line LN229. Int. J. Oncol. 2013, 43, 1456–1466. [Google Scholar] [CrossRef] [Green Version]
- Fan, P.; Yu, X.-Y.; Xie, X.-H.; Chen, C.-H.; Zhang, P.; Yang, C.; Peng, X.; Wang, Y.-T. Mitophagy is a protective response against oxidative damage in bone marrow mesenchymal stem cells. Life Sci. 2019, 229, 36–45. [Google Scholar] [CrossRef]
- Kilbride, S.M.; Telford, J.E.; Davey, G.P. Complex I controls mitochondrial and plasma membrane potentials in nerve terminals. Neurochem. Res. 2021, 46, 100–107. [Google Scholar] [CrossRef]
- Rhein, V.; Baysang, G.; Rao, S.; Meier, F.; Bonert, A.; Müller-Spahn, F.; Eckert, A. Amyloid-beta leads to impaired cellular respiration, energy production and mitochondrial electron chain complex activities in human neuroblastoma cells. Cell. Mol. Neurobiol. 2009, 29, 1063–1071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wajant, H. The Fas signaling pathway: More than a paradigm. Science 2002, 296, 1635–1636. [Google Scholar] [CrossRef]
- Shi, Y.; Holtzman, D.M. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat. Rev. Immunol. 2018, 18, 759–772. [Google Scholar] [CrossRef]
- Gupta, S.; Barrett, T.; Whitmarsh, A.J.; Cavanagh, J.; Sluss, H.K.; Dérijard, B.; Davis, R.J. Selective interaction of JNK protein kinase isoforms with transcription factors. EMBO J. 1996, 15, 2760–2770. [Google Scholar] [CrossRef] [Green Version]
- Ventura, J.-J.; Cogswell, P.; Flavell, R.A.; Baldwin, A.S.; Davis, R.J. JNK potentiates TNF-stimulated necrosis by increasing the production of cytotoxic reactive oxygen species. Genes Dev. 2004, 18, 2905–2915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kamata, H.; Honda, S.-I.; Maeda, S.; Chang, L.; Hirata, H.; Karin, M. Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell 2005, 120, 649–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tong, W.; Chen, X.; Song, X.; Chen, Y.; Jia, R.; Zou, Y.; Li, L.; Yin, L.; He, C.; Liang, X.; et al. Resveratrol inhibits LPS-induced inflammation through suppressing the signaling cascades of TLR4-NF-κB/MAPKs/IRF3. Exp. Ther. Med. 2020, 19, 1824–1834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, L.; Song, Y.; Liu, Y.; Chen, Q.; Han, Q.; Chen, W.; Pan, T.; Zhang, Y.; Cao, X.; Wang, Q. MicroRNA-92a negatively regulates Toll-like receptor (TLR)-triggered inflammatory response in macrophages by targeting MKK4 kinase. J. Biol. Chem. 2013, 288, 7956–7967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yim, M.-J.; Lee, J.M.; Choi, G.; Lee, D.-S.; Park, W.S.; Jung, W.-K.; Park, S.; Seo, S.-K.; Park, J.; Choi, I.-W.; et al. Anti-Inflammatory Potential of Carpomitra costata Ethanolic Extracts via Inhibition of NF-κB and AP-1 Activation in LPS-Stimulated RAW264.7 Macrophages. Evid. Based Complement. Alternat. Med. 2018, 2018, 6914514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, J.-W.; Kwon, O.-K.; Ryu, H.W.; Paik, J.-H.; Paryanto, I.; Yuniato, P.; Choi, S.; Oh, S.-R.; Ahn, K.-S. Anti-inflammatory effects of Passiflora foetida L. in LPS-stimulated RAW264.7 macrophages. Int. J. Mol. Med. 2018, 41, 3709–3716. [Google Scholar] [CrossRef] [Green Version]
