Brain Neurotrophins and Plant Polyphenols: A Powerful Connection
Abstract
:1. Introduction
2. Materials and Methods
3. Neurotrophins
4. Plant Polyphenols
5. Polyphenols and the Gut–Brain Axis
6. Brain and Plant Polyphenols
6.1. Epigenetic Modulation
6.2. Activation of Cellular Pathways
6.3. Mitigation of Neuroinflammation
6.4. Oxidative Stress Reduction
6.5. Model Studies
7. Polyphenols, Neurotrophins, and Addiction
7.1. Resveratrol
7.2. Curcumin
7.3. Green Tea Polyphenols
7.4. Olive Polyphenols
8. Discussion and Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
BBB | Blood–brain barrier |
BDNF | Brain-derived neurotrophic factor |
CREB | cAMP response element-binding protein |
DOPET | 3,4-dihydroxy-phenylethanol |
EGCG | Epigallocatechin gallate |
MAPK | Mitogen-activated protein kinase |
NGF | Nerve growth factor |
Nrf2 | Nuclear factor erythroid 2-related factor 2 |
NT-3 | Neurotrophin-3 |
NT-4/5 | Neurotrophin-4/5 |
p75NTR | p75 neurotrophin receptor |
SCFAs | Short-chain fatty acids |
SIRT1 | Sirtuin 1 |
SOD | Superoxide dismutase |
Trk | Tropomyosin receptor kinase |
WOS | Web of Science |
References
- Han, A.; Oster, R.; Yuen, H.; Jenkins, J.; Hawkins, J.; Edwards, L. Videoconference-Delivered Acceptance and Commitment Therapy for Family Caregivers of People with Dementia: Pilot Randomized Controlled Trial. JMIR Form. Res. 2025, 9, e67545. [Google Scholar] [CrossRef] [PubMed]
- Carlo, L.; Carlo, M.P.J.; O’Donnell, A.; Estrada, L.V. Technology-Based Mental Health Interventions in Dementia Care: A Systematic Review. J. Gerontol. Nurs. 2025, 51, 19–28. [Google Scholar] [CrossRef] [PubMed]
- van Husen, G.; Burger, T.J.; de Koning, M.B.; de Wit, M.A.S.; Segeren, M.W.; Beekman, A.T.F. Needs of the network: A qualitative study of the needs of family members, partners and close friends of people with a severe mental illness (SMI). BMC Psychiatry 2025, 25, 220. [Google Scholar] [CrossRef]
- Weerasinghe, T.; Dassanayake, R.; Senapathy, M.; Thennakoon, R.; Dayasiri, K. The role of primary caregivers’ knowledge, attitudes, and practices in paediatric medication safety. BMC Res. Notes 2025, 18, 94. [Google Scholar] [CrossRef] [PubMed]
- Teoh, Y.; Garg, P.; Karyadiguna, N.; Vivekanandarajah, S.; So, L.; Hurwitz, R.; Raman, S. Caregiver experiences of accessing a child developmental assessment service in a culturally diverse population in Australia: A mixed methods study. BMJ Paediatr. Open 2025, 9, e003003. [Google Scholar] [CrossRef]
- Scharf, A.; Michalowsky, B.; Rädke, A.; Kleinke, F.; Schade, S.; Platen, M.; Buchholz, M.; Pfaff, M.; Iskandar, A.; van den Berg, N.; et al. Identifying and Addressing Unmet Needs in Dementia: The Role of Care Access and Psychosocial Support. Int. J. Geriatr. Psychiatry 2025, 40, e70066. [Google Scholar] [CrossRef]
- Mao, J.; Yamakawa, M.; Hu, X.; Chikama, H.; Swa, T.; Takeya, Y. Negative Consequences of Sleep Deprivation Experienced by Informal Caregivers of People With Dementia on Caregivers and Care Recipients: A Scoping Review. Int. J. Nurs. Pract. 2025, 31, e70010. [Google Scholar] [CrossRef]
- Chen, T.H.; Ma, W.F. Exploring Socio-Economic Differences and Developer Medical Involvement of Dementia-Related English Version Mobile Health Applications. Int. J. Geriatr. Psychiatry 2025, 40, e70064. [Google Scholar] [CrossRef]
- Cappadona, I.; Ielo, A.; Pagano, M.; Anselmo, A.; Micali, G.; Giambò, F.M.; Duca, A.; D’Aleo, P.; Costanzo, D.; Carcione, G.; et al. Observational protocol on neuropsychological disorders in cardiovascular disease for holistic prevention and treatment. Future Cardiol. 2025, 21, 349–358. [Google Scholar] [CrossRef]
- Shen, W.C.; Lin, J.N.; Chan, S.H.; Yuh-Shiow, L.; Wang, J.J. Exploring Care Challenges and Needs of People with Diabetes Comorbid Cognitive Impairment from the Triangular Perspectives. Nurs. Health Sci. 2025, 27, e70081. [Google Scholar] [CrossRef]
- Bhalla, G.; Tanoto, P.; Vipin, A.; Chen, X.Y.J.; Leow, Y.J.; Chen, C.; Yap, P.L.K.; Merchant, R.A.; Hilal, S.; Ong, A.P.; et al. Current status and future directions for the diagnosis and management of mild cognitive impairment in Southeast Asia: A SEACURE consensus paper. J. Prev. Alzheimer’s Dis. 2025, 12, 100110. [Google Scholar] [CrossRef] [PubMed]
- Doerr, A.J.; Orwig, T.A.; McNulty, M.; Sison, S.D.M.; Paquette, D.R.; Leung, R.; Ding, H.; Erban, S.B.; Weinstein, B.R.; Guilarte-Walker, Y.; et al. Digital Assessment of Cognitive Health in Outpatient Primary Care: Usability Study. JMIR Form. Res. 2025, 9, e66695. [Google Scholar] [CrossRef] [PubMed]
- Yilmaz, B.; Erdogan, C.S.; Sandal, S.; Kelestimur, F.; Carpenter, D.O. Obesogens and Energy Homeostasis: Definition, Mechanisms of Action, Exposure, and Adverse Effects on Human Health. Neuroendocrinology 2025, 115, 72–100. [Google Scholar] [CrossRef] [PubMed]
- Zhukovsky, P.; Tio, E.S.; Coughlan, G.; Bennett, D.A.; Wang, Y.; Hohman, T.J.; Pizzagalli, D.A.; Mulsant, B.H.; Voineskos, A.N.; Felsky, D. Genetic influences on brain and cognitive health and their interactions with cardiovascular conditions and depression. Nat. Commun. 2024, 15, 5207. [Google Scholar] [CrossRef]
- Dudbridge, F.; Pashayan, N.; Yang, J. Predictive accuracy of combined genetic and environmental risk scores. Genet. Epidemiol. 2018, 42, 4–19. [Google Scholar] [CrossRef]
- Willemsen, G.; Vink, J.M.; Abdellaoui, A.; den Braber, A.; van Beek, J.H.D.A.; Draisma, H.H.M.; van Dongen, J.; van ’t Ent, D.; Geels, L.M.; van Lien, R.; et al. The Adult Netherlands Twin Register: Twenty-five years of survey and biological data collection. Twin Res. Hum. Genet. Off. J. Int. Soc. Twin Stud. 2013, 16, 271–281. [Google Scholar] [CrossRef]
- Chen, S.; Chen, S.; Hanewald, K.; Si, Y.; Bateman, H.; Li, B.; Xu, X.; Samtani, S.; Wu, C.; Brodaty, H. Social Environment, Lifestyle, and Genetic Predisposition with Dementia Risk: A Long-Term Longitudinal Study Among Older Adults. J. Gerontol. A. Biol. Sci. Med. Sci. 2024, 79, glae128. [Google Scholar] [CrossRef]
- Chermon, D.; Birk, R. Brain-derived neurotrophic factor gene rs925946 associates with Israeli females’ obesity predisposition: An interaction between genetics, eating habits, and physical inactivity. Nutr. Res. 2024, 125, 61–68. [Google Scholar] [CrossRef]
- van Dongen, J.; Willemsen, G.; de Geus, E.J.C.; Boomsma, D.I.; Neale, M.C. Effects of smoking on genome-wide DNA methylation profiles: A study of discordant and concordant monozygotic twin pairs. Elife 2023, 12, e83286. [Google Scholar] [CrossRef]
- Pérusse, L.; Jacob, R.; Drapeau, V.; Llewellyn, C.; Arsenault, B.J.; Bureau, A.; Labonté, M.-È.; Tremblay, A.; Vohl, M.-C. Understanding Gene-Lifestyle Interaction in Obesity: The Role of Mediation versus Moderation. Lifestyle Genom. 2022, 15, 67–76. [Google Scholar] [CrossRef]
- Barbu, M.C.; Shen, X.; Walker, R.M.; Howard, D.M.; Evans, K.L.; Whalley, H.C.; Porteous, D.J.; Morris, S.W.; Deary, I.J.; Zeng, Y.; et al. Epigenetic prediction of major depressive disorder. Mol. Psychiatry 2021, 26, 5112–5123. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; van der Werf, J.; Carson-Chahhoud, K.; Ni, G.; McGrath, J.; Hyppönen, E.; Lee, S.H. Whole-Genome Approach Discovers Novel Genetic and Nongenetic Variance Components Modulated by Lifestyle for Cardiovascular Health. J. Am. Heart Assoc. 2020, 9, e015661. [Google Scholar] [CrossRef] [PubMed]
- Gibson, J.; Russ, T.C.; Clarke, T.-K.; Howard, D.M.; Hillary, R.F.; Evans, K.L.; Walker, R.M.; Bermingham, M.L.; Morris, S.W.; Campbell, A.; et al. A meta-analysis of genome-wide association studies of epigenetic age acceleration. PLoS Genet. 2019, 15, e1008104. [Google Scholar] [CrossRef] [PubMed]
- Huat, T.J.; Camats-Perna, J.; Newcombe, E.A.; Valmas, N.; Kitazawa, M.; Medeiros, R. Metal Toxicity Links to Alzheimer’s Disease and Neuroinflammation. J. Mol. Biol. 2019, 431, 1843–1868. [Google Scholar] [CrossRef]
- Skaper, S.D. The neurotrophin family of neurotrophic factors: An overview. Methods Mol. Biol. 2012, 846, 1–12. [Google Scholar] [CrossRef]
- Numakawa, T.; Kajihara, R. The Role of Brain-Derived Neurotrophic Factor as an Essential Mediator in Neuronal Functions and the Therapeutic Potential of Its Mimetics for Neuroprotection in Neurologic and Psychiatric Disorders. Molecules 2025, 30, 848. [Google Scholar] [CrossRef]
- Carvalho, I.M.; Coelho, P.B.; Costa, P.C.; Marques, C.S.; Oliveira, R.S.; Ferreira, D.C. Current Neurogenic and Neuroprotective Strategies to Prevent and Treat Neurodegenerative and Neuropsychiatric Disorders. NeuroMolecular Med. 2015, 17, 404–422. [Google Scholar] [CrossRef]
- Lewin, G.R. Physiology of the Neurotrophins. Annu. Rev. Neurosci. 1996, 19, 289–317. [Google Scholar] [CrossRef]
- Harvey, T.; Rios, M. The Role of BDNF and TrkB in the Central Control of Energy and Glucose Balance: An Update. Biomolecules 2024, 14, 424. [Google Scholar] [CrossRef]
- Kim, J.; He, M.J.; Widmann, A.K.; Lee, F.S. The role of neurotrophic factors in novel, rapid psychiatric treatments. Neuropsychopharmacology 2024, 49, 227–245. [Google Scholar] [CrossRef]
- Abdolahi, S.; Zare-Chahoki, A.; Noorbakhsh, F.; Gorji, A. A Review of Molecular Interplay between Neurotrophins and miRNAs in Neuropsychological Disorders. Mol. Neurobiol. 2022, 59, 6260–6280. [Google Scholar] [CrossRef] [PubMed]
- Numakawa, T.; Odaka, H. Brain-derived neurotrophic factor signaling in the pathophysiology of alzheimer’s disease: Beneficial effects of flavonoids for neuroprotection. Int. J. Mol. Sci. 2021, 22, 5719. [Google Scholar] [CrossRef] [PubMed]
- Lippi, G.; Mattiuzzi, C.; Sanchis-Gomar, F. Updated overview on interplay between physical exercise, neurotrophins, and cognitive function in humans. J. Sport. Health Sci. 2020, 9, 74–81. [Google Scholar] [CrossRef] [PubMed]
- Kashyap, M.P.; Roberts, C.; Waseem, M.; Tyagi, P. Drug Targets in Neurotrophin Signaling in the Central and Peripheral Nervous System. Mol. Neurobiol. 2018, 55, 6939–6955. [Google Scholar] [CrossRef]
- Tang, T.; Li, Y.; Jiao, Q.; Du, X.; Jiang, H. Cerebral Dopamine Neurotrophic Factor: A Potential Therapeutic Agent for Parkinson’s Disease. Neurosci. Bull. 2017, 33, 568–575. [Google Scholar] [CrossRef]
- Mitre, M.; Mariga, A.; Chao, M.V. Neurotrophin signalling: Novel insights into mechanisms and pathophysiology. Clin. Sci. 2017, 131, 13–23. [Google Scholar] [CrossRef]
- Ciafrè, S.; Ferraguti, G.; Tirassa, P.; Iannitelli, A.; Ralli, M.; Greco, A.; Chaldakov, G.N.; Rosso, P.; Fico, E.; Messina, M.P.; et al. Nerve growth factor in the psychiatric brain. Riv. Psichiatr. 2020, 55, 4–15. [Google Scholar] [CrossRef]
- Frim, D.M.; Uhler, T.A.; Short, M.P.; Ezzedine, Z.D.; Klagsbrun, M.; Breakefield, X.O.; Isacson, O. Effects of biologically delivered NGF, bdnf and BFGF on striatal excitotoxic lesions. Neuroreport 1993, 4, 367–370. [Google Scholar] [CrossRef]
- Alizadeh Pahlavani, H. Possible role of exercise therapy on depression: Effector neurotransmitters as key players. Behav. Brain Res. 2024, 459, 114791. [Google Scholar] [CrossRef]
- Scuri, M.; Samsell, L.; Piedimonte, G. The role of neurotrophins in inflammation and allergy. Inflamm. Allergy Drug Targets 2010, 9, 173–180. [Google Scholar] [CrossRef]
- Rochlitzer, S.; Nassenstein, C.; Braun, A. The contribution of neurotrophins to the pathogenesis of allergic asthma. Biochem. Soc. Trans. 2006, 34, 594–599. [Google Scholar] [CrossRef] [PubMed]
- Chao, M.Y.; Rajagopal, R.; Lee, F.S. Neurotrophin signalling in health and disease. Clin. Sci. 2006, 110, 167–173. [Google Scholar] [CrossRef] [PubMed]
- Ricci, A.; Felici, L.; Mariotta, S.; Mannino, F.; Schmid, G.; Terzano, C.; Cardillo, G.; Amenta, F.; Bronzetti, E. Neurotrophin and Neurotrophin Receptor Protein Expression in the Human Lung. Am. J. Respir. Cell Mol. Biol. 2004, 30, 12–19. [Google Scholar] [CrossRef] [PubMed]
