Polyphenols in Parkinson’s Disease: A Systematic Review of In Vivo Studies
Abstract
:1. Introduction
2. Studies in Animal Models
2.1. Pure Compounds
2.1.1. 6−hydroxydopamine Model
2.1.2. N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine Model
2.1.3. Rotenone Model
2.1.4. Lipopolysaccharide Model
2.1.5. Homocysteine Model
2.2. Beverages and Extracts
3. Epidemiological Studies
4. The Permeability of Polyphenols Across the Blood-Brain Barrier
5. Polyphenols and the Microbiota–Gut–Brain Axis
6. Summary and Conclusions
Author Contributions
Conflicts of Interest
Abbreviations
4-HNE | 4-hydroxynonenal |
6-OHDA | 6-Hydroxydopamine |
AChE | Acetylcholinesterase |
AKT | serine/threonine protein kinase |
ARE | Antioxidant responsive element |
Bax | Bcl-2-like protein 4 |
Bcl-2 | B-cell lymphoma 2 |
BDNF | Brain-derived neurotrophic factor. |
b.w. | Body weight |
CaMKII | Ca2+/calmodulin-dependent protein kinase II |
Casp-3 | Caspase-3 |
Casp-8 | Caspase-8 |
Casp-9 | Caspase-9 |
CAT | Catalase |
CHIP | C terminus Hsp70 interacting protein |
CNS | Central nervous system |
COI | Cytochrome c oxidase I |
COX-2 | Cyclooxygenase-2 |
CUR | Curcumin |
DA | Dopamine |
DARPP-32 | Dopamine- and cAMP-regulated phosphoprotein |
DAT | Dopamine transporter |
DJ-1 | Protein deglycase |
DOPAC | 3,4-dihydroxyphenylacetic acid |
EC | (−)-Epicatechin |
ECG | Epicatechin gallate |
EGC | (−)-Epigallocatechin |
EGCG | Epigallocatechin-3-gallate |
ERK | Extracellular signal-regulated kinase protein-serine/threonine kinase |
FA | Ferulic acid |
GAPDH | Glyceraldehyde 3-phosphate dehydrogenase |
GABA | Gamma-aminobutyric acid |
GCLC | Glutamate cysteine ligase catalytic subunit |
GCLM | Glutamate cysteine ligase modifier subunit |
GDNF | Glial cell-derived neurotrophic factor |
GFAP | Glial fibrillary acidic protein |
GLu | Glutamate (or Glutamic acid) |
GPx | Glutathione peroxidase |
GR | Glutathione reductase |
GRP-78 | 78 kDa Glucose-regulated protein |
GS | Glutamine synthetase |
GSH | Reduced glutathione |
GSSG | Oxidized glutathione |
GTPs | Green tea polyphenols |
Hcy | Homocysteine |
HO-1 | Heme oxygenase-1 |
Hsp70s | The 70 kDa heat shock proteins |
HVA | Homovanillic acid |
Iba1 | Ionised calcium binding adapter molecule 1 |
i.p. | Intraperitoneally |
IL-1β | Interleukin 1 beta |
IL-6 | Interleukin 6 |
IL-10 | Interleukin 10 |
iNOS | Inducible nitric oxide synthase |
JNK | c-Jun N-terminal kinase |
JSE | Juglandis Semen extract |
L-DOPA | l-3,4-dihydroxyphenylalanine |
LPS | Lipopolysaccharide |
MAO | Monoamine oxidase |
MAPK | Mitogen-activated protein kinase |
MB | Maneb |
MDA | Malondialdehyde |
MMPs | N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine |
MPTP | Mitochondrial transmembrane potentials |
mTORC1 | Mammalian target of rapamycin complex 1 |
NF-кB | Nuclear factor-кB |
nNOS | Nuclear nitric oxide synthase |
NO | Nitric oxide |
Nrf2 | Nuclear factor (erythroid-derived 2)-like 2 |
NQO1 | NAD(P)H: quinone oxidoreductase 1 |
(p) | probenocid |
p-4E-BP1 | Phosphorylated 4E-binding protein 1 |
p38 | MAP Kinase (MAPK), CSBP Cytokinin-Specific Binding Protein or RK |
PARP | Poly(ADP-ribose) polymerase |
PCA | Protocatechuic acid |
PCs | Protein carbonyls |
PD | Parkinson disease |
PGE2 | Prostaglandin E2 |
PI3K | phosphoinositide 3-kinase |
PKC | Protein kinase C |
PLA2 | Phospholipases A2 |
p.o. | Orally |
PQ | Resveratrol |
RES | Paraquat |
ROS | Reactive oxygen species |
ROT | Rotenone |
SA | Syringic acid |
SIL | Silibinin |
SNpc | Substantia nigra pars compacta |
SOD | Superoxide dismutase |
TBARS | Thiobarbituric acid reactive substances |
TH | Tyrosine hydroxylase |
TNF-α | Tumor necrosis factor alpha |
TRAP | Total reactive antioxidant potential |
Trx | Thioredoxin |
TF | Theaflavin |
TPs | Tea polyphenols |
TrkB | Tropomyosin receptor kinase B |
TUNEL | Terminal deoxynucleotidyl transferase dUTP nick end labeling |
UA | Urolithin A |
VMAT2 | Vesicular monoamine transporter-2 |
References
- Reglodi, D.; Renaud, J.; Tamas, A.; Tizabi, Y.; Socías, S.B.; Del-Bel, E.; Raisman-Vozari, R. Novel tactics for neuroprotection in parkinson’s disease: Role of antibiotics, polyphenols and neuropeptides. Prog. Neurobiol. 2017, 155, 120–148. [Google Scholar] [CrossRef] [PubMed]
- Costa, S.L.; Silva, V.D.; 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] [PubMed]
- Mandel, S.; Youdim, M.B. Catechin polyphenols: Neurodegeneration and neuroprotection in neurodegenerative diseases. Free Radic. Biol. Med. 2004, 37, 304–317. [Google Scholar] [CrossRef] [PubMed]
- Costa, C.; Tsatsakis, A.; Mamoulakis, C.; Teodoro, M.; Briguglio, G.; Caruso, E.; Tsoukalas, D.; Margina, D.; Dardiotis, E.; Kouretas, D.; et al. Current evidence on the effect of dietary polyphenols intake on chronic diseases. Food Chem. Toxicol. 2017, 110, 286–299. [Google Scholar] [CrossRef] [PubMed]
- Moosavi, F.; Hosseini, R.; Saso, L.; Firuzi, O. Modulation of neurotrophic signaling pathways by polyphenols. Drug Des. Dev. Ther. 2016, 10, 23–42. [Google Scholar]
- Pan, T.; Jankovic, J.; Le, W. Potential therapeutic properties of green tea polyphenols in parkinson’s disease. Drugs Aging 2003, 20, 711–721. [Google Scholar] [CrossRef] [PubMed]
- Youdim, K.A.; Shukitt-Hale, B.; Joseph, J.A. Flavonoids and the brain: Interactions at the blood-brain barrier and their physiological effects on the central nervous system. Free Radic. Biol. Med. 2004, 37, 1683–1693. [Google Scholar] [CrossRef] [PubMed]
- Youdim, K.A.; Spencer, J.P.; Schroeter, H.; Rice-Evans, C. Dietary flavonoids as potential neuroprotectants. Biol. Chem. 2002, 383, 503–519. [Google Scholar] [CrossRef] [PubMed]
- Vauzour, D.; Vafeiadou, K.; Rodriguez-Mateos, A.; Rendeiro, C.; Spencer, J.P. The neuroprotective potential of flavonoids: A multiplicity of effects. Genes Nutr. 2008, 3, 115–126. [Google Scholar] [CrossRef] [PubMed]
- Zhao, B. Natural antioxidants protect neurons in alzheimer’s disease and parkinson’s disease. Neurochem. Res. 2009, 34, 630–638. [Google Scholar] [CrossRef] [PubMed]
- Weinreb, O.; Amit, T.; Mandel, S.; Youdim, M.B. Neuroprotective molecular mechanisms of (−)-epigallocatechin-3-gallate: A reflective outcome of its antioxidant, iron chelating and neuritogenic properties. Genes Nutr. 2009, 4, 283–296. [Google Scholar] [CrossRef] [PubMed]
- Mandel, S.; Amit, T.; Reznichenko, L.; Weinreb, O.; Youdim, M.B. Green tea catechins as brain-permeable, natural iron chelators-antioxidants for the treatment of neurodegenerative disorders. Mol. Nutr. Food Res. 2006, 50, 229–234. [Google Scholar] [CrossRef] [PubMed]
- Vingill, S.; Connor-Robson, N.; Wade-Martins, R. Are rodent models of parkinson’s disease behaving as they should? Behav Brain Res. 2017. [Google Scholar] [CrossRef] [PubMed]
- Zbarsky, V.; Datla, K.P.; Parkar, S.; Rai, D.K.; Aruoma, O.I.; Dexter, D.T. Neuroprotective properties of the natural phenolic antioxidants curcumin and naringenin but not quercetin and fisetin in a 6-ohda model of parkinson’s disease. Free Radic. Res. 2005, 39, 1119–1125. [Google Scholar] [CrossRef] [PubMed]