- Lim, M.X.; Png, C.W.; Tay, C.Y.B.; Teo, J.D.W.; Jiao, H.; Lehming, N.; Tan, K.S.W.; Zhang, Y. Differential regulation of proinflammatory cytokine expression by mitogen-activated protein kinases in macrophages in response to intestinal parasite infection. Infect. Immun. 2014, 82, 4789–4801. [Google Scholar] [CrossRef] [Green Version]
- Carlson, N.G.; Wieggel, W.A.; Chen, J.; Bacchi, A.; Rogers, S.W.; Gahring, L.C. Inflammatory cytokines IL-1 alpha, IL-1 beta, IL-6, and TNF-alpha impart neuroprotection to an excitotoxin through distinct pathways. J. Immunol. 1999, 163, 3963–3968. [Google Scholar]
- Yasukawa, H.; Ohishi, M.; Mori, H.; Murakami, M.; Chinen, T.; Aki, D.; Hanada, T.; Takeda, K.; Akira, S.; Hoshijima, M.; et al. IL-6 induces an anti-inflammatory response in the absence of SOCS3 in macrophages. Nat. Immunol. 2003, 4, 551–556. [Google Scholar] [CrossRef]
- Walker, D.G.; Whetzel, A.M.; Lue, L.F. Expression of suppressor of cytokine signaling genes in human elderly and Alzheimer’s disease brains and human microglia. Neuroscience 2015, 302, 121–137. [Google Scholar] [CrossRef] [Green Version]
- Wichmann, M.A.; Cruickshanks, K.J.; Carlsson, C.M.; Chappell, R.; Fischer, M.E.; Klein, B.E.K.; Klein, R.; Tsai, M.Y.; Schubert, C.R. Long-term systemic inflammation and cognitive impairment in a population-based cohort. J. Am. Geriatr. Soc. 2014, 62, 1683–1691. [Google Scholar] [CrossRef] [Green Version]
- Toulmond, S.; Vige, X.; Fage, D.; Benavides, J. Local infusion of interleukin-6 attenuates the neurotoxic effects of NMDA on rat striatal cholinergic neurons. Neurosci. Lett. 1992, 144, 49–52. [Google Scholar] [CrossRef]
- Yamada, M.; Hatanaka, H. Interleukin-6 protects cultured rat hippocampal neurons against glutamate-induced cell death. Brain Res. 1994, 643, 173–180. [Google Scholar] [CrossRef]
- Pizzi, M.; Sarnico, I.; Boroni, F.; Benarese, M.; Dreano, M.; Garotta, G.; Valerio, A.; Spano, P. Prevention of neuron and oligodendrocyte degeneration by interleukin-6 (IL-6) and IL-6 receptor/IL-6 fusion protein in organotypic hippocampal slices. Mol. Cell. Neurosci. 2004, 25, 301–311. [Google Scholar] [CrossRef]
- Erta, M.; Quintana, A.; Hidalgo, J. Interleukin-6, a major cytokine in the central nervous system. Int. J. Biol. Sci. 2012, 8, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
- Yang, D.D.; Kuan, C.Y.; Whitmarsh, A.J.; Rincón, M.; Zheng, T.S.; Davis, R.J.; Rakic, P.; Flavell, R.A. Absence of excitotoxicity-induced apoptosis in the hippocampus of mice lacking the Jnk3 gene. Nature 1997, 389, 865–870. [Google Scholar] [CrossRef] [PubMed]
- Hoque, A.; Williamson, N.A.; Ameen, S.S.; Ciccotosto, G.D.; Hossain, M.I.; Oakhill, J.S.; Ng, D.C.H.; Ang, C.-S.; Cheng, H.-C. Quantitative proteomic analyses of dynamic signalling events in cortical neurons undergoing excitotoxic cell death. Cell Death Dis. 2019, 10, 213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnson, J.W.; Kotermanski, S.E. Mechanism of action of memantine. Curr. Opin. Pharmacol. 2006, 6, 61–67. [Google Scholar] [CrossRef]