- Schulte-Herbruggen, O.; Braun, A.; Rochlitzer, S.; Jockers-Scherubl, M.C.; Hellweg, R. Neurotrophic factors--a tool for therapeutic strategies in neurological, neuropsychiatric and neuroimmunological diseases? Curr. Med. Chem. 2007, 14, 2318–2329. [Google Scholar] [CrossRef]
- Angelucci, F.; Mathé, A.A.; Aloe, L. Neurotrophic factors and CNS disorders: Findings in rodent models of depression and schizophrenia. Prog. Brain Res. 2004, 146, 151–165. [Google Scholar] [CrossRef]
- Martinotti, G.; Di Iorio, G.; Marini, S.; Ricci, V.; De Berardis, D.; Di Giannantonio, M. Nerve growth factor and brain-derived neurotrophic factor concentrations in schizophrenia: A review. J. Biol. Regul. Homeost. Agents 2012, 26, 347–356. [Google Scholar]
- Costa, S.L.; Silva, V.D.A.; dos Santos Souza, C.; Santos, C.C.; Paris, I.; Muñoz, P.; Segura-Aguilar, J. Impact of Plant-Derived Flavonoids on Neurodegenerative Diseases. Neurotox. Res. 2016, 30, 41–52. [Google Scholar] [CrossRef]
- Hussain, G.; Huang, J.; Rasul, A.; Anwar, H.; Imran, A.; Maqbool, J.; Razzaq, A.; Aziz, N.; Makhdoom, E.U.H.; Konuk, M.; et al. Putative roles of plant-derived tannins in neurodegenerative and neuropsychiatry disorders: An updated review. Molecules 2019, 24, 2213. [Google Scholar] [CrossRef]
- Mohsenpour, H.; Pesce, M.; Patruno, A.; Bahrami, A.; Pour, P.M.; Farzaei, M.H. A review of plant extracts and plant-derived natural compounds in the prevention/treatment of neonatal hypoxic-ischemic brain injury. Int. J. Mol. Sci. 2021, 22, 833. [Google Scholar] [CrossRef]
- Fakhri, S.; Abbaszadeh, F.; Moradi, S.Z.; Cao, H.; Khan, H.; Xiao, J. Effects of Polyphenols on Oxidative Stress, Inflammation, and Interconnected Pathways during Spinal Cord Injury. Oxidative Med. Cell. Longev. 2022, 2022, 8100195. [Google Scholar] [CrossRef]
- Tayab, M.A.; Islam, M.N.; Chowdhury, K.A.A.; Tasnim, F.M. Targeting neuroinflammation by polyphenols: A promising therapeutic approach against inflammation-associated depression. Biomed. Pharmacother. 2022, 147, 112668. [Google Scholar] [CrossRef]
- Davinelli, S.; Medoro, A.; Ali, S.; Passarella, D.; Intrieri, M.; Scapagnini, G. Dietary Flavonoids and Adult Neurogenesis: Potential Implications for Brain Aging. Curr. Neuropharmacol. 2022, 21, 651–668. [Google Scholar] [CrossRef] [PubMed]
- Shi, R.; Gao, D.; Stoika, R.; Liu, K.; Sik, A.; Jin, M. Potential implications of polyphenolic compounds in neurodegenerative diseases. Crit. Rev. Food Sci. Nutr. 2024, 64, 5491–5514. [Google Scholar] [CrossRef] [PubMed]
- Grabska-Kobyłecka, I.; Szpakowski, P.; Król, A.; Książek-Winiarek, D.; Kobyłecki, A.; Głąbiński, A.; Nowak, D. Polyphenols and Their Impact on the Prevention of Neurodegenerative Diseases and Development. Nutrients 2023, 15, 3454. [Google Scholar] [CrossRef]
- Roy, D.; Kaur, P.; Ghosh, M.; Choudhary, D.; Rangra, N.K. The therapeutic potential of typical plant-derived compounds for the management of metabolic disorders. Phyther. Res. 2024, 38, 3986–4008. [Google Scholar] [CrossRef] [PubMed]
- Moise, G.; Jîjie, A.R.; Moacă, E.A.; Predescu, I.A.; Dehelean, C.A.; Hegheș, A.; Vlad, D.C.; Popescu, R.; Vlad, C.S. Plants’ Impact on the Human Brain—Exploring the Neuroprotective and Neurotoxic Potential of Plants. Pharmaceuticals 2024, 17, 1339. [Google Scholar] [CrossRef]
- Miguel, C.A.; Noya-Riobó, M.V.; Mazzone, G.L.; Villar, M.J.; Coronel, M.F. Antioxidant, anti-inflammatory and neuroprotective actions of resveratrol after experimental nervous system insults. Special focus on the molecular mechanisms involved. Neurochem. Int. 2021, 150, 105188. [Google Scholar] [CrossRef]
- Goyal, S.; Seth, B.; Chaturvedi, R.K. Polyphenols and Stem Cells for Neuroregeneration in Parkinson’s Disease and Amyotrophic Lateral Sclerosis. Curr. Pharm. Des. 2021, 28, 806–828. [Google Scholar] [CrossRef]
- Özduran, G.; Becer, E.; Vatansever, H.S. The Role and Mechanisms of Action of Catechins in Neurodegenerative Diseases. J. Am. Nutr. Assoc. 2023, 42, 67–74. [Google Scholar] [CrossRef]
- Alesci, A.; Nicosia, N.; Fumia, A.; Giorgianni, F.; Santini, A.; Cicero, N. Resveratrol and Immune Cells: A Link to Improve Human Health. Molecules 2022, 27, 424. [Google Scholar] [CrossRef]
- Nor, N.A.M.; Budin, S.B.; Zainalabidin, S.; Jalil, J.; Sapian, S.; Jubaidi, F.F.; Anuar, N.N.M. The Role of Polyphenol in Modulating Associated Genes in Diabetes-Induced Vascular Disorders. Int. J. Mol. Sci. 2022, 23, 6396. [Google Scholar] [CrossRef]
- Zhang, W.; Dong, X.; Huang, R. Antiparkinsonian effects of polyphenols: A narrative review with a focus on the modulation of the gut-brain axis. Pharmacol. Res. 2023, 193, 106787. [Google Scholar] [CrossRef] [PubMed]
- Bhusal, C.K.; Uti, D.E.; Mukherjee, D.; Alqahtani, T.; Alqahtani, S.; Bhattacharya, A.; Akash, S. Unveiling Nature’s potential: Promising natural compounds in Parkinson’s disease management. Park. Relat. Disord. 2023, 115, 105799. [Google Scholar] [CrossRef] [PubMed]
- Gong, G.; Ganesan, K.; Wan, Y.; Liu, Y.; Huang, Y.; Luo, Y.; Wang, X.; Zhang, Z.; Zheng, Y. Unveiling the neuroprotective properties of isoflavones: Current evidence, molecular mechanisms and future perspectives. Crit. Rev. Food Sci. Nutr. 2024, 65, 3112–3148. [Google Scholar] [CrossRef]
- Carito, V.; Ceccanti, M.; Tarani, L.; Ferraguti, G.; NChaldakov, G.; Fiore, M. Neurotrophins’ Modulation by Olive Polyphenols. Curr. Med. Chem. 2016, 23, 3189–3197. [Google Scholar] [CrossRef]
- Carito, V.; Ceccanti, M.; Ferraguti, G.; Coccurello, R.; Ciafrè, S.; Tirassa, P.; Fiore, M. NGF and BDNF Alterations by Prenatal Alcohol Exposure. Curr. Neuropharmacol. 2019, 17, 308–317. [Google Scholar] [CrossRef]
- Tarani, L.; Carito, V.; Ferraguti, G.; Petrella, C.; Greco, A.; Ralli, M.; Messina, M.P.; Rasio, D.; De Luca, E.; Putotto, C.; et al. Neuroinflammatory Markers in the Serum of Prepubertal Children with down Syndrome. J. Immunol. Res. 2020, 2020, 6937154. [Google Scholar] [CrossRef]
- Ferraguti, G.; Terracina, S.; Tarani, L.; Fanfarillo, F.; Allushi, S.; Caronti, B.; Tirassa, P.; Polimeni, A.; Lucarelli, M.; Cavalcanti, L.; et al. Nerve Growth Factor and the Role of Inflammation in Tumor Development. Curr. Issues Mol. Biol. 2024, 46, 965–989. [Google Scholar] [CrossRef]
- Pardon, M.C. Role of Neurotrophic Factors in Behavioral Processes: Implications for the Treatment of Psychiatric and Neurodegenerative Disorders. Vitam. Horm. 2010, 82, 185–200. [Google Scholar] [CrossRef]
- Fujii, T.; Kunugi, H. p75NTR as a Therapeutic Target for Neuropsychiatric Diseases. Curr. Mol. Pharmacol. 2010, 2, 70–76. [Google Scholar] [CrossRef]
- Retamales-Ortega, R.; Oróstica, L.; Vera, C.; Cuevas, P.; Hernández, A.; Hurtado, I.; Vega, M.; Romero, C. Role of nerve growth factor (NGF) and miRNAs in epithelial ovarian cancer. Int. J. Mol. Sci. 2017, 18, 507. [Google Scholar] [CrossRef] [PubMed]
- Meeker, R.B.; Williams, K.S. The p75 neurotrophin receptor: At the crossroad of neural repair and death. Neural Regen. Res. 2015, 10, 721–725. [Google Scholar] [CrossRef] [PubMed]
- Kozlov, E.M.; Grechko, A.V.; Chegodaev, Y.S.; Wu, W.K.; Orekhov, A.N. Contribution of neurotrophins to the immune system regulation and possible connection to alcohol addiction. Biology 2020, 9, 63. [Google Scholar] [CrossRef]
- West, A.E.; Pruunsild, P.; Timmusk, T. Neurotrophins: Transcription and translation. Handb. Exp. Pharmacol. 2014, 220, 67–100. [Google Scholar] [CrossRef] [PubMed]
- Tonchev, A.B. Brain ischemia, neurogenesis, and neurotrophic receptor expression in primates. Arch. Ital. Biol. 2011, 149, 225–231. [Google Scholar] [CrossRef]
- Cirulli, F.; Alleva, E. The NGF saga: From animal models of psychosocial stress to stress-related psychopathology. Front. Neuroendocrinol. 2009, 30, 379–395. [Google Scholar] [CrossRef]
- Fields, J.; Dumaop, W.; Langford, T.D.; Rockenstein, E.; Masliah, E. Role of neurotrophic factor alterations in the neurodegenerative process in HIV associated neurocognitive disorders. J. Neuroimmune Pharmacol. 2014, 9, 102–116. [Google Scholar] [CrossRef]
- Von Bartheld, C.S. Neurotrophins in the developing and regenerating visual system. Histol. Histopathol. 1998, 13, 437–459. [Google Scholar]
- Huang, E.J.; Reichardt, L.F. Trk Receptors: Roles in Neuronal Signal Transduction. Annu. Rev. Biochem. 2003, 72, 609–642. [Google Scholar] [CrossRef]
- Leßmann, V. Neurotrophin-dependent modulation of glutamatergic synaptic transmission in the mammalian CNS. Gen. Pharmacol. 1998, 31, 667–674. [Google Scholar] [CrossRef]
- Meis, S.; Endres, T.; Lessmann, V. Neurotrophin signalling in amygdala-dependent cued fear learning. Cell Tissue Res. 2020, 382, 161–172. [Google Scholar] [CrossRef] [PubMed]
- Fernandez, G.M.; Stewart, W.N.; Savage, L.M. Chronic drinking during adolescence predisposes the adult rat for continued heavy drinking: Neurotrophin and behavioral adaptation after long-term, continuous ethanol exposure. PLoS ONE 2016, 11, e0149987. [Google Scholar] [CrossRef] [PubMed]
- Aloe, L.; Iannitelli, A.; Angelucci, F.; Bersani, G.; Fiore, M. Studies in animal models and humans suggesting a role of nerve growth factor in schizophrenia-like disorders. Behav. Pharmacol. 2000, 11, 235–242. [Google Scholar] [CrossRef] [PubMed]
- Bersani, G.; Iannitelli, A.; Fiore, M.; Angelucci, F.; Aloe, L. Data and hypotheses on the role of nerve growth factor and other neurotrophins in psychiatric disorders. Med. Hypotheses 2000, 55, 199–207. [Google Scholar] [CrossRef]
- Aloe, L.; Rocco, M.L.; Bianchi, P.; Manni, L. Nerve growth factor: From the early discoveries to the potential clinical use. J. Transl. Med. 2012, 10, 239. [Google Scholar] [CrossRef]
- Dechant, G.; Neumann, H. Neurotrophins. Adv. Exp. Med. Biol. 2002, 513, 303–334. [Google Scholar] [CrossRef]
- Longo, F.M.; Massa, S.M. Neurotrophin-based strategies for neuroprotection. J. Alzheimers Dis. 2004, 6, S13–S17. [Google Scholar] [CrossRef]
- Fiore, M.; Amendola, T.; Triaca, V.; Tirassa, P.; Alleva, E.; Aloe, L. Agonistic encounters in aged male mouse potentiate the expression of endogenous brain NGF and BDNF: Possible implication for brain progenitor cells’ activation. Eur. J. Neurosci. 2003, 17, 1455–1464. [Google Scholar] [CrossRef]
- Mooney, S.M.; Miller, M.W. Nerve growth factor neuroprotection of ethanol-induced neuronal death in rat cerebral cortex is age dependent. Neuroscience 2007, 149, 372–381. [Google Scholar] [CrossRef]
- Ebadi, M.; Bashir, R.M.; Heidrick, M.L.; Hamada, F.M.; Refaey, H.E.L.; Hamed, A.; Helal, G.; Baxi, M.D.; Cerutis, D.R.; Lassi, N.K. Neurotrophins and their receptors in nerve injury and repair. Neurochem. Int. 1997, 30, 347–374. [Google Scholar] [CrossRef]
- Ichim, G.; Tauszig-Delamasure, S.; Mehlen, P. Neurotrophins and cell death. Exp. Cell Res. 2012, 318, 1221–1228. [Google Scholar] [CrossRef] [PubMed]
- Meakin, S.O.; Shooter, E.M. The nerve growth factor family of receptors. Trends Neurosci. 1992, 15, 323–331. [Google Scholar] [CrossRef] [PubMed]
- Chen, E.; Mufson, E.J.; Kordower, J.H. TRK and p75 Neurotrophin Receptor Systems in the Developing Human Brain. J. Comp. Neurol. 1996, 618, 591–618. [Google Scholar] [CrossRef]
- Cacialli, P. Neurotrophins time point intervention after traumatic brain injury: From zebrafish to human. Int. J. Mol. Sci. 2021, 22, 1585. [Google Scholar] [CrossRef]
- Nakagawara, A. Trk receptor tyrosine kinases: A bridge between cancer and neural development. Cancer Lett. 2001, 169, 107–114. [Google Scholar] [CrossRef]
- Reichardt, L.F. Neurotrophin-regulated signalling pathways. Philos. Trans. R. Soc. B Biol. Sci. 2006, 361, 1545–1564. [Google Scholar] [CrossRef]
- French, S.J.; Humby, T.; Horner, C.H.; Sofroniew, M.V.; Rattray, M. Hippocampal neurotrophin and trk receptor mRNA levels are altered by local administration of nicotine, carbachol and pilocarpine. Mol. Brain Res. 1999, 67, 124–136. [Google Scholar] [CrossRef]
- Molloy, N.H.; Read, D.E.; Gorman, A.M. Nerve growth factor in cancer cell death and survival. Cancers 2011, 3, 510–530. [Google Scholar] [CrossRef]