- Lou, H.; Jing, X.; Wei, X.; Shi, H.; Ren, D.; Zhang, X. Naringenin protects against 6-ohda-induced neurotoxicity via activation of the nrf2/are signaling pathway. Neuropharmacology 2014, 79, 380–388. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.D.; Jeong, K.H.; Jung, U.J.; Kim, S.R. Naringin treatment induces neuroprotective effects in a mouse model of parkinson’s disease in vivo, but not enough to restore the lesioned dopaminergic system. J. Nutr. Biochem. 2016, 28, 140–146. [Google Scholar] [CrossRef] [PubMed]
- Antunes, M.S.; Goes, A.T.; Boeira, S.P.; Prigol, M.; Jesse, C.R. Protective effect of hesperidin in a model of parkinson’s disease induced by 6-hydroxydopamine in aged mice. Nutrition 2014, 30, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
- Kiasalari, Z.; Khalili, M.; Baluchnejadmojarad, T.; Roghani, M. Protective effect of oral hesperetin against unilateral striatal 6-hydroxydopamine damage in the rat. Neurochem. Res. 2016, 41, 1065–1072. [Google Scholar] [CrossRef] [PubMed]
- Kääriäinen, T.M.; Piltonen, M.; Ossola, B.; Kekki, H.; Lehtonen, S.; Nenonen, T.; Lecklin, A.; Raasmaja, A.; Männistö, P.T. Lack of robust protective effect of quercetin in two types of 6-hydroxydopamine-induced parkinsonian models in rats and dopaminergic cell cultures. Brain Res. 2008, 1203, 149–159. [Google Scholar] [CrossRef] [PubMed]
- Haleagrahara, N.; Siew, C.J.; Mitra, N.K.; Kumari, M. Neuroprotective effect of bioflavonoid quercetin in 6-hydroxydopamine-induced oxidative stress biomarkers in the rat striatum. Neurosci. Lett. 2011, 500, 139–143. [Google Scholar] [CrossRef] [PubMed]
- Khan, M.M.; Raza, S.S.; Javed, H.; Ahmad, A.; Khan, A.; Islam, F.; Safhi, M.M. Rutin protects dopaminergic neurons from oxidative stress in an animal model of parkinson’s disease. Neurotox. Res. 2012, 22, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Baluchnejadmojarad, T.; Jamali-Raeufy, N.; Zabihnejad, S.; Rabiee, N.; Roghani, M. Troxerutin exerts neuroprotection in 6-hydroxydopamine lesion rat model of parkinson’s disease: Possible involvement of pi3k/erβ signaling. Eur. J. Pharmacol. 2017, 801, 72–78. [Google Scholar] [CrossRef] [PubMed]
- Ma, Z.G.; Wang, J.; Jiang, H.; Liu, T.W.; Xie, J.X. Myricetin reduces 6-hydroxydopamine-induced dopamine neuron degeneration in rats. Neuroreport 2007, 18, 1181–1185. [Google Scholar] [CrossRef] [PubMed]
- Baluchnejadmojarad, T.; Roghani, M.; Nadoushan, M.R.; Bagheri, M. Neuroprotective effect of genistein in 6-hydroxydopamine hemi-parkinsonian rat model. Phytother. Res. 2009, 23, 132–135. [Google Scholar] [CrossRef] [PubMed]
- Zhu, G.; Wang, X.; Chen, Y.; Yang, S.; Cheng, H.; Wang, N.; Li, Q. Puerarin protects dopaminergic neurons against 6-hydroxydopamine neurotoxicity via inhibiting apoptosis and upregulating glial cell line-derived neurotrophic factor in a rat model of parkinson’s disease. Planta Med. 2010, 76, 1820–1826. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; He, G.R.; Sun, L.; Lan, X.; Shi, L.L.; Xuan, Z.H.; Du, G.H. Assessment of the treatment effect of baicalein on a model of parkinsonian tremor and elucidation of the mechanism. Life Sci. 2012, 91, 5–13. [Google Scholar] [CrossRef] [PubMed]
- Datla, K.P.; Christidou, M.; Widmer, W.W.; Rooprai, H.K.; Dexter, D.T. Tissue distribution and neuroprotective effects of citrus flavonoid tangeretin in a rat model of parkinson’s disease. Neuroreport 2001, 12, 3871–3875. [Google Scholar] [CrossRef] [PubMed]
- Leaver, K.R.; Allbutt, H.N.; Creber, N.J.; Kassiou, M.; Henderson, J.M. Oral pre-treatment with epigallocatechin gallate in 6-ohda lesioned rats produces subtle symptomatic relief but not neuroprotection. Brain Res. Bull. 2009, 80, 397–402. [Google Scholar] [CrossRef] [PubMed]
- Roghani, M.; Niknam, A.; Jalali-Nadoushan, M.R.; Kiasalari, Z.; Khalili, M.; Baluchnejadmojarad, T. Oral pelargonidin exerts dose-dependent neuroprotection in 6-hydroxydopamine rat model of hemi-parkinsonism. Brain Res. Bull. 2010, 82, 279–283. [Google Scholar] [CrossRef] [PubMed]
- Jin, F.; Wu, Q.; Lu, Y.F.; Gong, Q.H.; Shi, J.S. Neuroprotective effect of resveratrol on 6-ohda-induced parkinson’s disease in rats. Eur. J. Pharmacol. 2008, 600, 78–82. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Xu, H.; Fu, Q.; Ma, R.; Xiang, J. Protective effect of resveratrol derived from polygonum cuspidatum and its liposomal form on nigral cells in parkinsonian rats. J. Neurol. Sci. 2011, 304, 29–34. [Google Scholar] [CrossRef] [PubMed]
- Khan, M.M.; Ahmad, A.; Ishrat, T.; Khan, M.B.; Hoda, M.N.; Khuwaja, G.; Raza, S.S.; Khan, A.; Javed, H.; Vaibhav, K.; et al. Resveratrol attenuates 6-hydroxydopamine-induced oxidative damage and dopamine depletion in rat model of parkinson’s disease. Brain Res. 2010, 1328, 139–151. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Zhang, D.Q.; Liao, Z.; Wang, B.; Gong, S.; Wang, C.; Zhang, M.Z.; Wang, G.H.; Cai, H.; Liao, F.F.; et al. Anti-oxidant polydatin (piceid) protects against substantia nigral motor degeneration in multiple rodent models of parkinson’s disease. Mol. Neurodegener. 2015, 10, 4. [Google Scholar] [CrossRef] [PubMed]
- Du, X.X.; Xu, H.M.; Jiang, H.; Song, N.; Wang, J.; Xie, J.X. Curcumin protects nigral dopaminergic neurons by iron-chelation in the 6-hydroxydopamine rat model of parkinson’s disease. Neurosci. Bull. 2012, 28, 253–258. [Google Scholar] [CrossRef] [PubMed]
- Tripanichkul, W.; Jaroensuppaperch, E.O. Ameliorating effects of curcumin on 6-ohda-induced dopaminergic denervation, glial response, and sod1 reduction in the striatum of hemiparkinsonian mice. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 1360–1368. [Google Scholar] [PubMed]
- Wu, C.R.; Tsai, C.W.; Chang, S.W.; Lin, C.Y.; Huang, L.C. Carnosic acid protects against 6-hydroxydopamine-induced neurotoxicity in in vivo and in vitro model of parkinson’s disease: Involvement of antioxidative enzymes induction. Chem. Biol. Interact. 2015, 225, 40–46. [Google Scholar] [CrossRef] [PubMed]
- Zare, K.; Eidi, A.; Roghani, M.; Rohani, A.H. The neuroprotective potential of sinapic acid in the 6-hydroxydopamine-induced hemi-parkinsonian rat. Metab. Brain Dis. 2015, 30, 205–213. [Google Scholar] [CrossRef] [PubMed]
- Farbood, Y.; Sarkaki, A.; Dolatshahi, M.; Taqhi Mansouri, S.M.; Khodadadi, A. Ellagic acid protects the brain against 6-hydroxydopamine induced neuroinflammation in a rat model of parkinson’s disease. Basic Clin. Neurosci. 2015, 6, 83–89. [Google Scholar] [PubMed]
- Lastres-Becker, I.; Molina-Holgado, F.; Ramos, J.A.; Mechoulam, R.; Fernández-Ruiz, J. Cannabinoids provide neuroprotection against 6-hydroxydopamine toxicity in vivo and in vitro: Relevance to parkinson’s disease. Neurobiol. Dis. 2005, 19, 96–107. [Google Scholar] [CrossRef] [PubMed]
- Levites, Y.; Weinreb, O.; Maor, G.; Youdim, M.B.; Mandel, S. Green tea polyphenol (-)-epigallocatechin-3-gallate prevents n-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J. Neurochem. 2001, 78, 1073–1082. [Google Scholar] [CrossRef] [PubMed]
- Mandel, S.; Maor, G.; Youdim, M.B. Iron and alpha-synuclein in the substantia nigra of mptp-treated mice: Effect of neuroprotective drugs r-apomorphine and green tea polyphenol (-)-epigallocatechin-3-gallate. J. Mol. Neurosci. 2004, 24, 401–416. [Google Scholar] [CrossRef]
- Choi, J.Y.; Park, C.S.; Kim, D.J.; Cho, M.H.; Jin, B.K.; Pie, J.E.; Chung, W.G. Prevention of nitric oxide-mediated 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinson’s disease in mice by tea phenolic epigallocatechin 3-gallate. Neurotoxicology 2002, 23, 367–374. [Google Scholar] [CrossRef]