- Kim, B.-J.; Silverman, S.M.; Liu, Y.; Wordinger, R.J.; Pang, I.-H.; Clark, A.F. In vitro and in vivo neuroprotective effects of cJun N-terminal kinase inhibitors on retinal ganglion cells. Mol. Neurodegener. 2016, 11, 30. [Google Scholar] [CrossRef] [Green Version]
- Centeno, C.; Repici, M.; Chatton, J.Y.; Riederer, B.M.; Bonny, C.; Nicod, P.; Price, M.; Clarke, P.G.H.; Papa, S.; Franzoso, G.; et al. Role of the JNK pathway in NMDA-mediated excitotoxicity of cortical neurons. Cell Death Differ. 2007, 14, 240–253. [Google Scholar] [CrossRef]
- Marcelli, S.; Iannuzzi, F.; Ficulle, E.; Mango, D.; Pieraccini, S.; Pellegrino, S.; Corbo, M.; Sironi, M.; Pittaluga, A.; Nisticò, R.; et al. The selective disruption of presynaptic JNK2/STX1a interaction reduces NMDA receptor-dependent glutamate release. Sci. Rep. 2019, 9, 7146. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.-Q.; Peng, Y.-P.; Lu, J.-H.; Cao, B.-B.; Qiu, Y.-H. Neuroprotection of interleukin-6 against NMDA attack and its signal transduction by JAK and MAPK. Neurosci. Lett. 2009, 450, 122–126. [Google Scholar] [CrossRef]
- Jung, J.E.; Kim, G.S.; Chan, P.H. Neuroprotection by interleukin-6 is mediated by signal transducer and activator of transcription 3 and antioxidative signaling in ischemic stroke. Stroke 2011, 42, 3574–3579. [Google Scholar] [CrossRef] [Green Version]
- Knight, J.M.; Costanzo, E.S.; Singh, S.; Yin, Z.; Szabo, A.; Pawar, D.S.; Hillard, C.J.; Rizzo, J.D.; D’Souza, A.; Pasquini, M.; et al. The IL-6 antagonist tocilizumab is associated with worse depression and related symptoms in the medically ill. Transl. Psychiatry 2021, 11, 58. [Google Scholar] [CrossRef]
- Dafsari, F.S.; Jessen, F. Depression-an underrecognized target for prevention of dementia in Alzheimer’s disease. Transl. Psychiatry 2020, 10, 160. [Google Scholar] [CrossRef]
- Lim, G.Y.; Tam, W.W.; Lu, Y.; Ho, C.S.; Zhang, M.W.; Ho, R.C. Prevalence of Depression in the Community from 30 Countries between 1994 and 2014. Sci. Rep. 2018, 8, 2861. [Google Scholar] [CrossRef]
- Snowden, M.B.; Atkins, D.C.; Steinman, L.E.; Bell, J.F.; Bryant, L.L.; Copeland, C.; Fitzpatrick, A.L. Longitudinal association of dementia and depression. Am. J. Geriatr. Psychiatry 2015, 23, 897–905. [Google Scholar] [CrossRef] [Green Version]
- Gao, S.; Howard, S.; LoGrasso, P.V. Pharmacological Inhibition of c-Jun N-terminal Kinase Reduces Food Intake and Sensitizes Leptin’s Anorectic Signaling Actions. Sci. Rep. 2017, 7, 41795. [Google Scholar] [CrossRef] [Green Version]
- Ouyang, W.; O’Garra, A. IL-10 Family Cytokines IL-10 and IL-22: From Basic Science to Clinical Translation. Immunity 2019, 50, 871–891. [Google Scholar] [CrossRef] [PubMed]
- Mosser, D.M.; Zhang, X. Interleukin-10: New perspectives on an old cytokine. Immunol. Rev. 2008, 226, 205–218. [Google Scholar] [CrossRef] [PubMed]