- Nguyen, N.; Lee, S.B.; Lee, Y.S.; Lee, K.H.; Ahn, J.Y. Neuroprotection by NGF and BDNF against neurotoxin-exerted apoptotic death in neural stem cells are mediated through Trk receptors, activating PI3-kinase and MAPK pathways. Neurochem. Res. 2009, 34, 942–951. [Google Scholar] [CrossRef]
- Braunger, B.M.; Demmer, C.; Tamm, E.R. Programmed cell death during retinal development of the mouse eye. Adv. Exp. Med. Biol. 2014, 801, 9–13. [Google Scholar] [CrossRef]
- Chakravarthy, R.; Mnich, K.; Gorman, A.M. Nerve growth factor (NGF)-mediated regulation of p75NTR expression contributes to chemotherapeutic resistance in triple negative breast cancer cells. Biochem. Biophys. Res. Commun. 2016, 478, 1541–1547. [Google Scholar] [CrossRef] [PubMed]
- Yan, C.; Liang, Y.; Nylander, K.D.; Wong, J.; Rudavsky, R.M.; Saragovi, H.U.; Schor, N.F. p75-nerve growth factor as an antiapoptotic complex: Independence versus cooperativity in protection from enediyne chemotherapeutic agents. Mol. Pharmacol. 2002, 61, 710–719. [Google Scholar] [CrossRef] [PubMed]
- Lu, B.; Pang, P.T.; Woo, N.H. The yin and yang of neurotrophin action. Nat. Rev. Neurosci. 2005, 6, 603–614. [Google Scholar] [CrossRef] [PubMed]
- Björkholm, C.; Monteggia, L.M. BDNF—A key transducer of antidepressant effects. Neuropharmacology 2016, 102, 72–79. [Google Scholar] [CrossRef]
- Ryu, S.; Liu, X.; Guo, T.; Guo, Z.; Zhang, J.; Cao, Y.Q. Peripheral CCL2-CCR2 signalling contributes to chronic headache-related sensitization. Brain 2023, 146, 4274–4291. [Google Scholar] [CrossRef]
- Zhang, H.L.; Sun, Y.; Wu, Z.J.; Yin, Y.; Liu, R.Y.; Zhang, J.C.; Zhang, Z.J.; Yau, S.Y.; Wu, H.X.; Yuan, T.F.; et al. Hippocampal PACAP signaling activation triggers a rapid antidepressant response. Mil. Med. Res. 2024, 11, 49. [Google Scholar] [CrossRef]
- Contreras, E.; Bolívar, S.; Navarro, X.; Udina, E. New insights into peripheral nerve regeneration: The role of secretomes. Exp. Neurol. 2022, 354, 114069. [Google Scholar] [CrossRef]
- Osborne, A.; Khatib, T.Z.; Songra, L.; Barber, A.C.; Hall, K.; Kong, G.Y.X.; Widdowson, P.S.; Martin, K.R. Neuroprotection of retinal ganglion cells by a novel gene therapy construct that achieves sustained enhancement of brain-derived neurotrophic factor/tropomyosin-related kinase receptor-B signaling. Cell Death Dis. 2018, 9, 1007. [Google Scholar] [CrossRef]
- Peplow, P.V.; Baxter, G.D. Gene expression and release of growth factors during delayed wound healing: A review of studies in diabetic animals and possible combined laser phototherapy and growth factor treatment to enhance healing. Photomed. Laser Surg. 2012, 30, 617–636. [Google Scholar] [CrossRef]
- Hou, C.H.; Chen, W.L.; Lin, C.Y. Targeting nerve growth factor-mediated osteosarcoma metastasis: Mechanistic insights and therapeutic opportunities using larotrectinib. Cell Death Dis. 2024, 15, 381. [Google Scholar] [CrossRef]
- Manni, L.; Aloe, L.; Fiore, M. Changes in cognition induced by social isolation in the mouse are restored by electro-acupuncture. Physiol. Behav. 2009, 98, 537–542. [Google Scholar] [CrossRef] [PubMed]
- Wei, K.; Yang, J.; Lin, S.; Mei, Y.; An, N.; Cao, X.; Jiang, L.; Liu, C.; Li, C. Dietary Habits Modify the Association of Physical Exercise with Cognitive Impairment in Community-Dwelling Older Adults. J. Clin. Med. 2022, 11, 5122. [Google Scholar] [CrossRef] [PubMed]
- Barati, S.; Fabrizio, C.; Strafella, C.; Cascella, R.; Caputo, V.; Megalizzi, D.; Peconi, C.; Mela, J.; Colantoni, L.; Caltagirone, C.; et al. Relationship between Nutrition, Lifestyle, and Neurodegenerative Disease: Lessons from ADH1B, CYP1A2 and MTHFR. Genes 2022, 13, 1498. [Google Scholar] [CrossRef] [PubMed]
- Faurot, K.R.; Cole, W.R.; MacIntosh, B.A.; Dunlap, M.; Moore, C.B.; Roberson, B.; Guerra, M.; Domenichiello, A.F.; Palsson, O.; Rivera, W.; et al. Targeted dietary interventions to reduce pain in persistent post-traumatic headache among service members: Protocol for a randomized, controlled parallel-group trial. Contemp. Clin. Trials 2022, 119, 106851. [Google Scholar] [CrossRef]
- Driver, S.; McShan, E.; Swank, C.; Grobe, K.; Calhoun, S.; Bailey, R.; Kramer, K. Creating an appropriate adaptation of a healthy lifestyle intervention for people after stroke. Brain Inj. 2020, 34, 1497–1503. [Google Scholar] [CrossRef]
- Durany, N.; Thome, J. Neurotrophic factors and the pathophysiology of schizophrenic psychoses. Eur. Psychiatry 2004, 19, 326–337. [Google Scholar] [CrossRef]
- Jiang, C.; Salton, S.R. The role of neurotrophins in major depressive disorder. Transl. Neurosci. 2013, 4, 46–58. [Google Scholar] [CrossRef]
- Fukuyama, Y.; Kubo, M.; Harada, K. The search for, and chemistry and mechanism of, neurotrophic natural products. J. Nat. Med. 2020, 74, 648–671. [Google Scholar] [CrossRef]
- Memberg, S.P.; Hall, A.K. Proliferation, differentiation, and survival of rat sensory neuron precursors in vitro require specific trophic factors. Mol. Cell. Neurosci. 1995, 6, 323–335. [Google Scholar] [CrossRef]
- Alvarez-Dolado, M.; Iglesias, T.; Rodríguez-Peña, A.; Bernal, J.; Muñoz, A. Expression of neurotrophins and the trk family of neurotrophin receptors in normal and hypothyroid rat brain. Mol. Brain Res. 1994, 27, 249–257. [Google Scholar] [CrossRef]
- Fiore, M.; Grace, A.A.; Korf, J.; Stampachiacchiere, B.; Aloe, L. Impaired brain development in the rat following prenatal exposure to methylazoxymethanol acetate at gestational day 17 and neurotrophin distribution. Neuroreport 2004, 15, 1791–1795. [Google Scholar] [CrossRef]
- Valvassori, S.S.; Mariot, E.; Varela, R.B.; Bavaresco, D.V.; Dal-Pont, G.C.; Ferreira, C.L.; Andersen, M.L.; Tye, S.J.; Quevedo, J. The role of neurotrophic factors in manic-, anxious- and depressive-like behaviors induced by amphetamine sensitization: Implications to the animal model of bipolar disorder. J. Affect. Disord. 2019, 245, 1106–1113. [Google Scholar] [CrossRef] [PubMed]
- Ceccanti, M.; Mancinelli, R.; Tirassa, P.; Laviola, G.; Rossi, S.; Romeo, M.; Fiore, M. Early exposure to ethanol or red wine and long-lasting effects in aged mice. A study on nerve growth factor, brain-derived neurotrophic factor, hepatocyte growth factor, and vascular endothelial growth factor. Neurobiol. Aging 2012, 33, 359–367. [Google Scholar] [CrossRef] [PubMed]
- Van der Zee, C.E.E.M.; Rashid, K.; Le, K.; Moore, K.A.; Stanisz, J.; Diamond, J.; Racine, R.J.; Fahnestock, M. Intraventricular administration of antibodies to nerve growth factor retards kindling and blocks mossy fiber sprouting in adult rats. J. Neurosci. 1995, 15, 5316–5323. [Google Scholar] [CrossRef] [PubMed]
- Angelucci, F.; Mathé, A.A.; Aloe, L. Brain-derived neurotrophic factor and tyrosine kinase receptor TrkB in rat brain are significantly altered after haloperidol and risperidone administration. J. Neurosci. Res. 2000, 60, 783–794. [Google Scholar] [CrossRef]
- Bimonte-Nelson, H.A.; Hunter, C.L.; Nelson, M.E.; Granholm, A.C. Frontal cortex BDNF levels correlate with working memory in an animal model of Down syndrome. Behav. Brain Res. 2003, 139, 47–57. [Google Scholar] [CrossRef]
- Ferraguti, G.; Terracina, S.; Micangeli, G.; Lucarelli, M.; Tarani, L.; Ceccanti, M.; Spaziani, M.; D’Orazi, V.; Petrella, C.; Fiore, M. NGF and BDNF in pediatrics syndromes. Neurosci. Biobehav. Rev. 2023, 145, 105015. [Google Scholar] [CrossRef]
- Ciafrè, S.; Ferraguti, G.; Greco, A.; Polimeni, A.; Ralli, M.; Ceci, F.M.; Ceccanti, M.; Fiore, M. Alcohol as an early life stressor: Epigenetics, metabolic, neuroendocrine and neurobehavioral implications. Neurosci. Biobehav. Rev. 2020, 118, 654–668. [Google Scholar] [CrossRef]
- Manni, L.; Fausto, V.; Fiore, M.; Aloe, L. Repeated Restraint and Nerve Growth Factor Administration in Male and Female Mice: Effect on Sympathetic and Cardiovascular Mediators of the Stress Response. Curr. Neurovasc. Res. 2008, 5, 1–12. [Google Scholar] [CrossRef]
- Aloe, L.; Alleva, E.; Fiore, M. Stress and nerve growth factor: Findings in animal models and humans. Pharmacol. Biochem. Behav. 2002, 73, 159–166. [Google Scholar] [CrossRef]
- Ceci, F.M.; Ferraguti, G.; Petrella, C.; Greco, A.; Tirassa, P.; Iannitelli, A.; Ralli, M.; Vitali, M.; Ceccanti, M.; Chaldakov, G.N.; et al. Nerve Growth Factor, Stress and Diseases. Curr. Med. Chem. 2020, 28, 2943–2959. [Google Scholar] [CrossRef] [PubMed]
- Di Liegro, C.M.; Schiera, G.; Proia, P.; Di Liegro, I. Physical activity and brain health. Genes 2019, 10, 720. [Google Scholar] [CrossRef] [PubMed]
- Sanaeifar, F.; Pourranjbar, S.; Pourranjbar, M.; Ramezani, S.; Mehr, S.R.; Wadan, A.H.S.; Khazeifard, F. Beneficial effects of physical exercise on cognitive-behavioral impairments and brain-derived neurotrophic factor alteration in the limbic system induced by neurodegeneration. Exp. Gerontol. 2024, 195, 112539. [Google Scholar] [CrossRef] [PubMed]
- Almeida, M.F.; Chaves, R.S.; Silva, C.M.; Chaves, J.C.S.; Melo, K.P.; Ferrari, M.F.R. BDNF trafficking and signaling impairment during early neurodegeneration is prevented by moderate physical activity. IBRO Rep. 2016, 1, 19–31. [Google Scholar] [CrossRef]
- Fiore, M.; Talamini, L.; Angelucci, F.; Koch, T.; Aloe, L.; Korf, J. Prenatal methylazoxymethanol acetate alters behavior and brain NGF levels in young rats: A possible correlation with the development of schizophrenia-like deficits. Neuropharmacology 1999, 38, 857–869. [Google Scholar] [CrossRef]
- Weinstock, M. Prenatal stressors in rodents: Effects on behavior. Neurobiol. Stress 2017, 6, 3–13. [Google Scholar] [CrossRef]
- De Nicoló, S.; Tarani, L.; Ceccanti, M.; Maldini, M.; Natella, F.; Vania, A.; Chaldakov, G.N.; Fiore, M. Effects of olive polyphenols administration on nerve growth factor and brain-derived neurotrophic factor in the mouse brain. Nutrition 2013, 29, 681–687. [Google Scholar] [CrossRef]
- Moosavi, F.; Hosseini, R.; Saso, L.; Firuzi, O. Modulation of neurotrophic signaling pathways by polyphenols. Drug Des. Devel. Ther. 2015, 10, 23–42. [Google Scholar] [CrossRef]
- Ziegler, T.; Tsiountsioura, M.; Meixner-Goetz, L.; Cvirn, G.; Lamprecht, M. Polyphenols’ Impact on Selected Biomarkers of Brain Aging in Healthy Middle-Aged and Elderly Subjects: A Review of Clinical Trials. Nutrients 2023, 15, 3770. [Google Scholar] [CrossRef]
- Kopalli, S.R.; Behl, T.; Kyada, A.; Rekha, M.M.; Kundlas, M.; Rani, P.; Nathiya, D.; Satyam Naidu, K.; Gulati, M.; Bhise, M.; et al. Synaptic plasticity and neuroprotection: The molecular impact of flavonoids on neurodegenerative disease progression. Neuroscience 2025, 569, 161–183. [Google Scholar] [CrossRef]
- An, J.; Chen, B.; Tian, D.; Guo, Y.; Yan, Y.; Yang, H. Regulation of Neurogenesis and Neuronal Differentiation by Natural Compounds. Curr. Stem Cell Res. Ther. 2022, 17, 756–771. [Google Scholar] [CrossRef] [PubMed]
- Nicoletti, V.G.; Pajer, K.; Calcagno, D.; Pajenda, G.; Nógrádi, A. The Role of Metals in the Neuroregenerative Action of BDNF, GDNF, NGF and Other Neurotrophic Factors. Biomolecules 2022, 12, 1015. [Google Scholar] [CrossRef] [PubMed]
- Mitra, S.; Behbahani, H.; Eriksdotter, M. Innovative therapy for Alzheimer’s disease-with focus on biodelivery of NGF. Front. Neurosci. 2019, 13, 38. [Google Scholar] [CrossRef] [PubMed]
- Stepanichev, M.; Onufriev, M.; Aniol, V.; Freiman, S.; Brandstaetter, H.; Winter, S.; Lazareva, N.; Guekht, A.; Gulyaeva, N. Effects of cerebrolysin on nerve growth factor system in the aging rat brain. Restor. Neurol. Neurosci. 2017, 35, 571–581. [Google Scholar] [CrossRef]
- Forlenza, O.V.; Miranda, A.S.; Barbosa, I.G.; Talib, L.L.; Diniz, B.S.; Gattaz, W.F.; Teixeira, A.L. Decreased neurotrophic support is associated with cognitive decline in non-demented subjects. J. Alzheimer’s Dis. 2015, 46, 423–429. [Google Scholar] [CrossRef]
- Sable, P.; Kale, A.; Joshi, A.; Joshi, S. Maternal micronutrient imbalance alters gene expression of BDNF, NGF, TrkB and CREB in the offspring brain at an adult age. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 2014, 34, 24–32. [Google Scholar] [CrossRef]