- Xu, Q.; Langley, M.; Kanthasamy, A.G.; Reddy, M.B. Epigallocatechin gallate has a neurorescue effect in a mouse model of parkinson disease. J. Nutr. 2017, 147, 1926–1931. [Google Scholar] [CrossRef] [PubMed]
- Reznichenko, L.; Kalfon, L.; Amit, T.; Youdim, M.B.; Mandel, S.A. Low dosage of rasagiline and epigallocatechin gallate synergistically restored the nigrostriatal axis in mptp-induced parkinsonism. Neurodegener. Dis. 2010, 7, 219–231. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.S.; Kim, J.M.; O, J.J.; Jeon, B.S. Inhibition of inducible nitric oxide synthase expression and cell death by (-)-epigallocatechin-3-gallate, a green tea catechin, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of parkinson’s disease. J. Clin. Neurosci. 2010, 17, 1165–1168. [Google Scholar] [CrossRef] [PubMed]
- Zhou, T.; Zhu, M.; Liang, Z. (-)-epigallocatechin-3-gallate modulates peripheral immunity in the mptp-induced mouse model of parkinson’s disease. Mol. Med. Rep. 2018, 17, 4883–4888. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.G.; Ju, M.S.; Ha, S.K.; Lee, H.; Kim, S.Y.; Oh, M.S. Acacetin protects dopaminergic cells against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neuroinflammation in vitro and in vivo. Biol. Pharm. Bull. 2012, 35, 1287–1294. [Google Scholar] [CrossRef] [PubMed]
- Xue, X.; Liu, H.; Qi, L.; Li, X.; Guo, C.; Gong, D.; Qu, H. Baicalein ameliorated the upregulation of striatal glutamatergic transmission in the mice model of parkinson’s disease. Brain Res. Bull. 2014, 103, 54–59. [Google Scholar] [CrossRef] [PubMed]
- Lee, E.; Park, H.R.; Ji, S.T.; Lee, Y.; Lee, J. Baicalein attenuates astroglial activation in the 1-methyl-4-phenyl-1,2,3,4-tetrahydropyridine-induced parkinson’s disease model by downregulating the activations of nuclear factor-κb, erk, and jnk. J. Neurosci. Res. 2014, 92, 130–139. [Google Scholar] [CrossRef] [PubMed]
- Jang, S.W.; Liu, X.; Yepes, M.; Shepherd, K.R.; Miller, G.W.; Liu, Y.; Wilson, W.D.; Xiao, G.; Blanchi, B.; Sun, Y.E.; et al. A selective trkb agonist with potent neurotrophic activities by 7,8-dihydroxyflavone. Proc. Natl. Acad. Sci. USA 2010, 107, 2687–2692. [Google Scholar] [CrossRef] [PubMed]
- Takano, K.; Tabata, Y.; Kitao, Y.; Murakami, R.; Suzuki, H.; Yamada, M.; Iinuma, M.; Yoneda, Y.; Ogawa, S.; Hori, O. Methoxyflavones protect cells against endoplasmic reticulum stress and neurotoxin. Am. J. Physiol. Cell Physiol. 2007, 292, C353–C361. [Google Scholar] [CrossRef] [PubMed]
- Jeong, K.H.; Jeon, M.T.; Kim, H.D.; Jung, U.J.; Jang, M.C.; Chu, J.W.; Yang, S.J.; Choi, I.Y.; Choi, M.S.; Kim, S.R. Nobiletin protects dopaminergic neurons in the 1-methyl-4-phenylpyridinium-treated rat model of parkinson’s disease. J. Med. Food 2015, 18, 409–414. [Google Scholar] [CrossRef] [PubMed]
- Yabuki, Y.; Ohizumi, Y.; Yokosuka, A.; Mimaki, Y.; Fukunaga, K. Nobiletin treatment improves motor and cognitive deficits seen in mptp-induced parkinson model mice. Neuroscience 2014, 259, 126–141. [Google Scholar] [CrossRef] [PubMed]
- Leem, E.; Nam, J.H.; Jeon, M.T.; Shin, W.H.; Won, S.Y.; Park, S.J.; Choi, M.S.; Jin, B.K.; Jung, U.J.; Kim, S.R. Naringin protects the nigrostriatal dopaminergic projection through induction of gdnf in a neurotoxin model of parkinson’s disease. J. Nutr. Biochem. 2014, 25, 801–806. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Pu, X.P. Neuroprotective effect of kaempferol against a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse model of parkinson’s disease. Biol. Pharm. Bull. 2011, 34, 1291–1296. [Google Scholar] [CrossRef] [PubMed]
- Lv, C.; Hong, T.; Yang, Z.; Zhang, Y.; Wang, L.; Dong, M.; Zhao, J.; Mu, J.; Meng, Y. Effect of quercetin in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-induced mouse model of parkinson’s disease. Evid. Based Complement. Alternat. Med. 2012, 2012, 928643. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.T.; Cao, X.B.; Xiong, N.; Wang, H.C.; Huang, J.S.; Sun, S.G.; Wang, T. Morin exerts neuroprotective actions in parkinson disease models in vitro and in vivo. Acta Pharmacol. Sin. 2010, 31, 900–906. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.; Park, H.R.; Chun, H.J.; Lee, J. Silibinin prevents dopaminergic neuronal loss in a mouse model of parkinson’s disease via mitochondrial stabilization. J. Neurosci. Res. 2015, 93, 755–765. [Google Scholar] [CrossRef] [PubMed]
- Jung, U.J.; Jeon, M.T.; Choi, M.S.; Kim, S.R. Silibinin attenuates mpp⁺-induced neurotoxicity in the substantia nigra in vivo. J. Med. Food 2014, 17, 599–605. [Google Scholar] [CrossRef] [PubMed]
- Geed, M.; Garabadu, D.; Ahmad, A.; Krishnamurthy, S. Silibinin pretreatment attenuates biochemical and behavioral changes induced by intrastriatal mpp+ injection in rats. Pharmacol. Biochem. Behav. 2014, 117, 92–103. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.X.; Chen, W.F.; Xie, J.X.; Wong, M.S. Neuroprotective effects of genistein on dopaminergic neurons in the mice model of parkinson’s disease. Neurosci. Res. 2008, 60, 156–161. [Google Scholar] [CrossRef] [PubMed]
- Kumar, H.; Kim, I.S.; More, S.V.; Kim, B.W.; Bahk, Y.Y.; Choi, D.K. Gastrodin protects apoptotic dopaminergic neurons in a toxin-induced parkinson’s disease model. Evid. Based Complement. Alternat. Med. 2013, 2013, 514095. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Jing, H.; Yang, H.; Liu, Z.; Guo, H.; Chai, L.; Hu, L. Tanshinone i selectively suppresses pro-inflammatory genes expression in activated microglia and prevents nigrostriatal dopaminergic neurodegeneration in a mouse model of parkinson’s disease. J. Ethnopharmacol. 2015, 164, 247–255. [Google Scholar] [CrossRef] [PubMed]
- Nagarajan, S.; Chellappan, D.R.; Chinnaswamy, P.; Thulasingam, S. Ferulic acid pretreatment mitigates mptp-induced motor impairment and histopathological alterations in c57bl/6 mice. Pharm. Biol. 2015, 53, 1591–1601. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.L.; Qu, R.; Zhang, J.; Li, L.F.; Ma, S.P. Neuroprotective effects of madecassoside in early stage of parkinson’s disease induced by mptp in rats. Fitoterapia 2013, 90, 112–118. [Google Scholar] [CrossRef] [PubMed]
- Vajragupta, O.; Boonchoong, P.; Watanabe, H.; Tohda, M.; Kummasud, N.; Sumanont, Y. Manganese complexes of curcumin and its derivatives: Evaluation for the radical scavenging ability and neuroprotective activity. Free Radic. Biol. Med. 2003, 35, 1632–1644. [Google Scholar] [CrossRef] [PubMed]
- Rajeswari, A.; Sabesan, M. Inhibition of monoamine oxidase-b by the polyphenolic compound, curcumin and its metabolite tetrahydrocurcumin, in a model of parkinson’s disease induced by mptp neurodegeneration in mice. Inflammopharmacology 2008, 16, 96–99. [Google Scholar] [CrossRef] [PubMed]
- Ojha, R.P.; Rastogi, M.; Devi, B.P.; Agrawal, A.; Dubey, G.P. Neuroprotective effect of curcuminoids against inflammation-mediated dopaminergic neurodegeneration in the mptp model of parkinson’s disease. J. Neuroimmune Pharmacol. 2012, 7, 609–618. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.C.; Zhou, Y.C.; Chen, Y.; Zhu, Y.G.; Fang, F.; Chen, L.M. Ginsenoside rg1 reduces mptp-induced substantia nigra neuron loss by suppressing oxidative stress. Acta Pharmacol. Sin. 2005, 26, 56–62. [Google Scholar] [CrossRef] [PubMed]