- Wang, B.; Rao, Y.-H.; Inoue, M.; Hao, R.; Lai, C.-H.; Chen, D.; McDonald, S.L.; Choi, M.-C.; Wang, Q.; Shinohara, M.L.; et al. Microtubule acetylation amplifies p38 kinase signalling and anti-inflammatory IL-10 production. Nat. Commun. 2014, 5, 3479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guillot-Sestier, M.-V.; Doty, K.R.; Gate, D.; Rodriguez, J.; Leung, B.P.; Rezai-Zadeh, K.; Town, T. Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology. Neuron 2015, 85, 534–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chakrabarty, P.; Li, A.; Ceballos-Diaz, C.; Eddy, J.A.; Funk, C.C.; Moore, B.; DiNunno, N.; Rosario, A.M.; Cruz, P.E.; Verbeeck, C.; et al. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron 2015, 85, 519–533. [Google Scholar] [CrossRef] [Green Version]
- Loewenbrueck, K.F.; Tigno-Aranjuez, J.T.; Boehm, B.O.; Lehmann, P.V.; Tary-Lehmann, M. Th1 responses to beta-amyloid in young humans convert to regulatory IL-10 responses in Down syndrome and Alzheimer’s disease. Neurobiol. Aging 2010, 31, 1732–1742. [Google Scholar] [CrossRef]
- Lopes, K.O.; Sparks, D.L.; Streit, W.J. Microglial dystrophy in the aged and Alzheimer’s disease brain is associated with ferritin immunoreactivity. Glia 2008, 56, 1048–1060. [Google Scholar] [CrossRef]
- Ma, S.L.; Tang, N.L.S.; Lam, L.C.W.; Chiu, H.F.K. The association between promoter polymorphism of the interleukin-10 gene and Alzheimer’s disease. Neurobiol. Aging 2005, 26, 1005–1010. [Google Scholar] [CrossRef]
- Zheng, C.; Zhou, X.-W.; Wang, J.-Z. The dual roles of cytokines in Alzheimer’s disease: Update on interleukins, TNF-α, TGF-β and IFN-γ. Transl. Neurodegener. 2016, 5, 7. [Google Scholar] [CrossRef] [Green Version]
- Fiebich, B.L.; Batista, C.R.A.; Saliba, S.W.; Yousif, N.M.; de Oliveira, A.C.P. Role of microglia tlrs in neurodegeneration. Front. Cell. Neurosci. 2018, 12, 329. [Google Scholar] [CrossRef] [Green Version]
- Saraiva, M.; Christensen, J.R.; Tsytsykova, A.V.; Goldfeld, A.E.; Ley, S.C.; Kioussis, D.; O’Garra, A. Identification of a macrophage-specific chromatin signature in the IL-10 locus. J. Immunol. 2005, 175, 1041–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, W.; Lim, W.; Gee, K.; Aucoin, S.; Nandan, D.; Kozlowski, M.; Diaz-Mitoma, F.; Kumar, A. The p38 mitogen-activated kinase pathway regulates the human interleukin-10 promoter via the activation of Sp1 transcription factor in lipopolysaccharide-stimulated human macrophages. J. Biol. Chem. 2001, 276, 13664–13674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, C.; Sano, Y.; Todorova, K.; Carlson, B.A.; Arpa, L.; Celada, A.; Lawrence, T.; Otsu, K.; Brissette, J.L.; Arthur, J.S.C.; et al. The kinase p38 alpha serves cell type-specific inflammatory functions in skin injury and coordinates pro- and anti-inflammatory gene expression. Nat. Immunol. 2008, 9, 1019–1027. [Google Scholar] [CrossRef] [Green Version]