- Riccio, A.; Ahn, S.; Davenport, C.M.; Blendy, J.A.; Ginty, D.D. Mediation by a CREB family transcription factor of NGF-dependent survival of sympathetic neurons. Science 1999, 286, 2358–2361. [Google Scholar] [CrossRef]
- Cheng, A.; Hou, Y.; Mattson, M.P. Mitochondria and neuroplasticity. ASN Neuro 2010, 2, e00045. [Google Scholar] [CrossRef]
- Almeida, S.; Cunha-Oliveira, T.; Laço, M.; Oliveira, C.R.; Rego, A.C. Dysregulation of CREB activation and histone acetylation in 3-nitropropionic acid-treated cortical neurons: Prevention by BDNF and NGF. Neurotox. Res. 2010, 17, 399–405. [Google Scholar] [CrossRef]
- Fiore, M.; Petrella, C.; Coriale, G.; Rosso, P.; Fico, E.; Ralli, M.; Greco, A.; De Vincentiis, M.; Minni, A.; Polimeni, A.; et al. Markers of Neuroinflammation in the Serum of Prepubertal Children with Fetal Alcohol Spectrum Disorders. CNS Neurol. Disord. Drug Targets-CNS Neurol. Disord. 2022, 21, 854–868. [Google Scholar] [CrossRef]
- Pahlavani, H.A. Exercise therapy to prevent and treat Alzheimer’s disease. Front. Aging Neurosci. 2023, 15, 1243869. [Google Scholar] [CrossRef] [PubMed]
- Gong, A.G.W.; Wang, H.Y.; Dong, T.T.X.; Tsim, K.W.K.; Zheng, Y.Z. Danggui Buxue Tang, a simple Chinese formula containing Astragali Radix and Angelicae Sinensis Radix, stimulates the expressions of neurotrophic factors in cultured SH-SY5Y cells. Chin. Med. 2017, 12, 24. [Google Scholar] [CrossRef] [PubMed]
- Primiani, C.T.; Ryan, V.H.; Rao, J.S.; Cam, M.C.; Ahn, K.; Modi, H.R.; Rapoport, S.I. Coordinated gene expression of neuroinflammatory and cell signaling markers in dorsolateral prefrontal cortex during human brain development and aging. PLoS ONE 2014, 9, e110972. [Google Scholar] [CrossRef]
- Griñán-Ferré, C.; Marsal-García, L.; Bellver-Sanchis, A.; Kondengaden, S.M.; Turga, R.C.; Vázquez, S.; Pallàs, M. Pharmacological inhibition of G9a/GLP restores cognition and reduces oxidative stress, neuroinflammation and ß-Amyloid plaques in an early-onset Alzheimer’s disease mouse model. Aging 2019, 11, 11591–11608. [Google Scholar] [CrossRef]
- Liu, J.; Wang, Y.; Guo, J.; Sun, J.; Sun, Q. Salvianolic Acid B improves cognitive impairment by inhibiting neuroinflammation and decreasing Aß level in Porphyromonas gingivalis-infected mice. Aging 2020, 12, 10117–10128. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Shi, X.; Ohm, M.; Korte, M.; Zagrebelsky, M. Deciphering genetic causality between plasma BDNF and 91 circulating inflammatory proteins through bidirectional mendelian randomization. Sci. Rep. 2025, 15, 10312. [Google Scholar] [CrossRef]
- Braschi, C.; Capsoni, S.; Narducci, R.; Poli, A.; Sansevero, G.; Brandi, R.; Maffei, L.; Cattaneo, A.; Berardi, N. Intranasal delivery of BDNF rescues memory deficits in AD11 mice and reduces brain microgliosis. Aging Clin. Exp. Res. 2021, 33, 1223–1238. [Google Scholar] [CrossRef]
- Wang, Z.H.; Xiang, J.; Liu, X.; Yu, S.P.; Manfredsson, F.P.; Sandoval, I.M.; Wu, S.; Wang, J.Z.; Ye, K. Deficiency in BDNF/TrkB Neurotrophic Activity Stimulates δ-Secretase by Upregulating C/EBPβ in Alzheimer’s Disease. Cell Rep. 2019, 28, 655–669.e5. [Google Scholar] [CrossRef]
- Wu, C.C.; Lien, C.C.; Hou, W.H.; Chiang, P.M.; Tsai, K.J. Gain of BDNF Function in Engrafted Neural Stem Cells Promotes the Therapeutic Potential for Alzheimer’s Disease. Sci. Rep. 2016, 6, 27358. [Google Scholar] [CrossRef]
- Turnbull, M.T.; Boskovic, Z.; Coulson, E.J. Acute down-regulation of BDNF signaling does not replicate exacerbated amyloid-β levels and cognitive impairment induced by cholinergic basal forebrain lesion. Front. Mol. Neurosci. 2018, 11, 51. [Google Scholar] [CrossRef]
- Xu, J.; Kurup, P.; Azkona, G.; Baguley, T.D.; Saavedra, A.; Nairn, A.C.; Ellman, J.A.; Pérez-Navarro, E.; Lombroso, P.J. Down-regulation of BDNF in cell and animal models increases striatal-enriched protein tyrosine phosphatase 61 (STEP61) levels. J. Neurochem. 2016, 136, 285–294. [Google Scholar] [CrossRef] [PubMed]
- Velazquez, R.; Ferreira, E.; Tran, A.; Turner, E.C.; Belfiore, R.; Branca, C.; Oddo, S. Acute tau knockdown in the hippocampus of adult mice causes learning and memory deficits. Aging Cell 2018, 17, e12775. [Google Scholar] [CrossRef] [PubMed]
- Sartor, G.C.; Malvezzi, A.M.; Kumar, A.; Andrade, N.S.; Wiedner, H.J.; Vilca, S.J.; Janczura, K.J.; Bagheri, A.; Al-Ali, H.; Powell, S.K.; et al. Enhancement of BDNF expression and memory by HDAC inhibition requires BET bromodomain reader proteins. J. Neurosci. 2019, 39, 612–626. [Google Scholar] [CrossRef] [PubMed]
- Zhu, G.; Sun, X.; Yang, Y.; Du, Y.; Lin, Y.; Zhou, N.; Xiang, J. Reduction of BDNF Results in GABAergic Neuroplasticity Dysfunction and Contributes to Late-Life Anxiety Disorder. Behav. Neurosci. 2019, 133, 212–224. [Google Scholar] [CrossRef]
- Wang, L.; Mao, Y.; Lu, Y.; Yuan, Y.; Jin, Y. Knockdown of lncRNA BDNF-AS alleviates isoflurane-induced neuro-inflammation and cognitive dysfunction through modulating miR-214-3p. Folia Neuropathol. 2023, 61, 68–76. [Google Scholar] [CrossRef]
- Carrillo, J.Á.; Arcusa, R.; Xandri-Martínez, R.; Cerdá, B.; Zafrilla, P.; Marhuenda, J. Impact of Polyphenol-Rich Nutraceuticals on Cognitive Function and Neuroprotective Biomarkers: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2025, 17, 601. [Google Scholar] [CrossRef]
- Puhlmann, L.M.C.; Vrtička, P.; Linz, R.; Valk, S.L.; Papassotiriou, I.; Chrousos, G.P.; Engert, V.; Singer, T. Serum BDNF Increase After 9-Month Contemplative Mental Training Is Associated with Decreased Cortisol Secretion and Increased Dentate Gyrus Volume: Evidence from a Randomized Clinical Trial. Biol. Psychiatry Glob. Open Sci. 2025, 5, 100414. [Google Scholar] [CrossRef]
- Kim, N.; Parolin, B.; Renshaw, D.; Deb, S.K.; Zariwala, M.G. Formulated Palmitoylethanolamide Supplementation Improves Parameters of Cognitive Function and BDNF Levels in Young, Healthy Adults: A Randomised Cross-Over Trial. Nutrients 2024, 16, 489. [Google Scholar] [CrossRef]
- Schneider, E.; Doll, J.P.K.; Schweinfurth, N.; Kettelhack, C.; Schaub, A.C.; Yamanbaeva, G.; Varghese, N.; Mählmann, L.; Brand, S.; Eckert, A.; et al. Effect of short-term, high-dose probiotic supplementation on cognition, related brain functions and BDNF in patients with depression: A secondary analysis of a randomized controlled trial. J. Psychiatry Neurosci. 2023, 48, E23–E33. [Google Scholar] [CrossRef]
- Hsu, Y.C.; Huang, Y.Y.; Tsai, S.Y.; Kuo, Y.W.; Lin, J.H.; Ho, H.H.; Chen, J.F.; Hsia, K.C.; Sun, Y. Efficacy of Probiotic Supplements on Brain-Derived Neurotrophic Factor, Inflammatory Biomarkers, Oxidative Stress and Cognitive Function in Patients with Alzheimer’s Dementia: A 12-Week Randomized, Double-Blind Active-Controlled Study. Nutrients 2024, 16, 16. [Google Scholar] [CrossRef]
- Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef] [PubMed]
- Wan, M.L.Y.; Co, V.A.; El-Nezami, H. Dietary polyphenol impact on gut health and microbiota. Crit. Rev. Food Sci. Nutr. 2021, 61, 690–711. [Google Scholar] [CrossRef] [PubMed]
- Shen, N.; Wang, T.; Gan, Q.; Liu, S.; Wang, L.; Jin, B. Plant flavonoids: Classification, distribution, biosynthesis, and antioxidant activity. Food Chem. 2022, 383, 132531. [Google Scholar] [CrossRef] [PubMed]
- Iqbal, I.; Wilairatana, P.; Saqib, F.; Nasir, B.; Wahid, M.; Latif, M.F.; Iqbal, A.; Naz, R.; Mubarak, M.S. Plant Polyphenols and Their Potential Benefits on Cardiovascular Health: A Review. Molecules 2023, 28, 6403. [Google Scholar] [CrossRef]
- Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols: The role of metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef]
- Rana, A.; Samtiya, M.; Dhewa, T.; Mishra, V.; Aluko, R.E. Health benefits of polyphenols: A concise review. J. Food Biochem. 2022, 46, e14264. [Google Scholar] [CrossRef]
- Truzzi, F.; Tibaldi, C.; Zhang, Y.; Dinelli, G.; DAmen, E. An Overview on Dietary Polyphenols and Their Biopharmaceutical Classification System (BCS). Int. J. Mol. Sci. 2021, 22, 5514. [Google Scholar] [CrossRef]
- Zhang, L.; Han, Z.; Granato, D. Polyphenols in foods: Classification, methods of identification, and nutritional aspects in human health. Adv. Food Nutr. Res. 2021, 98, 1–33. [Google Scholar] [CrossRef]
- Ciupei, D.; Colişar, A.; Leopold, L.; Stănilă, A.; Diaconeasa, Z.M. Polyphenols: From Classification to Therapeutic Potential and Bioavailability. Foods 2024, 13, 4131. [Google Scholar] [CrossRef]
- Chong, J.R.; de Lucia, C.; Tovar-Rios, D.A.; Castellanos-Perilla, N.; Collins, C.; Kvernberg, S.M.; Ballard, C.; Siow, R.C.; Aarsland, D. A Randomised, Double-Blind, Placebo-Controlled, Cross-Over Clinical Trial to Evaluate the Biological Effects and Safety of a Polyphenol Supplement on Healthy Ageing. Antioxidants 2024, 13, 995. [Google Scholar] [CrossRef]
- Godos, J.; Caraci, F.; Castellano, S.; Currenti, W.; Galvano, F.; Ferri, R.; Grosso, G. Association between dietary flavonoids intake and cognitive function in an Italian cohort. Biomolecules 2020, 10, 1300. [Google Scholar] [CrossRef] [PubMed]
- Gorzynik-Debicka, M.; Przychodzen, P.; Cappello, F.; Kuban-Jankowska, A.; Gammazza, A.M.; Knap, N.; Wozniak, M.; Gorska-Ponikowska, M. Potential health benefits of olive oil and plant polyphenols. Int. J. Mol. Sci. 2018, 19, 686. [Google Scholar] [CrossRef] [PubMed]
- Abd El-Hack, M.E.; de Oliveira, M.C.; Attia, Y.A.; Kamal, M.; Almohmadi, N.H.; Youssef, I.M.; Khalifa, N.E.; Moustafa, M.; Al-Shehri, M.; Taha, A.E. The efficacy of polyphenols as an antioxidant agent: An updated review. Int. J. Biol. Macromol. 2023, 250, 126525. [Google Scholar] [CrossRef]
- Zhou, Y.; Zhang, S.; Fan, X. Role of Polyphenols as Antioxidant Supplementation in Ischemic Stroke. Oxid. Med. Cell. Longev. 2021, 2021, 5471347. [Google Scholar] [CrossRef] [PubMed]
- Panickar, K.S.; Anderson, R.A. Effect of polyphenols on oxidative stress and mitochondrial dysfunction in neuronal death and brain edema in cerebral ischemia. Int. J. Mol. Sci. 2011, 12, 8181–8207. [Google Scholar] [CrossRef]
- Zhou, Z.D.; Xie, S.P.; Saw, W.T.; Ho, P.G.H.; Wang, H.; Lei, Z.; Yi, Z.; Tan, E.K. The therapeutic implications of tea polyphenols against dopamine (DA) neuron degeneration in parkinson’s disease (PD). Cells 2019, 8, 911. [Google Scholar] [CrossRef]
- Cracco, P.; Montalesi, E.; Parente, M.; Cipolletti, M.; Iucci, G.; Battocchio, C.; Venditti, I.; Fiocchetti, M.; Marino, M. A Novel Resveratrol-Induced Pathway Increases Neuron-Derived Cell Resilience against Oxidative Stress. Int. J. Mol. Sci. 2023, 24, 5903. [Google Scholar] [CrossRef]
- Kung, H.C.; Lin, K.J.; Kung, C.T.; Lin, T.K. Oxidative stress, mitochondrial dysfunction, and neuroprotection of polyphenols with respect to resveratrol in parkinson’s disease. Biomedicines 2021, 9, 918. [Google Scholar] [CrossRef]
- Naoi, M.; Wu, Y.; Shamoto-Nagai, M.; Maruyama, W. Mitochondria in Neuroprotection by Phytochemicals: Bioactive Polyphenols Modulate Mitochondrial Apoptosis System, Function and Structure. Int. J. Mol. Sci. 2019, 20, 2451. [Google Scholar] [CrossRef]
- Zhou, Y.; Jiang, Z.; Lu, H.; Xu, Z.; Tong, R.; Shi, J.; Jia, G. Recent Advances of Natural Polyphenols Activators for Keap1-Nrf2 Signaling Pathway. Chem. Biodivers. 2019, 16, e1900400. [Google Scholar] [CrossRef]
- Nabavi, S.F.; Barber, A.J.; Spagnuolo, C.; Russo, G.L.; Daglia, M.; Nabavi, S.M.; Sobarzo-Sánchez, E. Nrf2 as molecular target for polyphenols: A novel therapeutic strategy in diabetic retinopathy. Crit. Rev. Clin. Lab. Sci. 2016, 53, 293–312. [Google Scholar] [CrossRef] [PubMed]
- Tascioglu Aliyev, A.; Panieri, E.; Stepanić, V.; Gurer-Orhan, H.; Saso, L. Involvement of NRF2 in Breast Cancer and Possible Therapeutical Role of Polyphenols and Melatonin. Molecules 2021, 26, 1853. [Google Scholar] [CrossRef] [PubMed]