- Anandhan, A.; Tamilselvam, K.; Radhiga, T.; Rao, S.; Essa, M.M.; Manivasagam, T. Theaflavin, a black tea polyphenol, protects nigral dopaminergic neurons against chronic mptp/probenecid induced parkinson’s disease. Brain Res. 2012, 1433, 104–113. [Google Scholar] [CrossRef] [PubMed]
- Anandhan, A.; Janakiraman, U.; Manivasagam, T. Theaflavin ameliorates behavioral deficits, biochemical indices and monoamine transporters expression against subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (mptp)-induced mouse model of parkinson’s disease. Neuroscience 2012, 218, 257–267. [Google Scholar] [CrossRef] [PubMed]
- Anandhan, A.; Essa, M.M.; Manivasagam, T. Therapeutic attenuation of neuroinflammation and apoptosis by black tea theaflavin in chronic mptp/probenecid model of parkinson’s disease. Neurotox. Res. 2013, 23, 166–173. [Google Scholar] [CrossRef] [PubMed]
- Patil, S.P.; Jain, P.D.; Sancheti, J.S.; Ghumatkar, P.J.; Tambe, R.; Sathaye, S. Neuroprotective and neurotrophic effects of apigenin and luteolin in mptp induced parkinsonism in mice. Neuropharmacology 2014, 86, 192–202. [Google Scholar] [CrossRef] [PubMed]
- Rekha, K.R.; Selvakumar, G.P.; Sivakamasundari, R.I. Effects of syringic acid on chronic mptp/probenecid induced motor dysfunction, dopaminergic markers expression and neuroinflammation in c57bl/6 mice. Biomed. Aging Pathol. 2014, 4, 95–104. [Google Scholar] [CrossRef]
- Karuppagounder, S.S.; Madathil, S.K.; Pandey, M.; Haobam, R.; Rajamma, U.; Mohanakumar, K.P. Quercetin up-regulates mitochondrial complex-i activity to protect against programmed cell death in rotenone model of parkinson’s disease in rats. Neuroscience 2013, 236, 136–148. [Google Scholar] [CrossRef] [PubMed]
- Cui, Q.; Li, X.; Zhu, H. Curcumin ameliorates dopaminergic neuronal oxidative damage via activation of the akt/nrf2 pathway. Mol. Med. Rep. 2016, 13, 1381–1388. [Google Scholar] [CrossRef] [PubMed]
- Ojha, S.; Javed, H.; Azimullah, S.; Abul Khair, S.B.; Haque, M.E. Neuroprotective potential of ferulic acid in the rotenone model of parkinson’s disease. Drug Des. Dev. Ther. 2015, 9, 5499–5510. [Google Scholar]
- Sonia Angeline, M.; Sarkar, A.; Anand, K.; Ambasta, R.K.; Kumar, P. Sesamol and naringenin reverse the effect of rotenone-induced pd rat model. Neuroscience 2013, 254, 379–394. [Google Scholar] [CrossRef] [PubMed]
- Al-Amri, J.S.; Hagras, M.M.; Mohamed I, M. Effect of epigallocatechin-3-gallate on inflammatory mediators release in lps-induced parkinson’s disease in rats. Indian J. Exp. Biol. 2013, 51, 357–362. [Google Scholar] [PubMed]
- Mansouri, Z.; Sabetkasaei, M.; Moradi, F.; Masoudnia, F.; Ataie, A. Curcumin has neuroprotection effect on homocysteine rat model of parkinson. J. Mol. Neurosci. 2012, 47, 234–242. [Google Scholar] [CrossRef] [PubMed]
- Grünblatt, E.; Mandel, S.; Youdim, M.B. Neuroprotective strategies in parkinson’s disease using the models of 6-hydroxydopamine and mptp. Ann. N. Y. Acad. Sci. 2000, 899, 262–273. [Google Scholar] [CrossRef] [PubMed]
- Shahpiri, Z.; Bahramsoltani, R.; Hosein Farzaei, M.; Farzaei, F.; Rahimi, R. Phytochemicals as future drugs for parkinson’s disease: A comprehensive review. Rev. Neurosci. 2016, 27, 651–668. [Google Scholar] [CrossRef] [PubMed]
- Xia, N.; Fang, F.; Zhang, P.; Cui, J.; Tep-Cullison, C.; Hamerley, T.; Lee, H.J.; Palmer, T.; Bothner, B.; Lee, J.H.; et al. A knockin reporter allows purification and characterization of mda neurons from heterogeneous populations. Cell Rep. 2017, 18, 2533–2546. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.D.; Jeong, K.H.; Jung, U.J.; Kim, S.R. Myricitrin ameliorates 6-hydroxydopamine-induced dopaminergic neuronal loss in the substantia nigra of mouse brain. J. Med. Food 2016, 19, 374–382. [Google Scholar] [CrossRef] [PubMed]
- Jung, U.J.; Kim, S.R. Beneficial effects of flavonoids against parkinson’s disease. J. Med. Food 2018. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Thong, P.S.; Lee, T.; Leong, S.K.; Mao, B.Y.; Dong, F.; Watt, F. Dopaminergic cell death precedes iron elevation in mptp-injected monkeys. Free Radic. Biol. Med. 2003, 35, 540–547. [Google Scholar] [CrossRef]
- Salazar, J.; Mena, N.; Hunot, S.; Prigent, A.; Alvarez-Fischer, D.; Arredondo, M.; Duyckaerts, C.; Sazdovitch, V.; Zhao, L.; Garrick, L.M.; et al. Divalent metal transporter 1 (dmt1) contributes to neurodegeneration in animal models of parkinson’s disease. Proc. Natl. Acad. Sci. USA 2008, 105, 18578–18583. [Google Scholar] [CrossRef] [PubMed]
- Lv, Z.; Jiang, H.; Xu, H.; Song, N.; Xie, J. Increased iron levels correlate with the selective nigral dopaminergic neuron degeneration in parkinson’s disease. J. Neural Transm (Vienna) 2011, 118, 361–369. [Google Scholar] [CrossRef] [PubMed]
- Lau, Y.S.; Patki, G.; Das-Panja, K.; Le, W.D.; Ahmad, S.O. Neuroprotective effects and mechanisms of exercise in a chronic mouse model of parkinson’s disease with moderate neurodegeneration. Eur. J. Neurosci. 2011, 33, 1264–1274. [Google Scholar] [CrossRef] [PubMed]
- Zaitone, S.A.; Abo-Elmatty, D.M.; Shaalan, A.A. Acetyl-l-carnitine and α-lipoic acid affect rotenone-induced damage in nigral dopaminergic neurons of rat brain, implication for parkinson’s disease therapy. Pharmacol. Biochem. Behav. 2012, 100, 347–360. [Google Scholar] [CrossRef] [PubMed]
- Jiang, P.; Dickson, D.W. Parkinson’s disease: Experimental models and reality. Acta Neuropathol. 2018, 135, 13–32. [Google Scholar] [CrossRef] [PubMed]
- Rein, M.J.; Renouf, M.; Cruz-Hernandez, C.; Actis-Goretta, L.; Thakkar, S.K.; da Silva Pinto, M. Bioavailability of bioactive food compounds: A challenging journey to bioefficacy. Br. J. Clin. Pharmacol. 2013, 75, 588–602. [Google Scholar] [CrossRef] [PubMed]
- Wahlqvist, M.L. Food structure is critical for optimal health. Food Funct. 2016, 7, 1245–1250. [Google Scholar] [CrossRef] [PubMed]
- Wu, W.R.; Zhu, X.Z. Involvement of monoamine oxidase inhibition in neuroprotective and neurorestorative effects of ginkgo biloba extract against mptp-induced nigrostriatal dopaminergic toxicity in c57 mice. Life Sci. 1999, 65, 157–164. [Google Scholar] [CrossRef]
- Datla, K.P.; Bennett, R.D.; Zbarsky, V.; Ke, B.; Liang, Y.F.; Higa, T.; Bahorun, T.; Aruoma, O.I.; Dexter, D.T. The antioxidant drink effective microorganism-x (em-x) pre-treatment attenuates the loss of nigrostriatal dopaminergic neurons in 6-hydroxydopamine-lesion rat model of parkinson’s disease. J. Pharm. Pharmacol. 2004, 56, 649–654. [Google Scholar] [CrossRef] [PubMed]
- Ahlemeyer, B.; Krieglstein, J. Neuroprotective effects of ginkgo biloba extract. Cell. Mol. Life Sci. 2003, 60, 1779–1792. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.S.; Lee, J.I.; Lee, W.Y.; Kim, S.E. Neuroprotective effect of ginkgo biloba l. Extract in a rat model of parkinson’s disease. Phytother. Res. 2004, 18, 663–666. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.F.; Wu, Q.; Sun, A.S.; Huang, X.N.; Shi, J.S. Protective effect and mechanism of ginkgo biloba leaf extracts for parkinson disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Acta Pharmacol. Sin. 2001, 22, 1089–1093. [Google Scholar] [PubMed]