- Jarnicki, A.G.; Conroy, H.; Brereton, C.; Donnelly, G.; Toomey, D.; Walsh, K.; Sweeney, C.; Leavy, O.; Fletcher, J.; Lavelle, E.C.; et al. Attenuating regulatory T cell induction by TLR agonists through inhibition of p38 MAPK signaling in dendritic cells enhances their efficacy as vaccine adjuvants and cancer immunotherapeutics. J. Immunol. 2008, 180, 3797–3806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Foey, A.D.; Parry, S.L.; Williams, L.M.; Feldmann, M.; Foxwell, B.M.; Brennan, F.M. Regulation of monocyte IL-10 synthesis by endogenous IL-1 and TNF-alpha: Role of the p38 and p42/44 mitogen-activated protein kinases. J. Immunol. 1998, 160, 920–928. [Google Scholar] [PubMed]
- Chanteux, H.; Guisset, A.C.; Pilette, C.; Sibille, Y. LPS induces IL-10 production by human alveolar macrophages via MAPKinases- and Sp1-dependent mechanisms. Respir. Res. 2007, 8, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saraiva, M.; O’Garra, A. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 2010, 10, 170–181. [Google Scholar] [CrossRef] [Green Version]
- Lowry, O.H.; Rosebrough, N.J.; Farr, A.L.; Randall, R.J. Protein measurement with the Folin phenol reagent. J. Biol. Chem. 1951, 193, 265–275. [Google Scholar] [CrossRef]
- Pukac, L.A.; Carter, J.E.; Morrison, K.S.; Karnovsky, M.J. Enhancement of diaminobenzidine colorimetric signal in immunoblotting. BioTechniques 1997, 23, 385–388. [Google Scholar] [CrossRef] [PubMed]
- Janes, K.A. An analysis of critical factors for quantitative immunoblotting. Sci. Signal 2015, 8, rs2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silva, J.; Alves, C.; Martins, A.; Susano, P.; Simões, M.; Guedes, M.; Rehfeldt, S.; Pinteus, S.; Gaspar, H.; Rodrigues, A.; et al. Loliolide, a New Therapeutic Option for Neurological Diseases? In Vitro Neuroprotective and Anti-Inflammatory Activities of a Monoterpenoid Lactone Isolated from Codium tomentosum. Int. J. Mol. Sci. 2021, 22, 1888. [Google Scholar] [CrossRef] [PubMed]
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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Rehfeldt, S.C.H.; Laufer, S.; Goettert, M.I. A Highly Selective In Vitro JNK3 Inhibitor, FMU200, Restores Mitochondrial Membrane Potential and Reduces Oxidative Stress and Apoptosis in SH-SY5Y Cells. Int. J. Mol. Sci. 2021, 22, 3701. https://doi.org/10.3390/ijms22073701
Rehfeldt SCH, Laufer S, Goettert MI. A Highly Selective In Vitro JNK3 Inhibitor, FMU200, Restores Mitochondrial Membrane Potential and Reduces Oxidative Stress and Apoptosis in SH-SY5Y Cells. International Journal of Molecular Sciences. 2021; 22(7):3701. https://doi.org/10.3390/ijms22073701
Chicago/Turabian StyleRehfeldt, Stephanie Cristine Hepp, Stefan Laufer, and Márcia Inês Goettert. 2021. "A Highly Selective In Vitro JNK3 Inhibitor, FMU200, Restores Mitochondrial Membrane Potential and Reduces Oxidative Stress and Apoptosis in SH-SY5Y Cells" International Journal of Molecular Sciences 22, no. 7: 3701. https://doi.org/10.3390/ijms22073701
APA StyleRehfeldt, S. C. H., Laufer, S., & Goettert, M. I. (2021). A Highly Selective In Vitro JNK3 Inhibitor, FMU200, Restores Mitochondrial Membrane Potential and Reduces Oxidative Stress and Apoptosis in SH-SY5Y Cells. International Journal of Molecular Sciences, 22(7), 3701. https://doi.org/10.3390/ijms22073701