- Plauth, A.; Geikowski, A.; Cichon, S.; Wowro, S.J.; Liedgens, L.; Rousseau, M.; Weidner, C.; Fuhr, L.; Kliem, M.; Jenkins, G.; et al. Hormetic shifting of redox environment by pro-oxidative resveratrol protects cells against stress. Free Radic. Biol. Med. 2016, 99, 608–622. [Google Scholar] [CrossRef] [PubMed]
- Figueira, I.; Menezes, R.; Macedo, D.; Costa, I.; dos Santos, C.N. Polyphenols Beyond Barriers: A Glimpse into the Brain. Curr. Neuropharmacol. 2016, 15, 562–594. [Google Scholar] [CrossRef]
- Gundimeda, U.; McNeill, T.H.; Schiffman, J.E.; Hinton, D.R.; Gopalakrishna, R. Green tea polyphenols potentiate the action of nerve growth factor to induce neuritogenesis: Possible role of reactive oxygen species. J. Neurosci. Res. 2010, 88, 3644–3655. [Google Scholar] [CrossRef]
- Bensalem, J.; Dudonne, S.; Gaudout, D.; Servant, L.; Calon, F.; Desjardins, Y.; Laye, S.; Lafenetre, P.; Pallet, V. Polyphenol-rich extract from grape and blueberry attenuates cognitive decline and improves neuronal function in aged mice. J. Nutr. Sci. 2018, 7, e19. [Google Scholar] [CrossRef]
- Fiore, M.; Messina, M.P.; Petrella, C.; D’Angelo, A.; Greco, A.; Ralli, M.; Ferraguti, G.; Tarani, L.; Vitali, M.; Ceccanti, M. Antioxidant properties of plant polyphenols in the counteraction of alcohol-abuse induced damage: Impact on the Mediterranean diet. J. Funct. Foods 2020, 71, 104012. [Google Scholar] [CrossRef]
- Bucio-Noble, D.; Kautto, L.; Krisp, C.; Ball, M.S.; Molloy, M.P. Polyphenol extracts from dried sugarcane inhibit inflammatory mediators in an in vitro colon cancer model. J. Proteom. 2018, 177, 1–10. [Google Scholar] [CrossRef]
- Pallauf, K.; Giller, K.; Huebbe, P.; Rimbach, G. Nutrition and healthy ageing: Calorie restriction or polyphenol-rich “mediterrAsian” diet? Oxidative Med. Cell. Longev. 2013, 2013, 707421. [Google Scholar] [CrossRef]
- Ajmo, J.M.; Liang, X.; Rogers, C.Q.; Pennock, B.; You, M. Resveratrol alleviates alcoholic fatty liver in mice. AJP Gastrointest. Liver Physiol. 2008, 295, G833–G842. [Google Scholar] [CrossRef]
- Han, S.; Choi, J.R.; Soon Shin, K.; Kang, S.J. Resveratrol upregulated heat shock proteins and extended the survival of G93A-SOD1 mice. Brain Res. 2012, 1483, 112–117. [Google Scholar] [CrossRef] [PubMed]
- El Hayek, L.; Khalifeh, M.; Zibara, V.; Abi Assaad, R.; Emmanuel, N.; Karnib, N.; El-Ghandour, R.; Nasrallah, P.; Bilen, M.; Ibrahim, P.; et al. Lactate mediates the effects of exercise on learning and memory through sirt1-dependent activation of hippocampal brain-derived neurotrophic factor (BDNF). J. Neurosci. 2019, 39, 2369–2382. [Google Scholar] [CrossRef] [PubMed]
- Wu, W.F.; Chen, C.; Lin, J.T.; Jiao, X.H.; Dong, W.; Wan, J.; Liu, Q.; Qiu, Y.K.; Sun, A.; Liu, Y.Q.; et al. Impaired synaptic plasticity and decreased glutamatergic neuron excitability induced by SIRT1/BDNF downregulation in the hippocampal CA1 region are involved in postoperative cognitive dysfunction. Cell. Mol. Biol. Lett. 2024, 29, 79. [Google Scholar] [CrossRef]
- Caruso, G.I.; Spampinato, S.F.; Costantino, G.; Merlo, S.; Sortino, M.A. SIRT1-dependent upregulation of BDNF in human microglia challenged with Aβ: An early but transient response rescued by melatonin. Biomedicines 2021, 9, 466. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Li, C.; Li, R.; Yin, D.; Hong, Y.; Lu, M.; Xia, B.; Li, Y. Resveratrol by elevating the SIRT1 BDNF, GDNF and PSD95 levels reduce heroin addiction related behaviors. Neurosci. Lett. 2024, 841, 137934. [Google Scholar] [CrossRef]
- Wang, F.; Li, Y.; Tang, D.; Yang, B.; Tian, T.; Tian, M.; Meng, N.; Xie, W.; Zhang, C.; He, Z.; et al. Exploration of the SIRT1-mediated BDNF–TrkB signaling pathway in the mechanism of brain damage and learning and memory effects of fluorosis. Front. Public Heal. 2023, 11, 1247294. [Google Scholar] [CrossRef]
- Zhang, L.; Fang, Y.; Xu, Y.; Lian, Y.; Xie, N.; Wu, T.; Zhang, H.; Sun, L.; Zhang, R.; Wang, Z. Curcumin improves amyloid β-peptide (1-42) induced spatial memory deficits through BDNF-ERK signaling pathway. PLoS ONE 2015, 10, e0131525. [Google Scholar] [CrossRef]
- Goozee, K.G.; Shah, T.M.; Sohrabi, H.R.; Rainey-Smith, S.R.; Brown, B.; Verdile, G.; Martins, R.N. Examining the potential clinical value of curcumin in the prevention and diagnosis of Alzheimer’s disease. Br. J. Nutr. 2016, 115, 449–465. [Google Scholar] [CrossRef]
- Lou, S.; Gong, D.; Yang, M.; Qiu, Q.; Luo, J.; Chen, T. Curcumin Improves Neurogenesis in Alzheimer’s Disease Mice via the Upregulation of Wnt/β-Catenin and BDNF. Int. J. Mol. Sci. 2024, 25, 5123. [Google Scholar] [CrossRef]
- Pervin, M.; Unno, K.; Ohishi, T.; Tanabe, H.; Miyoshi, N.; Nakamura, Y. Beneficial Effects of Green Tea Catechins on Neurodegenerative Diseases. Molecules 2018, 23, 1297. [Google Scholar] [CrossRef]
- Zhang, S.; Zhu, Q.; Chen, J.Y.; OuYang, D.; Lu, J.H. The pharmacological activity of epigallocatechin-3-gallate (EGCG) on Alzheimer’s disease animal model: A systematic review. Phytomedicine 2020, 79, 153316. [Google Scholar] [CrossRef] [PubMed]
- Sharma, R.; Bhate, L.; Agrawal, Y.; Aspatwar, A. Advanced nutraceutical approaches to Parkinson’s disease: Bridging nutrition and neuroprotection. Nutr. Neurosci. 2025, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Valverde-Salazar, V.; Ruiz-Gabarre, D.; García-Escudero, V. Alzheimer’s Disease and Green Tea: Epigallocatechin-3-Gallate as a Modulator of Inflammation and Oxidative Stress. Antioxidants 2023, 12, 1460. [Google Scholar] [CrossRef] [PubMed]
- Sebastiani, G.; Almeida-Toledano, L.; Serra-Delgado, M.; Navarro-Tapia, E.; Sailer, S.; Valverde, O.; Garcia-Algar, O.; Andreu-Fernández, V. Therapeutic effects of catechins in less common neurological and neurodegenerative disorders. Nutrients 2021, 13, 2232. [Google Scholar] [CrossRef]
- Xu, Y.; Xie, M.; Xue, J.; Xiang, L.; Li, Y.; Xiao, J.; Xiao, G.; Wang, H.L. EGCG ameliorates neuronal and behavioral defects by remodeling gut microbiota and TotM expression in Drosophila models of Parkinson’s disease. FASEB J. 2020, 34, 5931–5950. [Google Scholar] [CrossRef]
- Shi, W.; Li, L.; Ding, Y.; Yang, K.; Chen, Z.; Fan, X.; Jiang, S.; Guan, Y.; Liu, Z.; Xu, D.; et al. The critical role of epigallocatechin gallate in regulating mitochondrial metabolism. Future Med. Chem. 2018, 10, 795–809. [Google Scholar] [CrossRef]
- de Oliveira, M.R.; Nabavi, S.F.; Daglia, M.; Rastrelli, L.; Nabavi, S.M. Epigallocatechin gallate and mitochondria—A story of life and death. Pharmacol. Res. 2016, 104, 70–85. [Google Scholar] [CrossRef]
- Ayyalasomayajula, N.; Bandaru, L.J.M.; Chetty, C.S.; Dixit, P.K.; Challa, S. Mitochondria-Mediated Moderation of Apoptosis by EGCG in Cytotoxic Neuronal Cells Induced by Lead (Pb) and Amyloid Peptides. Biol. Trace Elem. Res. 2022, 200, 3582–3593. [Google Scholar] [CrossRef]
- Tseng, H.L.; Li, C.J.; Huang, L.H.; Chen, C.Y.; Tsai, C.H.; Lin, C.N.; Hsu, H.Y. Quercetin 3-O-methyl ether protects FL83B cells from copper induced oxidative stress through the PI3K/Akt and MAPK/Erk pathway. Toxicol. Appl. Pharmacol. 2012, 264, 104–113. [Google Scholar] [CrossRef]
- Safi, A.; Heidarian, E.; Ahmadi, R. Quercetin Synergistically Enhances the Anticancer Efficacy of Docetaxel through Induction of Apoptosis and Modulation of PI3K/AKT, MAPK/ERK, and JAK/STAT3 Signaling Pathways in MDA-MB-231 Breast Cancer Cell Line. Int. J. Mol. Cell. Med. 2021, 10, 11. [Google Scholar] [CrossRef]
- Terracina, S.; Petrella, C.; Francati, S.; Lucarelli, M.; Barbato, C.; Minni, A.; Ralli, M.; Greco, A.; Tarani, L.; Fiore, M.; et al. Antioxidant Intervention to Improve Cognition in the Aging Brain: The Example of Hydroxytyrosol and Resveratrol. Int. J. Mol. Sci. 2022, 23, 15674. [Google Scholar] [CrossRef] [PubMed]
- Boronat, A.; Serreli, G.; Rodríguez-Morató, J.; Deiana, M.; de la Torre, R. Olive Oil Phenolic Compounds’ Activity against Age-Associated Cognitive Decline: Clinical and Experimental Evidence. Antioxidants 2023, 12, 1472. [Google Scholar] [CrossRef] [PubMed]
- Casamenti, F.; Stefani, M. Olive polyphenols: New promising agents to combat aging-associated neurodegeneration. Expert Rev. Neurother. 2017, 17, 345–358. [Google Scholar] [CrossRef] [PubMed]
- Micheli, L.; Bertini, L.; Bonato, A.; Villanova, N.; Caruso, C.; Caruso, M.; Bernini, R.; Tirone, F. Role of Hydroxytyrosol and Oleuropein in the Prevention of Aging and Related Disorders: Focus on Neurodegeneration, Skeletal Muscle Dysfunction and Gut Microbiota. Nutrients 2023, 15, 1767. [Google Scholar] [CrossRef]
- Mititelu, M.; Lupuliasa, D.; Neacșu, S.M.; Olteanu, G.; Busnatu, Ș.S.; Mihai, A.; Popovici, V.; Măru, N.; Boroghină, S.C.; Mihai, S.; et al. Polyunsaturated Fatty Acids and Human Health: A Key to Modern Nutritional Balance in Association with Polyphenolic Compounds from Food Sources. Foods 2025, 14, 46. [Google Scholar] [CrossRef]
- Vidyanti, A.N.; Rahmawati, F.; Rahman, R.H.; Prodjohardjono, A.; Gofir, A. Lifestyle interventions for dementia risk reduction: A review on the role of physical activity and diet in Western and Asian Countries. J. Prev. Alzheimer’s Dis. 2025, 12, 100028. [Google Scholar] [CrossRef]
- Stefani, M.; Rigacci, S. Beneficial properties of natural phenols: Highlight on protection against pathological conditions associated with amyloid aggregation. BioFactors 2014, 40, 482–493. [Google Scholar] [CrossRef]
- Kozioł-Kozakowska, A.; Wójcik, M.; Herceg-Čavrak, V.; Cobal, S.; Radovanovic, D.; Alvarez-Pitti, J.; Hartgring, I.; Piórecka, B.; Gabbianelli, R.; Drożdż, D. Dietary Strategies in the Prevention and Treatment of Hypertension in Children and Adolescents: A Narrative Review. Nutrients 2024, 16, 2786. [Google Scholar] [CrossRef]
- Sudarshan, K.; Boda, A.K.; Dogra, S.; Bose, I.; Yadav, P.N.; Aidhen, I.S. Discovery of an isocoumarin analogue that modulates neuronal functions via neurotrophin receptor TrkB. Bioorganic Med. Chem. Lett. 2019, 29, 585–590. [Google Scholar] [CrossRef]
- Román, G.C.; Jackson, R.E.; Gadhia, R.; Román, A.N.; Reis, J. Mediterranean diet: The role of long-chain ω-3 fatty acids in fish; polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine; probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Rev. Neurol. 2019, 175, 724–741. [Google Scholar] [CrossRef]
- Canudas, S.; Becerra-Tomas, N.; Hernandez-Alonso, P.; Galie, S.; Leung, C.; Crous-Bou, M.; De Vivo, I.; Gao, Y.; Gu, Y.; Meinila, J.; et al. Mediterranean Diet and Telomere Length: A Systematic Review and Meta-Analysis. Adv. Nutr. 2020, 11, 1544–1554. [Google Scholar] [CrossRef] [PubMed]
- van Soest, A.P.; Beers, S.; van de Rest, O.; de Groot, L.C. The Mediterranean-Dietary Approaches to Stop Hypertension Intervention for Neurodegenerative Delay (MIND) Diet for the Aging Brain: A Systematic Review. Adv. Nutr. 2024, 15, 100184. [Google Scholar] [CrossRef] [PubMed]
- McGrattan, A.M.; McGuinness, B.; McKinley, M.C.; Kee, F.; Passmore, P.; Woodside, J.V.; McEvoy, C.T. Diet and Inflammation in Cognitive Ageing and Alzheimer’s Disease. Curr. Nutr. Rep. 2019, 8, 53–65. [Google Scholar] [CrossRef] [PubMed]
- Morris, M.C.; Tangney, C.C.; Wang, Y.; Sacks, F.M.; Barnes, L.L.; Bennett, D.A.; Aggarwal, N.T. MIND diet slows cognitive decline with aging. Alzheimer’s Dement. 2015, 11, 1015–1022. [Google Scholar] [CrossRef]
- Stefaniak, O.; Dobrzyńska, M.; Drzymała-Czyż, S.; Przysławski, J. Diet in the Prevention of Alzheimer’s Disease: Current Knowledge and Future Research Requirements. Nutrients 2022, 14, 4564. [Google Scholar] [CrossRef]
- Valls-Pedret, C.; Lamuela-Raventós, R.M.; Medina-Remón, A.; Quintana, M.; Corella, D.; Pintó, X.; Martínez-González, M.Á.; Estruch, R.; Ros, E. Polyphenol-rich foods in the mediterranean diet are associated with better cognitive function in elderly subjects at high cardiovascular risk. J. Alzheimer’s Dis. 2012, 29, 773–782. [Google Scholar] [CrossRef]