- Ahmad, M.; Saleem, S.; Ahmad, A.S.; Yousuf, S.; Ansari, M.A.; Khan, M.B.; Ishrat, T.; Chaturvedi, R.K.; Agrawal, A.K.; Islam, F. Ginkgo biloba affords dose-dependent protection against 6-hydroxydopamine-induced parkinsonism in rats: Neurobehavioural, neurochemical and immunohistochemical evidences. J. Neurochem. 2005, 93, 94–104. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.; Yan, J.; Yang, T.; Yang, X.; Bezard, E.; Zhao, B. Protective effects of green tea polyphenols in the 6-ohda rat model of parkinson’s disease through inhibition of ros-no pathway. Biol. Psychiatry 2007, 62, 1353–1362. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.; Wang, T.; Yue, F.; Li, X.; Wang, P.; Li, Y.; Chan, P.; Yu, S. Tea polyphenols alleviate motor impairments, dopaminergic neuronal injury, and cerebral α-synuclein aggregation in mptp-intoxicated parkinsonian monkeys. Neuroscience 2015, 286, 383–392. [Google Scholar] [CrossRef] [PubMed]
- Pérez-H, J.; Carrillo-S, C.; García, E.; Ruiz-Mar, G.; Pérez-Tamayo, R.; Chavarría, A. Neuroprotective effect of silymarin in a mptp mouse model of parkinson’s disease. Toxicology 2014, 319, 38–43. [Google Scholar] [CrossRef] [PubMed]
- Singhal, N.K.; Srivastava, G.; Patel, D.K.; Jain, S.K.; Singh, M.P. Melatonin or silymarin reduces maneb- and paraquat-induced parkinson’s disease phenotype in the mouse. J. Pineal Res. 2011, 50, 97–109. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Matsumoto, K.; Murakami, Y.; Tezuka, Y.; Wu, Y.; Kadota, S. Neuroprotective effects of polygonum multiflorum on nigrostriatal dopaminergic degeneration induced by paraquat and maneb in mice. Pharmacol. Biochem. Behav. 2005, 82, 345–352. [Google Scholar] [CrossRef] [PubMed]
- Datla, K.P.; Zbarsky, V.; Rai, D.; Parkar, S.; Osakabe, N.; Aruoma, O.I.; Dexter, D.T. Short-term supplementation with plant extracts rich in flavonoids protect nigrostriatal dopaminergic neurons in a rat model of parkinson’s disease. J. Am. Coll. Nutr. 2007, 26, 341–349. [Google Scholar] [CrossRef] [PubMed]
- Moreira, E.L.; Rial, D.; Aguiar, A.S.; Figueiredo, C.P.; Siqueira, J.M.; DalBó, S.; Horst, H.; de Oliveira, J.; Mancini, G.; dos Santos, T.S.; et al. Proanthocyanidin-rich fraction from croton celtidifolius baill confers neuroprotection in the intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine rat model of parkinson’s disease. J. Neural Transm (Vienna) 2010, 117, 1337–1351. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.G.; Park, G.; Kim, H.G.; Oh, D.S.; Kim, H.; Oh, M.S. In vitro and in vivo neuroprotective effects of walnut (juglandis semen) in models of parkinson’s disease. Int. J. Mol. Sci. 2016, 17. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Li, G.; Szeto, S.S.W.; Chong, C.M.; Quan, Q.; Huang, C.; Cui, W.; Guo, B.; Wang, Y.; Han, Y.; et al. Examining the neuroprotective effects of protocatechuic acid and chrysin on in vitro and in vivo models of parkinson disease. Free Radic. Biol. Med. 2015, 84, 331–343. [Google Scholar] [CrossRef] [PubMed]
- Gao, X.; Cassidy, A.; Schwarzschild, M.A.; Rimm, E.B.; Ascherio, A. Habitual intake of dietary flavonoids and risk of parkinson disease. Neurology 2012, 78, 1138–1145. [Google Scholar] [CrossRef] [PubMed]
- Chan, D.K.; Mellick, G.D.; Hung, W.T.; Woo, J. Genetic and environmental risk factors and their interactions for parkinson’s disease in a chinese population. J. Clin Neurosci. 2003, 10, 313–315. [Google Scholar] [CrossRef]
- Checkoway, H.; Powers, K.; Smith-Weller, T.; Franklin, G.M.; Longstreth, W.T.; Swanson, P.D. Parkinson’s disease risks associated with cigarette smoking, alcohol consumption, and caffeine intake. Am. J. Epidemiol. 2002, 155, 732–738. [Google Scholar] [CrossRef] [PubMed]
- Tan, E.K.; Tan, C.; Fook-Chong, S.M.; Lum, S.Y.; Chai, A.; Chung, H.; Shen, H.; Zhao, Y.; Teoh, M.L.; Yih, Y.; et al. Dose-dependent protective effect of coffee, tea, and smoking in parkinson’s disease: A study in ethnic chinese. J. Neurol. Sci. 2003, 216, 163–167. [Google Scholar] [CrossRef] [PubMed]
- Tan, L.C.; Koh, W.P.; Yuan, J.M.; Wang, R.; Au, W.L.; Tan, J.H.; Tan, E.K.; Yu, M.C. Differential effects of black versus green tea on risk of parkinson’s disease in the singapore chinese health study. Am. J. Epidemiol. 2008, 167, 553–560. [Google Scholar] [CrossRef] [PubMed]
- Kandinov, B.; Giladi, N.; Korczyn, A.D. Smoking and tea consumption delay onset of parkinson’s disease. Parkinsonism Relat. Disord. 2009, 15, 41–46. [Google Scholar] [CrossRef] [PubMed]
- Mak, J.C. Potential role of green tea catechins in various disease therapies: Progress and promise. Clin. Exp. Pharmacol. Physiol. 2012, 39, 265–273. [Google Scholar] [CrossRef] [PubMed]
- Li, F.J.; Ji, H.F.; Shen, L. A meta-analysis of tea drinking and risk of parkinson’s disease. Sci. World J. 2012, 2012, 923464. [Google Scholar] [CrossRef] [PubMed]
- Dutta, D.; Mohanakumar, K.P. Tea and parkinson’s disease: Constituents of tea synergize with antiparkinsonian drugs to provide better therapeutic benefits. Neurochem. Int. 2015, 89, 181–190. [Google Scholar] [CrossRef] [PubMed]
- Youdim, K.A.; Dobbie, M.S.; Kuhnle, G.; Proteggente, A.R.; Abbott, N.J.; Rice-Evans, C. Interaction between flavonoids and the blood-brain barrier: In vitro studies. J. Neurochem. 2003, 85, 180–192. [Google Scholar] [CrossRef] [PubMed]
- Youdim, K.A.; Qaiser, M.Z.; Begley, D.J.; Rice-Evans, C.A.; Abbott, N.J. Flavonoid permeability across an in situ model of the blood-brain barrier. Free Radic. Biol. Med. 2004, 36, 592–604. [Google Scholar] [CrossRef] [PubMed]
- de Boer, V.C.; Dihal, A.A.; van der Woude, H.; Arts, I.C.; Wolffram, S.; Alink, G.M.; Rietjens, I.M.; Keijer, J.; Hollman, P.C. Tissue distribution of quercetin in rats and pigs. J. Nutr. 2005, 135, 1718–1725. [Google Scholar] [CrossRef] [PubMed]
- Peng, H.W.; Cheng, F.C.; Huang, Y.T.; Chen, C.F.; Tsai, T.H. Determination of naringenin and its glucuronide conjugate in rat plasma and brain tissue by high-performance liquid chromatography. J. Chromatogr. B Biomed. Sci. Appl. 1998, 714, 369–374. [Google Scholar] [CrossRef]
- Tsai, T.H.; Chen, Y.F. Determination of unbound hesperetin in rat blood and brain by microdialysis coupled to microbore liquid chromatography. J. Food Drug Anal. 2000, 8, 331–336. [Google Scholar]
- Suganuma, M.; Okabe, S.; Oniyama, M.; Tada, Y.; Ito, H.; Fujiki, H. Wide distribution of (3h)(-)-epigallocatechin gallate, a cancer preventive tea polyphenol, in mouse tissue. Carcinogenesis 1998, 19, 1771–1776. [Google Scholar] [CrossRef] [PubMed]
- Abd El Mohsen, M.M.; Kuhnle, G.; Rechner, A.R.; Schroeter, H.; Rose, S.; Jenner, P.; Rice-Evans, C.A. Uptake and metabolism of epicatechin and its access to the brain after oral ingestion. Free Radic. Biol. Med. 2002, 33, 1693–1702. [Google Scholar] [CrossRef]
- Wang, J.; Ferruzzi, M.G.; Ho, L.; Blount, J.; Janle, E.M.; Gong, B.; Pan, Y.; Gowda, G.A.; Raftery, D.; Arrieta-Cruz, I.; et al. Brain-targeted proanthocyanidin metabolites for alzheimer’s disease treatment. J. Neurosci. 2012, 32, 5144–5150. [Google Scholar] [CrossRef] [PubMed]