- Lopresti, A.L.; Smith, S.J.; Pouchieu, C.; Pourtau, L.; Gaudout, D.; Pallet, V.; Drummond, P.D. Effects of a polyphenol-rich grape and blueberry extract (MemophenolTM) on cognitive function in older adults with mild cognitive impairment: A randomized, double-blind, placebo-controlled study. Front. Psychol. 2023, 14, 1144231. [Google Scholar] [CrossRef]
- Sasaki, A.; Kawai, E.; Watanabe, K.; Yamano, E.; Oba, C.; Nakamura, K.; Natsume, M.; Mizuno, K.; Watanabe, Y. Cacao Polyphenol-Rich Dark Chocolate Intake Contributes to Efficient Brain Activity during Cognitive Tasks: A Randomized, Single-Blinded, Crossover, and Dose-Comparison fMRI Study. Nutrients 2024, 16, 41. [Google Scholar] [CrossRef]
- Rajaram, S.; Jones, J.; Lee, G.J. Plant-based dietary patterns, plant foods, and age-related cognitive decline. Adv. Nutr. 2019, 10, 422–436. [Google Scholar] [CrossRef]
- Riegelman, E.; Xue, K.S.; Wang, J.S.; Tang, L. Gut–Brain Axis in Focus: Polyphenols, Microbiota, and Their Influence on α-Synuclein in Parkinson’s Disease. Nutrients 2024, 16, 2041. [Google Scholar] [CrossRef]
- Sarubbo, F.; Moranta, D.; Tejada, S.; Jiménez, M.; Esteban, S. Impact of Gut Microbiota in Brain Ageing: Polyphenols as Beneficial Modulators. Antioxidants 2023, 12, 812. [Google Scholar] [CrossRef] [PubMed]
- Prakash Reddy, V.; Aryal, P.; Robinson, S.; Rafiu, R.; Obrenovich, M.; Perry, G. Polyphenols in alzheimer’s disease and in the gut–brain axis. Microorganisms 2020, 8, 199. [Google Scholar] [CrossRef]
- Filosa, S.; Di Meo, F.; Crispi, S. Polyphenols-gut microbiota interplay and brain neuromodulation. Neural Regen. Res. 2018, 13, 2055–2059. [Google Scholar] [CrossRef] [PubMed]
- Kurhaluk, N.; Kamiński, P.; Bilski, R.; Kołodziejska, R.; Woźniak, A.; Tkaczenko, H. Role of Antioxidants in Modulating the Microbiota–Gut–Brain Axis and Their Impact on Neurodegenerative Diseases. Int. J. Mol. Sci. 2025, 26, 3658. [Google Scholar] [CrossRef]
- Tahiri, M.; Gilbert, J.A. Examining the potential prebiotic effect of almonds. J. Appl. Microbiol. 2025, 136, lxaf078. [Google Scholar] [CrossRef]
- Domínguez-López, I.; López-Yerena, A.; Vallverdú-Queralt, A.; Pallàs, M.; Lamuela-Raventós, R.M.; Pérez, M. From the gut to the brain: The long journey of phenolic compounds with neurocognitive effects. Nutr. Rev. 2024, 83, e533–e546. [Google Scholar] [CrossRef]
- Cao, Q.; Shen, M.; Li, R.; Liu, Y.; Zeng, Z.; Zhou, J.; Niu, D.; Zhang, Q.; Wang, R.; Yao, J.; et al. Elucidating the specific mechanisms of the gut-brain axis: The short-chain fatty acids-microglia pathway. J. Neuroinflammation 2025, 22, 133. [Google Scholar] [CrossRef]
- Molska, M.; Mruczyk, K.; Cisek-Woźniak, A.; Prokopowicz, W.; Szydełko, P.; Jakuszewska, Z.; Marzec, K.; Trocholepsza, M. The Influence of Intestinal Microbiota on BDNF Levels. Nutrients 2024, 16, 2891. [Google Scholar] [CrossRef]
- Arora, I.; Sharma, M.; Sun, L.Y.; Tollefsbol, T.O. The epigenetic link between polyphenols, aging and age-related diseases. Genes 2020, 11, 1094. [Google Scholar] [CrossRef]
- Pan, M.-H.; Lai, C.-S.; Wu, J.-C.; Ho, C.-T. Epigenetic and Disease Targets by Polyphenols. Curr. Pharm. Des. 2013, 19, 6156–6185. [Google Scholar] [CrossRef]
- Prasanth, M.I.; Sivamaruthi, B.S.; Cheong, C.S.Y.; Verma, K.; Tencomnao, T.; Brimson, J.M.; Prasansuklab, A. Role of Epigenetic Modulation in Neurodegenerative Diseases: Implications of Phytochemical Interventions. Antioxidants 2024, 13, 606. [Google Scholar] [CrossRef] [PubMed]
- Borsoi, F.T.; Neri-Numa, I.A.; de Oliveira, W.Q.; de Araújo, F.F.; Pastore, G.M. Dietary polyphenols and their relationship to the modulation of non-communicable chronic diseases and epigenetic mechanisms: A mini-review. Food Chem. Mol. Sci. 2023, 6, 100155. [Google Scholar] [CrossRef] [PubMed]
- Branco, C.S.; Duong, A.; Machado, A.K.; Scola, G.; Andreazza, A.C.; Salvador, M. Modulation of Mitochondrial and Epigenetic Targets by Polyphenols-rich Extract from Araucaria angustifolia in Larynx Carcinoma. Anticancer Agents Med. Chem. 2018, 19, 130–139. [Google Scholar] [CrossRef] [PubMed]
- Cuevas, A.; Saavedra, N.; Salazar, L.A.; Abdalla, D.S.P. Modulation of immune function by polyphenols: Possible contribution of epigenetic factors. Nutrients 2013, 5, 2314–2332. [Google Scholar] [CrossRef]
- Bankole, O.; Scambi, I.; Parrella, E.; Muccilli, M.; Bonafede, R.; Turano, E.; Pizzi, M.; Mariotti, R. Beneficial and Dimorphic Response to Combined HDAC Inhibitor Valproate and AMPK/SIRT1 Pathway Activator Resveratrol in the Treatment of ALS Mice. Int. J. Mol. Sci. 2022, 23, 1047. [Google Scholar] [CrossRef]
- Schiaffino, L.; Bonafede, R.; Scambi, I.; Parrella, E.; Pizzi, M.; Mariotti, R. Acetylation state of RelA modulated by epigenetic drugs prolongs survival and induces a neuroprotective effect on ALS murine model. Sci. Rep. 2018, 8, 12875. [Google Scholar] [CrossRef]
- Zhu, X.; Li, Q.; Chang, R.; Yang, D.; Song, Z.; Guo, Q.; Huang, C. Curcumin alleviates neuropathic pain by inhibiting p300/CBP histone acetyltransferase activity-regulated expression of BDNF and Cox-2 in a rat model. PLoS ONE 2014, 9, e91303. [Google Scholar] [CrossRef]
- Liang, D.Y.; Li, X.; Clark, J.D. Epigenetic regulation of opioid-induced hyperalgesia, dependence, and tolerance in mice. J. Pain 2013, 14, 36–47. [Google Scholar] [CrossRef]
- Lim, U.; Song, M.A. Dietary and lifestyle factors of DNA methylation. Methods Mol. Biol. 2012, 863, 359–376. [Google Scholar] [CrossRef]
- Ribarič, S. Diet and aging. Oxid. Med. Cell. Longev. 2012, 2012, 741468. [Google Scholar] [CrossRef]
- Divyajanani, S.; Harithpriya, K.; Ganesan, K.; Ramkumar, K.M. Dietary Polyphenols Remodel DNA Methylation Patterns of NRF2 in Chronic Disease. Nutrients 2023, 15, 3347. [Google Scholar] [CrossRef] [PubMed]
- Ramalingam, M.; Kim, S.J. Pharmacological activities and applications of spicatoside A. Biomol. Ther. 2016, 24, 469–474. [Google Scholar] [CrossRef] [PubMed]
- Minichiello, L.; Calella, A.M.; Medina, D.L.; Bonhoeffer, T.; Klein, R.; Korte, M. Mechanism of TrkB-mediated hippocampal long-term potentiation. Neuron 2002, 36, 121–137. [Google Scholar] [CrossRef] [PubMed]
- Fiorentino, H.; Kuczewski, N.; Diabira, D.; Ferrand, N.; Pangalos, M.N.; Porcher, C.; Gaiarsa, J.-L. GABAB Receptor Activation Triggers BDNF Release and Promotes the Maturation of GABAergic Synapses. J. Neurosci. 2009, 29, 11650–11661. [Google Scholar] [CrossRef]
- Chen, L.; Zhang, H.Y. Cancer preventive mechanisms of the green tea polyphenol (-)-epigallocatechin-3-gallate. Molecules 2007, 12, 946–957. [Google Scholar] [CrossRef]
- Singh, M.; Arseneault, M.; Sanderson, T.; Murthy, V.; Ramassamy, C. Challenges for research on polyphenols from foods in Alzheimer’s disease: Bioavailability, metabolism, and cellular and molecular mechanisms. J. Agric. Food Chem. 2008, 56, 4855–4873. [Google Scholar] [CrossRef]
- Piroddi, M.; Albini, A.; Fabiani, R.; Giovannelli, L.; Luceri, C.; Natella, F.; Rosignoli, P.; Rossi, T.; Taticchi, A.; Servili, M.; et al. Nutrigenomics of extra-virgin olive oil: A review. BioFactors 2017, 43, 17–41. [Google Scholar] [CrossRef]
- Strasser, B.; Gostner, J.M.; Fuchs, D. Mood, food, and cognition: Role of tryptophan and serotonin. Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 55–61. [Google Scholar] [CrossRef]
- Li, Y.R.; Li, S.; Lin, C.C. Effect of resveratrol and pterostilbene on aging and longevity. BioFactors 2018, 44, 69–82. [Google Scholar] [CrossRef]
- Weinreb, O.; Mandel, S.; Amit, T.; Youdim, M.B. Neurological mechanisms of green tea polyphenols in Alzheimer’s and Parkinson’s diseases. J. Nutr. Biochem. 2004, 15, 506–516. [Google Scholar] [CrossRef]
- Gandhi, G.R.; Antony, P.J.; de Paula Lana, M.J.M.; da Silva, B.F.X.; Oliveira, R.V.; Jothi, G.; Hariharan, G.; Mohana, T.; Gan, R.Y.; Gurgel, R.Q.; et al. Natural products modulating interleukins and other inflammatory mediators in tumor-bearing animals: A systematic review. Phytomedicine 2022, 100, 154038. [Google Scholar] [CrossRef] [PubMed]
- Cao, S.; Fu, X.; Yang, S.; Tang, S. The anti-inflammatory activity of resveratrol in acute kidney injury: A systematic review and meta-analysis of animal studies. Pharm. Biol. 2022, 60, 2088–2097. [Google Scholar] [CrossRef] [PubMed]
- Jomova, K.; Alomar, S.Y.; Valko, R.; Liska, J.; Nepovimova, E.; Kuca, K.; Valko, M. Flavonoids and their role in oxidative stress, inflammation, and human diseases. Chem. Biol. Interact. 2025, 413, 111489. [Google Scholar] [CrossRef] [PubMed]
- Tiwari, V.; Chopra, K. Resveratrol abrogates alcohol-induced cognitive deficits by attenuating oxidative-nitrosative stress and inflammatory cascade in the adult rat brain. Neurochem. Int. 2013, 62, 861–869. [Google Scholar] [CrossRef]
- Tiwari, V.; Chopra, K. Resveratrol prevents alcohol-induced cognitive deficits and brain damage by blocking inflammatory signaling and cell death cascade in neonatal rat brain. J. Neurochem. 2011, 117, 678–690. [Google Scholar] [CrossRef]
- Kita, T.; Asanuma, M.; Miyazaki, I.; Takeshima, M. Protective Effects of Phytochemical Antioxidants Against Neurotoxin-Induced Degeneration of Dopaminergic Neurons. J. Pharmacol. Sci. 2014, 124, 313–319. [Google Scholar] [CrossRef]
- Chandler, D.; Woldu, A.; Rahmadi, A.; Shanmugam, K.; Steiner, N.; Wright, E.; Benavente-García, O.; Schulz, O.; Castillo, J.; Münch, G. Effects of plant-derived polyphenols on TNF-alpha and nitric oxide production induced by advanced glycation endproducts. Mol. Nutr. Food Res. 2010, 54, S141–S150. [Google Scholar] [CrossRef]
- Asadi, S.; Ahmadiani, A.; Esmaeili, M.A.; Sonboli, A.; Ansari, N.; Khodagholi, F. In vitro antioxidant activities and an investigation of neuroprotection by six Salvia species from Iran: A comparative study. Food Chem. Toxicol. 2010, 48, 1341–1349. [Google Scholar] [CrossRef]
- Goutham, G.; Manikandan, R.; Arulvasu, C.; Arumugam, M.; Beulaja, M.; Thiagarajan, R.; Setzer, W.N.; Daglia, M.; Nabavi, S.F.; Nabavi, S.M. A focus on resveratrol and ocular problems, especially cataract: From chemistry to medical uses and clinical relevance. Biomed. Pharmacother. 2017, 86, 232–241. [Google Scholar] [CrossRef]
- Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The immunomodulatory and anti-inflammatory role of polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef]
- Edgecombe, S.C.; Stretch, G.L.; Hayball, P.J. Oleuropein, an antioxidant polyphenol from olive oil, is poorly absorbed from isolated perfused rat intestine. J. Nutr. 2000, 130, 2996–3002. [Google Scholar] [CrossRef] [PubMed]
- Segovia-Bravo, K.A.; Arroyo-Lopez, F.N.; Garcia-Garcia, P.; Duran-Quintana, M.C.; Garrido-Fernandez, A. Reuse of ozonated alkaline solutions as fermentation brines in Spanish green table olives. J. Food Sci. 2007, 72, M126–M133. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Wang, Z.; Yu, Z.; Niu, T. Dietary Flavonoid Intake and Anemia Risk in Children and Adolescents: Insights from National Health and Nutrition Examination Survey. Antioxidants 2025, 14, 395. [Google Scholar] [CrossRef] [PubMed]
- Amidfar, M.; Garcez, M.L.; Askari, G.; Bagherniya, M.; Khorvash, F.; Golpour-Hamedani, S.; de Oliveira, J. Role of BDNF Signaling in the Neuroprotective and Memory-enhancing Effects of Flavonoids in Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2024, 23, 984–995. [Google Scholar] [CrossRef]
- Melgar-Locatelli, S.; Mañas-Padilla, M.C.; Castro-Zavala, A.; Rivera, P.; del Carmen Razola-Díaz, M.; Monje, F.J.; Rodríguez-Pérez, C.; Castilla-Ortega, E. Diet enriched with high-phenolic cocoa potentiates hippocampal brain-derived neurotrophic factor expression and neurogenesis in healthy adult micewith subtle effects on memory. Food Funct. 2024, 15, 8310–8329. [Google Scholar] [CrossRef]
- Tan, L.; Zhang, H.; Li, H.; Sun, S.; Lyu, Q.; Jiang, Y. Blueberry extracts antagonize Aβ25–35 neurotoxicity and exert a neuroprotective effect through MEK-ERK-BDNF/UCH-L1 signaling pathway in rat and mouse hippocampus. Nutr. Neurosci. 2024, 27, 745–760. [Google Scholar] [CrossRef]