- Talavéra, S.; Felgines, C.; Texier, O.; Besson, C.; Gil-Izquierdo, A.; Lamaison, J.L.; Rémésy, C. Anthocyanin metabolism in rats and their distribution to digestive area, kidney, and brain. J. Agric. Food Chem. 2005, 53, 3902–3908. [Google Scholar] [CrossRef] [PubMed]
- Andres-Lacueva, C.; Shukitt-Hale, B.; Galli, R.L.; Jauregui, O.; Lamuela-Raventos, R.M.; Joseph, J.A. Anthocyanins in aged blueberry-fed rats are found centrally and may enhance memory. Nutr. Neurosci. 2005, 8, 111–120. [Google Scholar] [CrossRef] [PubMed]
- Kalt, W.; Blumberg, J.B.; McDonald, J.E.; Vinqvist-Tymchuk, M.R.; Fillmore, S.A.; Graf, B.A.; O’Leary, J.M.; Milbury, P.E. Identification of anthocyanins in the liver, eye, and brain of blueberry-fed pigs. J. Agric. Food Chem. 2008, 56, 705–712. [Google Scholar] [CrossRef] [PubMed]
- Zhao, W.; Wang, J.; Bi, W.; Ferruzzi, M.; Yemul, S.; Freire, D.; Mazzola, P.; Ho, L.; Dubner, L.; Pasinetti, G.M. Novel application of brain-targeting polyphenol compounds in sleep deprivation-induced cognitive dysfunction. Neurochem. Int. 2015, 89, 191–197. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.Y.; Kritchevsky, J.; Hargett, K.; Feller, K.; Klobusnik, R.; Song, B.J.; Cooper, B.; Jouni, Z.; Ferruzzi, M.G.; Janle, E.M. Plasma bioavailability and regional brain distribution of polyphenols from apple/grape seed and bilberry extracts in a young swine model. Mol. Nutr. Food Res. 2015, 59, 2432–2447. [Google Scholar] [CrossRef] [PubMed]
- Passamonti, S.; Vrhovsek, U.; Vanzo, A.; Mattivi, F. Fast access of some grape pigments to the brain. J. Agric. Food Chem. 2005, 53, 7029–7034. [Google Scholar] [CrossRef] [PubMed]
- El Mohsen, M.A.; Marks, J.; Kuhnle, G.; Moore, K.; Debnam, E.; Kaila Srai, S.; Rice-Evans, C.; Spencer, J.P. Absorption, tissue distribution and excretion of pelargonidin and its metabolites following oral administration to rats. Br. J. Nutr. 2006, 95, 51–58. [Google Scholar] [CrossRef] [PubMed]
- Koutsos, A.; Tuohy, K.M.; Lovegrove, J.A. Apples and cardiovascular health—Is the gut microbiota a core consideration? Nutrients 2015, 7, 3959–3998. [Google Scholar] [CrossRef] [PubMed]
- Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
- Espín, J.C.; González-Sarrías, A.; Tomás-Barberán, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly)phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef] [PubMed]
- Requena, T.; Martínez-Cuesta, M.C.; Peláez, C. Diet and microbiota linked in health and disease. Food Funct. 2018, 9, 688–704. [Google Scholar] [CrossRef] [PubMed]
- Selma, M.V.; Beltrán, D.; Luna, M.C.; Romo-Vaquero, M.; García-Villalba, R.; Mira, A.; Espín, J.C.; Tomás-Barberán, F.A. Isolation of human intestinal bacteria capable of producing the bioactive metabolite isourolithin a from ellagic acid. Front. Microbiol. 2017, 8, 1521. [Google Scholar] [CrossRef] [PubMed]
- Yuan, T.; Ma, H.; Liu, W.; Niesen, D.B.; Shah, N.; Crews, R.; Rose, K.N.; Vattem, D.A.; Seeram, N.P. Pomegranate’s neuroprotective effects against alzheimer’s disease are mediated by urolithins, its ellagitannin-gut microbial derived metabolites. ACS Chem. Neurosci. 2016, 7, 26–33. [Google Scholar] [CrossRef] [PubMed]
- Verzelloni, E.; Pellacani, C.; Tagliazucchi, D.; Tagliaferri, S.; Calani, L.; Costa, L.G.; Brighenti, F.; Borges, G.; Crozier, A.; Conte, A.; et al. Antiglycative and neuroprotective activity of colon-derived polyphenol catabolites. Mol. Nutr. Food Res. 2011, 55 (Suppl. 1), S35–S43. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; Ma, H.; Frost, L.; Yuan, T.; Dain, J.A.; Seeram, N.P. Pomegranate phenolics inhibit formation of advanced glycation endproducts by scavenging reactive carbonyl species. Food Funct. 2014, 5, 2996–3004. [Google Scholar] [CrossRef] [PubMed]
- Bookheimer, S.Y.; Renner, B.A.; Ekstrom, A.; Li, Z.; Henning, S.M.; Brown, J.A.; Jones, M.; Moody, T.; Small, G.W. Pomegranate juice augments memory and fmri activity in middle-aged and older adults with mild memory complaints. Evid. Based Complement. Alternat. Med. 2013, 2013, 946298. [Google Scholar] [CrossRef] [PubMed]
- Kerr, J.S.; Adriaanse, B.A.; Greig, N.H.; Mattson, M.P.; Cader, M.Z.; Bohr, V.A.; Fang, E.F. Mitophagy and alzheimer’s disease: Cellular and molecular mechanisms. Trends Neurosci. 2017, 40, 151–166. [Google Scholar] [CrossRef] [PubMed]
- Ryu, D.; Mouchiroud, L.; Andreux, P.A.; Katsyuba, E.; Moullan, N.; Nicolet-Dit-Félix, A.A.; Williams, E.G.; Jha, P.; Lo Sasso, G.; Huzard, D.; et al. Urolithin a induces mitophagy and prolongs lifespan in c. Elegans and increases muscle function in rodents. Nat. Med. 2016, 22, 879–888. [Google Scholar] [CrossRef] [PubMed]
- Gasperotti, M.; Passamonti, S.; Tramer, F.; Masuero, D.; Guella, G.; Mattivi, F.; Vrhovsek, U. Fate of microbial metabolites of dietary polyphenols in rats: Is the brain their target destination? ACS Chem. Neurosci. 2015, 6, 1341–1352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seeram, N.P.; Zhang, Y.J.; Sartipipour, M.; Henning, S.M.; Lee, R.P.; Harris, D.M.; Moro, A.; Heber, D. Pharmacokinetics and tissue disposition of urolithin a, an ellagitannin-derived metabolite, in mice. FASEB J. 2007, 21, A1081. [Google Scholar]
- DaSilva, N.A.; Nahar, P.P.; Ma, H.; Eid, A.; Wei, Z.; Meschwitz, S.; Zawia, N.H.; Slitt, A.L.; Seeram, N.P. Pomegranate ellagitannin-gut microbial-derived metabolites, urolithins, inhibit neuroinflammation in vitro. Nutr. Neurosci. 2017, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Thakur, P.; Nehru, B. Inhibition of neuroinflammation and mitochondrial dysfunctions by carbenoxolone in the rotenone model of parkinson’s disease. Mol. Neurobiol. 2015, 51, 209–219. [Google Scholar] [CrossRef] [PubMed]
- Dhanasekaran, D.N.; Reddy, E.P. Jnk signaling in apoptosis. Oncogene 2008, 27, 6245–6251. [Google Scholar] [CrossRef] [PubMed]
Compound, Route and Dosage | Species (Sex) | Neurotoxin | Neuroprotective Effects | References |
---|---|---|---|---|
Naringenin (50 mg/kg; p.o.) 4-day pre-treatment | Sprague- Dawley rats (male) | 6-OHDA | ↑TH-positive cells ↑DA, DOPAC, and HVA | Zbarsky et al. 2005 [14] |
Naringenin (70 mg/kg; p.o.) 4-day pre-treatment | C57BL/6 mice (male) | 6-OHDA | ↑rotational behavior ↑TH-positive neurons ↑DA, DOPAC, and HVA ↑protein expression of NRf2, HO-1, GCLC, GCLM, HO-1 ↑GSH ↓ROS ↑protein expression of JNK and p38 | Lou et al. 2014 [15] |
Naringin (80 mg/kg; i.p.) 1-day pre-treatment and 6-day post-treatment | C57BL/6 mice (male) | 6-OHDA | ↑TH-positive neurons ↓Iba1-positive microglia ↓IL-1β positive microglia ↓6-OHDA—inducedp-4E-BP1 | Kim et al. 2016 [16] |
Hesperidin (50 mg/kg; p.o.) 28-day post-treatment | aged C57BL/6 mice (female) | 6-OHDA | ↓time spent immobile (antidepressant-like activity) ↑spatial learning and memory skill ↑DA, DOPAC and HVA ↑GSH level and TRAP ↑CAT and GPx activity ↓ROS levels ↓6-OHDA—induced GR activity | Antunes et al. 2014 [17] |
Hesperetin (50 mg/kg; p.o.) 1-week post-treatment | Wistar rats (male) | 6-OHDA | ↑rotational behavior ↑motor balance and coordination ↓MDA ↑GSH ↑CAT ↑Bcl-2 ↓GFAP expression and DNA fragmentation | Kiasalari et al. 2016 [18] |
Quercetin (50 mg/kg; i.g.) 4-day pre-treatment | Sprague-Dawley rats (male) | 6-OHDA | No effects | Zbarsky et al. 2005 [14] |
Quercetin (50–200 mg/kg; i.p.) once a day for one week before and for one week after the 6-OHDA-infusion (100 mg/kg; i.p.) twice a day for one week before and for one week after the 6-OHDA-infusion | Wistar rats (male) | 6-OHDA | No effects | Kääriäinen et al. 2008 [19] |
Quercetin (30 mg/kg; i.p.) 14-day post-treatment | Sprague-Dawley rats (male) | 6-OHDA | ↑DA ↑neuronal density of striatum ↑Nissl-stained neurons ↓PCs ↓LPO ↑GSH | Haleagrahara et al. 2011 [20] |
Rutin (25 mg/kg; p.o.) 3-week pre-treatment | Wistar rats (male) | 6-OHDA | ↑motor co-ordination ↑working performance ↑DA and DOPAC ↓TNF-α and IL-1β ↓NO ↓TBARS, H2O2, and PCs ↑GSH ↑GPx, GR, SOD and CAT activity | Khan et al. 2012 [21] |
Troxerutin (150 mg/kg; p.o.) 1-week pre-treatment | Wistar rats (male) | 6-OHDA | ↑motor function ↑numbers of TH-positive ↑Nissl positive neurons ↑DA and DOPAC ↓MDA, ROS, NO2 ↓GFAP ↓TBARS, H2O2, and PCs ↓DNA fragmentation ↑GPx, GR, SOD and CAT activity | Baluchnejadmojarad et al. 2017 [22] |
Myricitrin (60 mg/kg; i.p.) 1-day pre-treatment and 6-day post-treatment | C57BL/6 mice (male) | 6-OHDA | ↑motor function ↑TH-positive neurons ↓Iba1-positive microglia ↓TNF-α-positive microglia ↑TH activity ↑p-4E-BP1 and p-TH levels ↑mTORC1 | Kim et al. 2016 [16] |
Myricetin (5 µl of solution 0.5 mg/mL injected into lateral cerebral ventricle) 7-day post-treatment | Wistar rats (female) | 6-OHDA | ↑TH-positive neurons ↓iron-staining cells ↑DA, DOPAC and HVA ↑TH and GAPDH mRNA | Ma et al. 2007 [23] |
Genistein (10 mg/kg; i.p.) a single dose 1 h before surgery | Sprague–Dawley rats (male) | 6-OHDA | ↑rotational behavior ↑Nissl-stained neurons | Baluchnejadmojarad et al. 2009 [24] |
Puerarin (0.12 mg/kg; i.p.) 10-day co-treatment t | Sprague–Dawley rats (male) | 6-OHDA | ↑TH-positive cells ↑DA, DOPAC and HVA ↓degeneration of DAargic neurons ↓apoptosis (TUNEL assay) ↓Bax protein expression ↑GDNF protein expression | Zhu et al. 2010 [25] |
Baicalein (200 mg/kg; i.g.) 1-week pre-treatment | Sprague–Dawley rats (male) | 6-OHDA | ↓muscle tremor ↑GABA level ↓ GLu level ↓COI mRNA expression ↓GABA-T protein expression ↑GS protein expression | Yu et al. 2012 [26] |
Tangeretin (20 mg/kg; p.o.) 4-day pre-treatment | Sprague-Dawley rats (male) | 6-OHDA | ↑TH-positive cells ↑DA | Datla et al. 2001 [27] |
EGCG (1 and 2 mg/kg; p.o.) 14-day pre-treatment | Sprague–Dawley rats (female) | 6-OHDA | ↓postural abnormalities ↑ability to cross a narrow beam | Leaver et al. 2009 [28] |
Pelargonidin (10 and/or 20 mg/kg; p.o.) 1 day before and on the day of surgery. | Wistar rats (male) | 6-OHDA | ↑rotational behavior ↑SN neurons ↓TBARS | Roghani et al. 2010 [29] |
RES (10, 20 and 40 mg/kg; i.g.) 10-week post-treatment | Sprague–Dawley rats (male) | 6-OHDA | ↑rotational behavior ↓COX-2 and TNF-α mRNA ↓COX-2 protein | Jin et al. 2008 [30] |
RES (20 mg/kg; i.g.) 14-day post-treatment treatment | Wistar rats (male) | 6-OHDA | ↑rotational behavior ↑total nigral cells and DA neurons ↑total antioxidant capacity ↓number of apoptotic nigral cells ↓ROS | Wang et al. 2011 [31] |
RES (20 mg/kg; i.p.) 15-day pre-treatment | Wistar rats (male) | 6-OHDA | ↑rotational behaviour, motor coordination ↑TH-positive cells ↑DA, DOPAC ↓TBARS, PCs ↓PLA2, COX-2 protein ↑activity of GSH, SOD, CAT, GPx, GR ↑Na+/K+-ATPase activity | Khan et al. 2010 [32] |
Piceid (50 mg/kg; p.o.), 14-day co-treatment | Sprague-Dawley rats (male) | 6-OHDA | ↓motor defects ↓MDA ↑SOD activity | Chen et al. 2015 [33] |
CUR (200 mg/kg; i.g.) twice a day, 24-day pre-treatment | Wistar rats (female) | 6-OHDA | ↑TH-positive cells ↑DA, DOPAC and HVA ↓iron-stained cells | Du et al. 2012 [34] |
CUR (200 mg/kg; i.p.) 7-day post-treatment | ICR mice (male) | 6-OHDA | ↑TH-positive cells ↓GFAP, Iba1 protein ↑SOD1 level | Tripanichkul et al. 2013 [35] |
CA (20 mg/kg; p.o.) 3 times per week, 3-week pre-treatment | Wistar rats (male) | 6-OHDA | ↑locomotor time and distance traveled ↓TBARS ↑GSH ↑GCLC, GCLM, GR, and SOD protein ↓phosphorylated JNK and p38 protein (↓activation) ↑Bcl-2/Bax ↑cleaved caspase-3/caspase-3 and cleaved PARP/PARP | Wu et al. 2015 [36] |
SA (20 mg/kg; p.o.) 2-day pre-treatment | Wistar rats (male) | 6-OHDA | ↑rotational behavior ↑Nissl-stained, TH-positive and total SN neurons ↓iron-stained cells ↓MDA and nitrite level | Zare et al. 2015 [37] |
Ellagic acid (50 mg/kg; i.g) 10-day post-treatment | Wistar rats (male) | 6-OHDA | ↓motor deficiencies ↓IL-1β and TNF-α protein | Farbood et al. 2015 [38] |
Delta 9-tetrahydrocannabinol (3 mg/kg; i.p.) 2-week post-treatment | Sprague-Dawley rats (male) | 6-OHDA | ↑DA ↑TH mRNA and activity | Lastres-Becker et al. 2005 [39] |
EGCG (2 and 10 mg/kg; p.o.) 10-day pre-treatment | C57-BL mice (male) | MPTP | ↑DA ↑TH level and activity ↓MPTP-induced activity of SOD and CAT | Levites et al. 2001 [40] |
EGCG (2 mg/kg; p.o.) 10-day pre-treatment | C57-BL mice (male) | MPTP | ↑DA ↑TH level and activity ↓α-synuclein ↓Bcl-2, Bax protein ↑PKCα protein | Mandel et al. 2004 [41] |
EGCG (25 mg/kg; p.o.) 1-day pre-treatment and 5-day co-treatment | C57B6 mice (male) | MPTP | ↑TH-positive neurons ↑DA, DOPAC and HVA ↑TH activity ↓nNOS mRNAs, nNOS level and activity | Choi et al. 2002 [42] |
EGCG (25 mg/kg; p.o.) 7-day post-treatment | C57 black mice (male) | MPTP | ↑rotational behaviour ↑DA and DOPAC ↓PCs ↑ferroportin protein | Xu et al. 2017 [43] |
EGCG (10 mg/kg; i.g.) 14-day post-treatment | C57/BL6 mice (male) | MPTP | ↑DA, DOPAC and HVA ↑Ras expression | Reznichenko et al. 2010 [44] |
EGCG (10 , 50 mg/kg; i.p.) 10-day pre-treatment and 4-day co-treatment | C57Bl/6 mice (male) | MPTP | ↑TH-positive neurons ↓iNOS expression | Kim et al. 2010 [45] |
EGCG (25 , 50 mg/kg/day; i.g.) 1-day pre-treatment and 20-day post-treatment | C57BL/6 mice (male) | MPTP | ↓motor coordination ↑TH-positive neurons ↓CD3+CD4+/CD3+CD8+ T cells ↓TNF-α and IL-6 protein in plasma | Zhou et al. 2018 [46] |
Acacetin (10 mg/kg; p.o) 3-day co-treatment | C57BL/6 mice (male) | MPTP | ↓movement impairment ↑TH-positive neurons ↓damage in DAergic cells ↑DA ↓iNOS mRNA and COX-2 mRNA | Kim et al. 2012 [47] |
Baicalein (10 mg/kg; i.g.) 5-day co-treatment | C57BL/6 mice (male) | MPTP | ↓motor dysfunction ↓MPTP-induced glutamatergic transmission, presynaptic glutamate release and upregulation of synaptic GluR1 subunit | Xue et al. 2014 [48] |
Baicalein (1 and 10 mg/kg; i.g.) 7-day pre-treatment | C57BL/6 mice (male) | MPTP | ↓motor dysfunction ↑TH-positive neurons ↓GFAP, Iba1 protein ↓phosphorylated ERK and JNK- ↓activation | Lee et al. 2014 [49] |
7,8-dihydroxyflavone (5, 20, 40, and 100 mg/kg; i.p) 7-day pre-and 7-day co-treatment | B57/BL mice (male) | MPTP | ↑TH-positive neurons ↑TrkB activity ↓caspase-3 protein | Jang et al. 2010 [50] |
Tangeretin (10 mg/kg; i.p.) 4-day pre-treatment | C57BL/6 mice (male) | MPTP | ↑TH-positive neurons ↑DA ↑GRP-78 protein | Takano et al. 2007 [51] |
Nobiletin (10 mg/kg; i.p.) 1-day pre-treatment and 6-day-post-treatment | Sprague Dawley (SD) rats (female) | MPTP | ↑TH-positive neurons ↑DA ↓Iba1 protein and IL-1β ↑GDNF protein | Jeong et al. 2015 [52] |
Nobiletin (50 mg/kg; i.p.) 14-day post-treatment | C57BL/6 mice (male) | MPTP | ↓motor and cognitive impairment ↑Ca2+/calmodulin-dependent protein kinase II (CaMKII) ↑DARPP-32, dopamine- and cAMP-regulated phosphoprotein-32 | Yabuki et al. 2014 [53] |