- Babaei, F.G.; Saburi, E.; Forouzanfar, F.; Asgari, M.; Keshavarzi, Z.; Hajali, V. Effect of epigallocatechin-3-gallate (EGCG) on cognitive functioning and the expression of APP and BDNF in the hippocampus of rats with streptozotocin -induced Alzheimer-like disease. Biochem. Biophys. Rep. 2025, 41, 101930. [Google Scholar] [CrossRef]
- Hammad, A.M.; Alzaghari, L.F.; Alfaraj, M.; Lux, V.; Sunoqrot, S. Green Tea Polyphenol Nanoparticles Reduce Anxiety Caused by Tobacco Smoking Withdrawal in Rats by Suppressing Neuroinflammation. Toxics 2024, 12, 598. [Google Scholar] [CrossRef]
- Jalouli, M.; Rahman, M.A.; Biswas, P.; Rahman, H.; Harrath, A.H.; Lee, I.S.; Kang, S.; Choi, J.; Park, M.N.; Kim, B. Targeting natural antioxidant polyphenols to protect neuroinflammation and neurodegenerative diseases: A comprehensive review. Front. Pharmacol. 2025, 16, 1492517. [Google Scholar] [CrossRef]
- Infante, R.; Infante, M.; Pastore, D.; Pacifici, F.; Chiereghin, F.; Malatesta, G.; Donadel, G.; Tesauro, M.; Della-Morte, D. An Appraisal of the Oleocanthal-Rich Extra Virgin Olive Oil (EVOO) and Its Potential Anticancer and Neuroprotective Properties. Int. J. Mol. Sci. 2023, 24, 17323. [Google Scholar] [CrossRef]
- Chou, L.M.; Lin, C.I.; Chen, Y.H.; Liao, H.; Lin, S.H. A diet containing grape powder ameliorates the cognitive decline in aged rats with a long-term high-fructose-high-fat dietary pattern. J. Nutr. Biochem. 2016, 34, 52–60. [Google Scholar] [CrossRef] [PubMed]
- Jaberi, K.R.; Alamdari-palangi, V.; Savardashtaki, A.; Vatankhah, P.; Jamialahmadi, T.; Tajbakhsh, A.; Sahebkar, A. Modulatory Effects of Phytochemicals on Gut–Brain Axis: Therapeutic Implication. Curr. Dev. Nutr. 2024, 8, 103785. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Villegas, A.; Galbete, C.; Martinez-González, M.A.; Martinez, J.A.; Razquin, C.; Salas-Salvadó, J.; Estruch, R.; Buil-Cosiales, P.; Martí, A. The effect of the Mediterranean diet on plasma brain-derived neurotrophic factor (BDNF) levels: The PREDIMED-NAVARRA randomized trial. Nutr. Neurosci. 2011, 14, 195–201. [Google Scholar] [CrossRef] [PubMed]
- Bańkowski, S.; Wójcik, Z.B.; Grabara, M.; Ozner, D.; Pałka, T.; Stanek, A.; Sadowska-Krępa, E. Does curcumin supplementation affect inflammation, blood count and serum brain-derived neurotropic factor concentration in amateur long-distance runners? PLoS ONE 2025, 20, e0317446. [Google Scholar] [CrossRef]
- Huhn, S.; Beyer, F.; Zhang, R.; Lampe, L.; Grothe, J.; Kratzsch, J.; Willenberg, A.; Breitfeld, J.; Kovacs, P.; Stumvoll, M.; et al. Effects of resveratrol on memory performance, hippocampus connectivity and microstructure in older adults—A randomized controlled trial. Neuroimage 2018, 174, 177–190. [Google Scholar] [CrossRef]
- O’neill Rothenberg, D.; Zhang, L. Mechanisms underlying the anti-depressive effects of regular tea consumption. Nutrients 2019, 11, 1361. [Google Scholar] [CrossRef]
- García-Cordero, J.; Pino, A.; Cuevas, C.; Puertas-Martín, V.; San Román, R.; de Pascual-Teresa, S. Neurocognitive effects of cocoa and red-berries consumption in healthy adults. Nutrients 2022, 14, 1. [Google Scholar] [CrossRef]
- Cimini, A.; Gentile, R.; D’Angelo, B.; Benedetti, E.; Cristiano, L.; Avantaggiati, M.L.; Giordano, A.; Ferri, C.; Desideri, G. Cocoa powder triggers neuroprotective and preventive effects in a human Alzheimer’s disease model by modulating BDNF signaling pathway. J. Cell. Biochem. 2013, 114, 2209–2220. [Google Scholar] [CrossRef]
- Volkow, N.D.; Michaelides, M.; Baler, R. The neuroscience of drug reward and addiction. Physiol. Rev. 2019, 99, 2115–2140. [Google Scholar] [CrossRef]
- Volkow, N.D.; Morales, M. The Brain on Drugs: From Reward to Addiction. Cell 2015, 162, 712–725. [Google Scholar] [CrossRef]
- Kauer, J.A.; Malenka, R.C. Synaptic plasticity and addiction. Nat. Rev. Neurosci. 2007, 8, 844–858. [Google Scholar] [CrossRef] [PubMed]
- Ceci, F.M.; Ferraguti, G.; Petrella, C.; Greco, A.; Ralli, M.; Iannitelli, A.; Carito, V.; Tirassa, P.; Chaldakov, G.N.; Messina, M.P.; et al. Nerve Growth Factor in Alcohol Use Disorders. Curr. Neuropharmacol. 2020, 19, 45–60. [Google Scholar] [CrossRef] [PubMed]
- Angelucci, F.; Ricci, V.; Pomponi, M.; Conte, G.; Mathé, A.A.; Attilio Tonali, P.; Bria, P. Chronic heroin and cocaine abuse is associated with decreased serum concentrations of the nerve growth factor and brain-derived neurotrophic factor. J. Psychopharmacol. 2007, 21, 820–825. [Google Scholar] [CrossRef] [PubMed]
- Inal-Emiroglu, F.N.; Karabay, N.; Resmi, H.; Guleryuz, H.; Baykara, B.; Alsen, S.; Senturk-Pilan, B.; Akay, A.; Kose, S. Correlations between amygdala volumes and serum levels of BDNF and NGF as a neurobiological markerin adolescents with bipolar disorder. J. Affect. Disord. 2015, 182, 50–56A. [Google Scholar] [CrossRef]
- Tseilikman, V.E.; Shatilov, V.A.; Zhukov, M.S.; Buksha, I.A.; Epitashvily, A.E.; Lipatov, I.A.; Aristov, M.R.; Koshelev, A.G.; Karpenko, M.N.; Traktirov, D.S.; et al. Limited Cheese Intake Paradigm Replaces Patterns of Behavioral Disorders in Experimental PTSD: Focus on Resveratrol Supplementation. Int. J. Mol. Sci. 2023, 24, 14343. [Google Scholar] [CrossRef]
- Gu, Z.; Chu, L.; Han, Y. Therapeutic effect of resveratrol on mice with depression. Exp. Ther. Med. 2019, 17, 3061–3064. [Google Scholar] [CrossRef]
- Chakraborty, A.K.; Tiwari, P.; Khobragade, D.S.; Kadiri, S.K.; Sheikh, I.A.; Thakur, J. The Neuroprotective Action of Resveratrol Against Cognitive Impairments Induced by Lorazepam in Male Rats. Curr. Drug Saf. 2024, 20, 341–348. [Google Scholar] [CrossRef]
- Li, Y.; Yu, L.; Zhao, L.; Zeng, F.; Liu, Q.S. Resveratrol modulates cocaine-induced inhibitory synaptic plasticity in VTA dopamine neurons by inhibiting phosphodiesterases (PDEs). Sci. Rep. 2017, 7, 15657. [Google Scholar] [CrossRef]
- Shuto, T.; Kuroiwa, M.; Koga, Y.; Kawahara, Y.; Sotogaku, N.; Toyomasu, K.; Nishi, A. Acute effects of resveratrol to enhance cocaine-induced dopamine neurotransmission in the striatum. Neurosci. Lett. 2013, 542, 107–112. [Google Scholar] [CrossRef]
- Nguyen, H.D. Resveratrol, Endocrine Disrupting Chemicals, Neurodegenerative Diseases and Depression: Genes, Transcription Factors, microRNAs, and Sponges Involved. Neurochem. Res. 2023, 48, 604–624. [Google Scholar] [CrossRef]
- Petrella, C.; Carito, V.; Carere, C.; Ferraguti, G.; Ciafrè, S.; Natella, F.; Bello, C.; Greco, A.; Ralli, M.; Mancinelli, R.; et al. Oxidative stress inhibition by resveratrol in alcohol-dependent mice. Nutrition 2020, 79–80, 110783. [Google Scholar] [CrossRef] [PubMed]
- Petrella, C.; Di Certo, M.G.; Gabanella, F.; Barbato, C.; Ceci, F.M.; Greco, A.; Ralli, M.; Polimeni, A.; Angeloni, A.; Severini1, C.; et al. Mediterranean Diet, Brain and Muscle: Olive Polyphenols and Resveratrol Protection in Neurodegenerative and Neuromuscular Disorders. Curr. Med. Chem. 2021, 28, 7595–7613. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.-M.; Lin, X.; Su, Y.-J.; Xue, D.; Zhang, C. Effects of curcumin on liver injury induced by chronic alcohol addiction. Zhongguo Ying Yong Sheng Li Xue Za Zhi 2022, 38, 782–786. [Google Scholar] [CrossRef] [PubMed]
- Soltaninejad, M.; Amleshi, R.S.; Shabani, M.; Ilaghi, M. Unraveling the protective effects of curcumin against drugs of abuse. Heliyon 2024, 10, e30468. [Google Scholar] [CrossRef]
- Bandyopadhyaya, G.; Sinha, S.; Chattopadhyay, B.D.; Chakraborty, A. Protective role of curcumin against nicotine-induced genotoxicity on rat liver under restricted dietary protein. Eur. J. Pharmacol. 2008, 588, 151–157. [Google Scholar] [CrossRef]
- Poulose, S.M.; Miller, M.G.; Scott, T.; Shukitt-Hale, B. Nutritional factors affecting adult neurogenesis and cognitive function. Adv. Nutr. 2017, 8, 804–811. [Google Scholar] [CrossRef]
- Kulkarni, S.K.; Bhutani, M.K.; Bishnoi, M. Antidepressant activity of curcumin: Involvement of serotonin and dopamine system. Psychopharmacology 2008, 201, 435–442. [Google Scholar] [CrossRef]
- Chang, X.R.; Wang, L.; Li, J.; Wu, D.S. Analysis of anti-depressant potential of curcumin against depression induced male albino wistar rats. Brain Res. 2016, 1642, 219–225. [Google Scholar] [CrossRef]
- Arora, V.; Kuhad, A.; Tiwari, V.; Chopra, K. Curcumin ameliorates reserpine-induced pain-depression dyad: Behavioural, biochemical, neurochemical and molecular evidences. Psychoneuroendocrinology 2011, 36, 1570–1581. [Google Scholar] [CrossRef]
- Rahmadi, M.; Nurhan, A.D.; Rahmawati, R.I.A.; Damayanti, T.F.; Purwanto, D.A.; Khotib, J. Epigallocatechin Gallate Ameliorates Nicotine Withdrawal Conditions-Induced Somatic and Affective Behavior Changes in Mice and Its Molecular Mechanism. Behav. Neurol. 2023, 2023, 5581893. [Google Scholar] [CrossRef]
- Lardner, A.L. Neurobiological effects of the green tea constituent theanine and its potential role in the treatment of psychiatric and neurodegenerative disorders. Nutr. Neurosci. 2014, 17, 145–155. [Google Scholar] [CrossRef] [PubMed]
- Sangiovanni, E.; Brivio, P.; Dell’Agli, M.; Calabrese, F. Botanicals as Modulators of Neuroplasticity: Focus on BDNF. Neural Plast. 2017, 2017, 5965371. [Google Scholar] [CrossRef] [PubMed]
- Meeusen, R. Exercise, nutrition and the brain. Sport. Med. 2014, 44, S47–S56. [Google Scholar] [CrossRef] [PubMed]
- Si, Z.; Wang, X.; Yu, Z.; Ruan, Y.; Qian, L.; Lin, S.; Gong, X.; Li, L.; Huang, J.; Liu, Y. EGCG attenuates METH self-administration and reinstatement of METH seeking in mice. Addict. Biol. 2023, 28, e13307. [Google Scholar] [CrossRef]
- Vieiros, M.; Almeida-Toledano, L.; Serra-Delgado, M.; Navarro-Tapia, E.; Ramos-Triguero, A.; Muñoz-Lozano, C.; Martínez, L.; Marchei, E.; Gómez-Roig, M.D.; Algar, Ó.G.; et al. Effects of maternal drinking patterns and epigallocatechin-3-gallate treatment on behavioural and molecular outcomes in a mouse model of fetal alcohol spectrum disorders. Biomed. Pharmacother. 2025, 187, 118138. [Google Scholar] [CrossRef]
- Wang, Y.; Wu, S.; Li, Q.; Lang, W.; Li, W.; Jiang, X.; Wan, Z.; Chen, J.; Wang, H. Epigallocatechin-3-gallate: A phytochemical as a promising drug candidate for the treatment of Parkinson’s disease. Front. Pharmacol. 2022, 13, 977521. [Google Scholar] [CrossRef]
- Kostrzewa, R.M.; Segura-Aguilar, J. Novel mechanisms and approaches in the study of neurodegeneration and neuroprotection. A review. Neurotox. Res. 2003, 5, 375–383. [Google Scholar] [CrossRef]
- Lerner, T.N.; Holloway, A.L.; Seiler, J.L. Dopamine, Updated: Reward Prediction Error and Beyond. Curr. Opin. Neurobiol. 2021, 67, 123–130. [Google Scholar] [CrossRef]
- Schultz, W. Predictive reward signal of dopamine neurons. J. Neurophysiol. 1998, 80, 1–27. [Google Scholar] [CrossRef]
- Petrella, C.; Ferraguti, G.; Tarani, L.; Chaldakov, G.N.; Ceccanti, M.; Greco, A.; Ralli, M.; Fiore, M. Olive Polyphenols and Chronic Alcohol Protection. In Olives and Olive Oil in Health and Disease Prevention, 2nd ed.; Preedy, V.R., Ross Watson, R., Eds.; Elsevier: Amsterdam, The Netherlands, 2021; Chapter 39; pp. 471–478. [Google Scholar]
- Nani, A.; Murtaza, B.; Khan, A.S.; Khan, N.A.; Hichami, A. Antioxidant and anti-inflammatory potential of polyphenols contained in Mediterranean diet in obesity: Molecular mechanisms. Molecules 2021, 26, 985. [Google Scholar] [CrossRef]
- Robles-Almazan, M.; Pulido-Moran, M.; Moreno-Fernandez, J.; Ramirez-Tortosa, C.; Rodriguez-Garcia, C.; Quiles, J.L.; Ramirez-Tortosa, M. Hydroxytyrosol: Bioavailability, toxicity, and clinical applications. Food Res. Int. 2018, 105, 654–667. [Google Scholar] [CrossRef] [PubMed]
- Carito, V.; Venditti, A.; Bianco, A.; Ceccanti, M.; Serrilli, A.M.; Chaldakov, G.; Tarani, L.; De Nicolò, S.; Fiore, M. Effects of olive leaf polyphenols on male mouse brain NGF, BDNF and their receptors TrkA, TrkB and p75. Nat. Prod. Res. 2014, 28, 1970–1984. [Google Scholar] [CrossRef] [PubMed]