Naringin (8 and 800 mg/kg; i.p) 1-day pre-and 6-day post-treatment | Sprague Dawley (SD) rats (female) | MPTP | ↑TH-positive neurons ↑GDNF protein ↑mTORC1 activity ↓TNF-α protein | Leem et al. 2014 [54] |
Kaempferol (25, 50 and 100 mg/kg; p.o) 14-day pre-treatment | C57BL/6 mice (male) | MPTP | ↑Motor behavioral ↑TH-positive neurons ↑DA and DOPAC ↓MDA ↑activity of SOD and GPx | Li and Pu 2011 [55] |
Quercetin (100, 200 mg/kg; p.o.) 10-day pre-treatment and 4-day co-treatment | C57BL/6 mice (male) | MPTP | ↑motor balance and coordination ↑DA ↑GPx, SOD activity ↑Na+, K+-ATPase ↓4-HNE ↑AChE activity | Lv et al. 2012 [56] |
Morin (5, 20, 40, and 100 mg/kg; i.p) 5-day pre-and 5-day co-treatment | B57/BL mice (male) | MPTP | ↓cataleptic time (bar test) and ↑ number of steps (drag test) ↑DA ↑TH-positive neurons | Zhang et al. 2010 [57] |
Silibinin (1 or 10 mg/kg; i.p.) 5-day co-treatment | C57BL/6 mice (male) | MPTP | ↓Motor dysfunction ↓DAergic neuron damage | Lee et al. 2015 [58] |
Silibinin (50, 100 mg/kg; i.p.) 5-day co-treatment | Sprague Dawley (female) | MPTP | ↑TH-positive neurons ↓IL-1β, TNF-α and iNOS protein | Jung et al. 2014 [59] |
Silibinin (100, 200 mg/kg; i.g.) 7-day co-treatment | albino rats of Charles– Foster strain (male) | MPTP | ↑spatial memory and locomotor activity ↓mitochondrial complex-I and IV activity ↑mitochondrial complex-II and V activity ↑mitochondrial membrane potential ↓NO, MDA ↑SOD activity ↓caspase-3 and NFκB protein | Geed et al. 2014 [60] |
Genistein (10 mg/kg; i.p.) 3-day pre-treatment and 5-day-co-treatment | C57BL/6 mice (male) | MPTP | ↑TH-positive neurons ↑DA, DOPAC and HVA ↑TH and DAT mRNA ↓Bcl-2 mRNA | Liu et al. 2008 [61] |
Gastrodin (10, 30 and 60 mg/kg; p.o.) 15-day co-treatment | C57BL/6 mice (male) | MPTP | ↓bradykinesia and motor impairment ↑TH protein ↓GFAP protein ↑Bcl-2 protein ↓Bax and caspase-3 protein ↓PARP cleavage | Kumar et al. 2013 [62] |
Tanshinone I (5 and 10 mg/kg; i.g.) 7-day pre- and co-treatment | C57BL/6 mice (male) | MPTP | ↓Motor dysfunction ↑DA, DOPAC and HVA ↑TH-positive neurons ↓Iba1-positive activated microglia cells ↓TNF-α and IL-10 protein | Wang et al. 2015 [63] |
Ferulic acid (40 mg/kg; i.g.) 3-day pre-treatment and 4-day co-treatment | C57BL/6 mice (male) | MPTP | ↑motor balance and coordination ↓anxiety ↓degeneration of DAargic neurons ↓Bax/Bcl-2 ratio | Nagarajan et al. 2015 [64] |
Madecassoside (15, 30 and 60 mg/kg; i.g) 7-day pre-treatment and 14-day co-treatment | Wistar rats m (male) | MPTP | ↑limb coordination and limb placing ↑DA, DOPAC and HVA ↓MDA ↑GSH ↑Bcl-2/Bax ratio ↑BDNF protein | Xu et al. 2013 [65] |
Curcumin (50 mg/kg; i.p.) 3-times at time points 1, 3, and 7 h post first MPTP injection | ICR mice (male) | MPTP | ↑DA ↑density of DA neurons | Vajragupta et al. 2003 [66] |
Curcumin (80 mg/kg; i.p.) 7-day co-treatment | Swiss albino mice (male) | MPTP | ↑DA and DOPAC ↓MAO B activity | Rajeswari et al. 2008 [67] |
Curcumin (150 mg/kg; p.o.) 1-week pre-treatment | C57BL/6 mice (male) | MPTP | ↑motor performance (rotarod test) ↓steep reduction intotal ambulation time (open field test) ↑DA, DOPAC and HVA ↓GFAP over-expression ↓IL-6 and TNF-α protein ↓iNOS protein and NO content | Ojha et al. 2012 [68] |
Ginsenoside Rg1 (5, 10 and 20 mg/kg; i.p.) 3-day pre-treatment and 5-day co-treatment | C57-BL mice (male) | MPTP | ↑numbers of TH-positive and Nissl positive neurons in SN ↓TUNEL positive neurons in SN ↑GSH ↓MPTP-induced SOD activity ↓MPTP-induced phosphor—JNK and phospho-c-Jun level | Chen et al. 2005 [69] |
Piceid (100 and 200 mg/kg; p.o.) 7-day co-treatment and 7-day post-treatment | C57BL/6 mice (male) | MPTP | ↑locomotor activity ↑TH-positive neurons ↑SOD activity ↓MDA level ↑p-Akt expression ↓activated caspase 3 expression | Chen et al. 2015 [33] |
TF (10 mg/kg; i.g.) 35-day co-treatment | C57BL/6 mice (male) | MPTP/p | ↑Locomotor activity ↑TH- and DAT-positive neurons ↓caspase-3, 8, and 9 protein | Anandhan et al. 2012 [70] |
TF (10 mg/kg; i.g.) 3-day pre-treatment and 4-day co-treatment | C57BL/6 mice (male) | MPTP | ↑Locomotor activity ↑DAT-positive neurons ↓TBARS, ↑ GSH ↑SOD, CAT and GPx activity ↓MAO-B activity | Anandhan et al. 2012b [71] |
TF (10 mg/kg; i.g.) 35-day co-treatment | C57BL/6 mice (male) | MPTP/p | ↓Akinesia, catalepsy ↓IL-1β, TNF-α, IL-6 as well as MPTP-induced IL-4 and IL-10 ↓GFAP and COX-2 protein ↑Bcl-2 protein ↓Bax protein | Anandhan et al. 2013 [72] |
Apigenin (5, 10 and 20 mg/kg; p.o.), luteolin (10 and 20 mg/kg; p.o.) 5-day pre-treatment and 21-day co-treatment | Swiss-albino mice (male) | MPTP/p | ↑Locomotor activity ↑TH-positive neurons ↓MDA, ↑GSH ↑SOD and CAT activity ↓GFAP and TNF-α protein ↑BDNF protein ↓MAO-B activity | Patil et al. 2014 [73] |
SA (20 mg/kg; i.g.) 35-day co-treatment | C57BL/6 mice (male) | MPTP/p | ↓impairment of motor coordination ↑TH- and DAT-positive neurons ↑TH, DAT and VMAT2 protein ↑DA, DOPAC and HVA ↓TBARS, ↑ GSH ↑SOD, CAT and GPx activity ↓IL-1β, TNF-α protein ↓COX-2 protein | Rekha et al. 2014 [74] |
Quercetin (25, 50, 75 mg/kg; i.p.) 4-day post-treatment | Sprague Dawley rats (male) | ROT | ↑rotational behavior ↑TH-positive neurons ↓TUNEL positive neurons ↑DA ↑complex-I activity ↑GSH, GSSG ↓ROT-induced activity of SOD and CAT | Karuppagounder et al. 2013 [75] |
CUR (20 mg/kg; i.g.) 50-day co-treatment | Lewis rats (male) | ROT | ↓postural impairment ↑motor coordination ↑TH activity ↓MDA, ↑ GSH ↑HO-1 and NQO1 protein ↑Akt/Nrf2 phosphorylation (activation) | Cui et al. 2016 [76] |
Piceid (80 mg/kg; i.g.) 5-week co-treatment | Sprague Dawley rats (male) | ROT | ↓postural impairment ↑motor coordination ↑TH-positive neurons ↑ATP ↑Trx, GSH ↑SOD activity | Chen et al. 2015 [33] |
FA (80 mg/kg; i.g.) 4-week co-treatment | Wistar rats (male) | ROT | ↑TH-positive neurons ↑SOD and CAT activity ↓MDA, ↑ GSH ↓GFAP and Iba1 protein ↓IL-1β, TNF-α, IL-6 protein ↓COX-2 and iNOS protein | Ojha et al. 2015 [77] |
Sesamol (15 mg/kg; p.o.) naringenin(10 mg/kg; p.o.) 10-day post-treatment | Wistar rats (male) | ROT | ↑motor coordination ↑body weight ↓degenerated neurons ↑TH and ubiquitin expression ↑parkin, CHIP, and DJ1 expression ↓caspase-9 and 3 protein ↑Hsp70 and Hsp90 protein | Sonia Angeline et al. 2013 [78] |
EGCG (10 mg/kg; i.p.) 24 h before LPS and continues for 7 days | Sprague Dawley rats (male) | LPS | ↑TH-positive neurons ↑DA ↓TNF-α and nitrite | AL-amri et al. 2013 [79] |
CUR (50 mg/kg; i.p.) 10-day treatment beginning 5 days prior to Hcy | Lewis rats (male) | Hcy | ↑Locomotor Activity ↑Nissl-stained neurons ↑Bcl-2 protein and ↓ Bax protein ↓Bax/Bcl-2 | Mansouri et al. 2012 [80] |
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kujawska, M.; Jodynis-Liebert, J. Polyphenols in Parkinson’s Disease: A Systematic Review of In Vivo Studies. Nutrients 2018, 10, 642. https://doi.org/10.3390/nu10050642
Kujawska M, Jodynis-Liebert J. Polyphenols in Parkinson’s Disease: A Systematic Review of In Vivo Studies. Nutrients. 2018; 10(5):642. https://doi.org/10.3390/nu10050642
Chicago/Turabian StyleKujawska, Małgorzata, and Jadwiga Jodynis-Liebert. 2018. "Polyphenols in Parkinson’s Disease: A Systematic Review of In Vivo Studies" Nutrients 10, no. 5: 642. https://doi.org/10.3390/nu10050642