- Carito, V.; Ceccanti, M.; Cestari, V.; Natella, F.; Bello, C.; Coccurello, R.; Mancinelli, R.; Fiore, M. Olive polyphenol effects in a mouse model of chronic ethanol addiction. Nutrition 2017, 33, 65–69. [Google Scholar] [CrossRef] [PubMed]
- Pasban-Aliabadi, H.; Esmaeili-Mahani, S.; Sheibani, V.; Abbasnejad, M.; Mehdizadeh, A.; Yaghoobi, M.M. Inhibition of 6-hydroxydopamine-induced PC12 cell apoptosis by olive (Olea europaea L.) leaf extract is performed by its main component oleuropein. Rejuvenation Res. 2013, 16, 134–142. [Google Scholar] [CrossRef]
- Achour, I.; Arel-Dubeau, A.M.; Renaud, J.; Legrand, M.; Attard, E.; Germain, M.; Martinoli, M.G. Oleuropein prevents neuronal death, mitigates mitochondrial superoxide production and modulates autophagy in a dopaminergic cellular model. Int. J. Mol. Sci. 2016, 17, 1293. [Google Scholar] [CrossRef]
- Badr, A.M.; Attia, H.A.; Al-Rasheed, N. Oleuropein Reverses Repeated Corticosterone-Induced Depressive-Like Behavior in mice: Evidence of Modulating Effect on Biogenic Amines. Sci. Rep. 2020, 10, 3336. [Google Scholar] [CrossRef]
- Marcelino, G.; Hiane, P.A.; Freitas, K.D.C.; Santana, L.F.; Pott, A.; Donadon, J.R.; Guimarães, R.D.C.A. Effects of olive oil and its minor components on cardiovascular diseases, inflammation, and gut microbiota. Nutrients 2019, 11, 1826. [Google Scholar] [CrossRef]
- Pojero, F.; Aiello, A.; Gervasi, F.; Caruso, C.; Ligotti, M.E.; Calabrò, A.; Procopio, A.; Candore, G.; Accardi, G.; Allegra, M. Effects of Oleuropein and Hydroxytyrosol on Inflammatory Mediators: Consequences on Inflammaging. Int. J. Mol. Sci. 2023, 24, 380. [Google Scholar] [CrossRef]
- Pérez-Mañá, C.; Farré, M.; Pujadas, M.; Mustata, C.; Menoyo, E.; Pastor, A.; Langohr, K.; De La Torre, R. Ethanol induces hydroxytyrosol formation in humans. Pharmacol. Res. 2015, 95–96, 27–33. [Google Scholar] [CrossRef]
- De La Torre, R. Bioavailability of olive oil phenolic compounds in humans. Inflammopharmacology 2008, 16, 245–247. [Google Scholar] [CrossRef]
- Hersant, H.; He, S.; Maliha, P.; Grossberg, G. Over the Counter Supplements for Memory: A Review of Available Evidence. CNS Drugs 2023, 37, 797–817. [Google Scholar] [CrossRef] [PubMed]
- Thirumdas, R.; Kothakota, A.; Pandiselvam, R.; Bahrami, A.; Barba, F.J. Role of food nutrients and supplementation in fighting against viral infections and boosting immunity: A review. Trends Food Sci. Technol. 2021, 110, 66–77. [Google Scholar] [CrossRef] [PubMed]
- Solfrizzi, V.; Agosti, P.; Lozupone, M.; Custodero, C.; Schilardi, A.; Valiani, V.; Sardone, R.; Dibello, V.; Di Lena, L.; Lamanna, A.; et al. Nutritional Intervention as a Preventive Approach for Cognitive-Related Outcomes in Cognitively Healthy Older Adults: A Systematic Review. J. Alzheimer’s Dis. 2018, 64, S229–S254. [Google Scholar] [CrossRef] [PubMed]
- Neshatdoust, S.; Saunders, C.; Castle, S.M.; Vauzour, D.; Williams, C.; Butler, L.; Lovegrove, J.A.; Spencer, J.P.E. High-flavonoid intake induces cognitive improvements linked to changes in serum brain-derived neurotrophic factor: Two randomised, controlled trials. Nutr. Heal. Aging 2016, 4, 81–93. [Google Scholar] [CrossRef]
- Nehlig, A. The neuroprotective effects of cocoa flavanol and its influence on cognitive performance. Br. J. Clin. Pharmacol. 2013, 75, 716–727. [Google Scholar] [CrossRef]
- Abdelmeguid, N.E.; Hammad, T.M.; Abdel-Moneim, A.M.; Salam, S.A. Effect of Epigallocatechin-3-gallate on Stress-Induced Depression in a Mouse Model: Role of Interleukin-1β and Brain-Derived Neurotrophic Factor. Neurochem. Res. 2022, 47, 3464–3475. [Google Scholar] [CrossRef]
- Sloan, R.P.; Wall, M.; Yeung, L.K.; Feng, T.; Feng, X.; Provenzano, F.; Schroeter, H.; Lauriola, V.; Brickman, A.M.; Small, S.A. Insights into the role of diet and dietary flavanols in cognitive aging: Results of a randomized controlled trial. Sci. Rep. 2021, 11, 3837. [Google Scholar] [CrossRef]
- Moriya, J.; Chen, R.; Yamakawa, J.; Sasaki, K.; Ishigaki, Y.; Takahashi, T. Resveratrol improves hippocampal atrophy in chronic fatigue mice by enhancing neurogenesis and inhibiting apoptosis of granular cells. Biol. Pharm. Bull. 2011, 34, 354–359. [Google Scholar] [CrossRef]
- Chen, J.J.; Shen, J.X.; Yu, Z.H.; Pan, C.; Han, F.; Zhu, X.L.; Xu, H.; Xu, R.T.; Wei, T.Y.; Lu, Y.P. The Antidepressant Effects of Resveratrol are Accompanied by the Attenuation of Dendrite/Dendritic Spine Loss and the Upregulation of BDNF/p-cofilin1 Levels in Chronic Restraint Mice. Neurochem. Res. 2021, 46, 660–674. [Google Scholar] [CrossRef]
- García-Aguilar, A.; Palomino, O.; Benito, M.; Guillén, C. Dietary polyphenols in metabolic and neurodegenerative diseases: Molecular targets in autophagy and biological effects. Antioxidants 2021, 10, 142. [Google Scholar] [CrossRef]
- Farhan, M.; Rizvi, A. Understanding the Prooxidant Action of Plant Polyphenols in the Cellular Microenvironment of Malignant Cells: Role of Copper and Therapeutic Implications. Front. Pharmacol. 2022, 13, 929853. [Google Scholar] [CrossRef] [PubMed]
- Rahim, R.A.; Jayusman, P.A.; Muhammad, N.; Ahmad, F.; Mokhtar, N.; Mohamed, I.N.; Mohamed, N.; Shuid, A.N. Recent advances in nanoencapsulation systems using plga of bioactive phenolics for protection against chronic diseases. Int. J. Environ. Res. Public. Health 2019, 16, 4962. [Google Scholar] [CrossRef]
- Khatoon, S.; Kalam, N.; Shaikh, M.F.; Hasnain, M.S.; Hafiz, A.K.; Ansari, M.T. Nanoencapsulation of Polyphenols as Drugs and Supplements for Enhancing Therapeutic Profile—A Review. Curr. Mol. Pharmacol. 2021, 15, 77–107. [Google Scholar] [CrossRef]
- Trindade, L.R.; da Silva, D.V.T.; Baião, D.D.S.; Paschoalin, V.M.F. Increasing the power of polyphenols through nanoencapsulation for adjuvant therapy against cardiovascular diseases. Molecules 2021, 26, 4621. [Google Scholar] [CrossRef]
- Noor, N.; Gani, A.; Gani, A.; Shah, A.; Ashraf, Z. Exploitation of polyphenols and proteins using nanoencapsulation for anti-viral and brain boosting properties—Evoking a synergistic strategy to combat COVID-19 pandemic. Int. J. Biol. Macromol. 2021, 180, 375–384. [Google Scholar] [CrossRef]
- Jayusman, P.A.; Nasruddin, N.S.; Mahamad Apandi, N.I.; Ibrahim, N.; Budin, S.B. Therapeutic Potential of Polyphenol and Nanoparticles Mediated Delivery in Periodontal Inflammation: A Review of Current Trends and Future Perspectives. Front. Pharmacol. 2022, 13, 847702. [Google Scholar] [CrossRef]
- El-Saadony, M.T.; Yang, T.; Korma, S.A.; Sitohy, M.; Abd El-Mageed, T.A.; Selim, S.; Al Jaouni, S.K.; Salem, H.M.; Mahmmod, Y.; Soliman, S.M.; et al. Impacts of turmeric and its principal bioactive curcumin on human health: Pharmaceutical, medicinal, and food applications: A comprehensive review. Front. Nutr. 2023, 9, 1040259. [Google Scholar] [CrossRef]
- Elanthendral, G.; Shobana, N.; Meena, R.; Samrot, A.V. Utilizing pharmacological properties of polyphenolic curcumin in nanotechnology. Biocatal. Agric. Biotechnol. 2021, 38, 102212. [Google Scholar] [CrossRef]
- Cas, M.D.; Ghidoni, R. Dietary curcumin: Correlation between bioavailability and health potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef]
- Górnicka, J.; Mika, M.; Wróblewska, O.; Siudem, P.; Paradowska, K. Methods to Improve the Solubility of Curcumin from Turmeric. Life 2023, 13, 207. [Google Scholar] [CrossRef]
- Hegde, M.; Girisa, S.; BharathwajChetty, B.; Vishwa, R.; Kunnumakkara, A.B. Curcumin Formulations for Better Bioavailability: What We Learned from Clinical Trials Thus Far? ACS Omega 2023, 8, 10713–10746. [Google Scholar] [CrossRef] [PubMed]
- Bansal, S.S.; Goel, M.; Aqil, F.; Vadhanam, M.V.; Gupta, R.C. Advanced drug delivery systems of curcumin for cancer chemoprevention. Cancer Prev. Res. 2011, 4, 1158–1171. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Wang, J.; Chen, Z.; Luo, J.; Guo, W.; Sun, L.; Lin, L. Targeting M2-like tumor-associated macrophages is a potential therapeutic approach to overcome antitumor drug resistance. npj Precis. Oncol. 2024, 8, 31. [Google Scholar] [CrossRef] [PubMed]
- Muzio, L.; Viotti, A.; Martino, G. Microglia in Neuroinflammation and Neurodegeneration: From Understanding to Therapy. Front. Neurosci. 2021, 15, 742065. [Google Scholar] [CrossRef]
- Abe, N.; Nishihara, T.; Yorozuya, T.; Tanaka, J. Microglia and Macrophages in the Pathological Central and Peripheral Nervous Systems. Cells 2020, 9, 2132. [Google Scholar] [CrossRef]
- Miyamoto, Y.; Kubota, K.; Asawa, Y.; Hoshi, K.; Hikita, A. M1-like macrophage contributes to chondrogenesis in vitro. Sci. Rep. 2021, 11, 21307. [Google Scholar] [CrossRef]
- Yamasaki, R. Elucidation of the role of microglia-macrophage-based neuroinflammation in neurological diseases. Neurol. Clin. Neurosci. 2024, 12, 329–339. [Google Scholar] [CrossRef]
- Wes, P.D.; Holtman, I.R.; Boddeke, E.W.G.M.; Möller, T.; Eggen, B.J.L. Next generation transcriptomics and genomics elucidate biological complexity of microglia in health and disease. Glia 2016, 64, 197–213. [Google Scholar] [CrossRef]
- Nakaso, K. Roles of Microglia in Neurodegenerative Diseases. Yonago Acta Med. 2024, 67, 1–8. [Google Scholar] [CrossRef]
- Jie, S.; Fu, A.; Wang, C.; Rajabi, S. A comprehensive review on the impact of polyphenol supplementation and exercise on depression and brain function parameters. Behav. Brain Funct. 2025, 21, 10. [Google Scholar] [CrossRef]
Polyphenols | Proposed Mechanisms | Molecular Pathway/Targets | Biological Outcomes |
---|---|---|---|
Resveratrol, epigallocatechin gallate (EGCG), curcumin | CREB activation |
| Enhanced synaptic plasticity, memory consolidation, and neuronal survival |
EGCG, resveratrol, quercetin | Reduction of oxidative stress |
| Decreased reactive oxygen species levels, protection against lipid peroxidation and cellular damage |
Curcumin, resveratrol, hydroxytyrosol (olive polyphenols) | Modulation of neuroinflammatory mediators |
| Attenuated neuroinflammatory responses and improved overall neuronal homeostasis |
Polyphenol | Targeted Neurotrophins/Receptors | Signaling Pathways Affected | Neurological Effects | Experimental Models |
---|---|---|---|---|
Resveratrol | BDNF, NGF, TrkB, TrkA | -↑ CREB phosphorylation—Activation of PI3K/Akt and ERK cascade | Improves memory, enhances synaptic plasticity | Rodent models, in vitro neuronal culture studies |
EGCG (green tea) | BDNF, TrkB | -↑ BDNF expression—Suppression of NF-κB-driven inflammation | Protects neurons from oxidative stress, reduces apoptosis | Mouse models of cognitive dysfunction, aging studies |
Curcumin | NGF, BDNF, TrkA, TrkB | - Modulates NF-κB—↑ Anti-apoptotic Bcl-2 pathways | Enhances neurogenesis, prevents inflammation | Parkinson’s and Alzheimer’s animal studies |
Quercetin | BDNF, NGF | -↑ MAPK/ERK signaling—Regulates neurotrophin transcription factors | Enhances neuronal resilience, neuroprotection | In vitro hippocampal neuron models |
Hydroxytyrosol (olive polyphenols) | NGF, BDNF, TrkA, TrkB | - Increases NGF synthesis—Regulates mitochondrial oxidative metabolism | Supports neuronal survival, counters neurodegeneration | Preclinical models of Alzheimer’s and aging disorders |
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. |
© 2025 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
Fiore, M.; Terracina, S.; Ferraguti, G. Brain Neurotrophins and Plant Polyphenols: A Powerful Connection. Molecules 2025, 30, 2657. https://doi.org/10.3390/molecules30122657
Fiore M, Terracina S, Ferraguti G. Brain Neurotrophins and Plant Polyphenols: A Powerful Connection. Molecules. 2025; 30(12):2657. https://doi.org/10.3390/molecules30122657
Chicago/Turabian StyleFiore, Marco, Sergio Terracina, and Giampiero Ferraguti. 2025. "Brain Neurotrophins and Plant Polyphenols: A Powerful Connection" Molecules 30, no. 12: 2657. https://doi.org/10.3390/molecules30122657
APA StyleFiore, M., Terracina, S., & Ferraguti, G. (2025). Brain Neurotrophins and Plant Polyphenols: A Powerful Connection. Molecules, 30(12), 2657. https://doi.org/10.3390/molecules30122657