Iron Dysregulation and Inflammagens Related to Oral and Gut Health Are Central to the Development of Parkinson’s Disease
Abstract
:1. Introduction
2. Parkinson’s Disease, Iron, Oxidative Stress, and Chronic Systemic Inflammation
3. An Integrated Physiological Systems Disease
A Continuum of Pathological Events or the Distinct Events Hypothesis?
4. Oral and Gut Dysbiosis and Parkinson’s Disease
4.1. Bacterial Inflammagens
4.2. Contribution of LPSs to Parkinson’s Disease
4.2.1. LPS as a Potent Inflammagen
4.2.2. Formation of Nucleated Molecular Intermediates as Induced by LPS
4.3. Oral Microbiota Translocation in Parkinson’s Disease
Gingipains as Potent Inflammagens from P. gingivalis
5. Therapeutic Possibilities and Pharmaceutical Interventions
5.1. Iron Chelation
5.2. Antibiotics and Probiotics
5.3. Faecal Microbiota Transplantation
5.4. Additional Therapeutic Options
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
α-Syn | Alpha-synuclein |
AD | Alzheimer’s disease |
Aβ | Beta-amyloid |
BBB | Blood–brain barrier |
CD | Cluster of differentiation 14 |
CNS | Central nervous system |
COX-2 | Cyclooxygenase-2 |
CRP | C-reactive protein |
CT51 | Coumarin-tris hybrid |
DHC12 | 7,8-dihydroxycoumarin-12 |
Hgb | Haemoglobin |
IFNγ | Interferon-gamma |
IL | Interleukin |
iNOS | Inducible nitric oxide synthase |
LB | Lewy body |
LPS | Lipopolysaccharide |
LTA | Lipoteichoic acid |
MRC | Medical Research Council of South Africa |
mRNA | Messenger ribonucleic acid |
MOA | Monoamine oxidase |
NADPH | Nicotinamide adenine dinucleotide phosphate reduced |
NF-кB | Nuclear factor kappa-light-chain-enhancer of activated B cells |
PARK | Parkinson’s disease genes |
PD | Parkinson’s disease |
RANTES | Regulated upon activation, normal T cell expressed and presumably secreted |
REM | Rapid eye movement |
Rgp | Gingipain |
ROS | Reactive oxygen species |
SAA | Serum amyloid A |
SNc | Substantia nigra pars compacta |
SUMO | Small ubiquitin-like modifier |
TEM | Transmission electron microscope |
TLR | Toll-like receptor |
TNF-α | Tumour necrosis factor-alpha |
TFN | Transferrin |
References
- Dorsey, E.R.; Elbaz, A.; Nichols, E.; Abd-Allah, F.; Abdelalim, A.; Adsuar, J.C.; Ansha, M.G.; Brayne, C.; Choi, J.-Y.J.; Collado-Mateo, D. Global, regional, and national burden of Parkinson’s disease, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2018, 17, 939–953. [Google Scholar] [CrossRef] [Green Version]
- Bach, J.P.; Ziegler, U.; Deuschl, G.; Dodel, R.; Doblhammer-Reiter, G. Projected numbers of people with movement disorders in the years 2030 and 2050. Mov. Disord. 2011, 26, 2286–2290. [Google Scholar] [CrossRef] [PubMed]
- Dorsey, E.R.; Bloem, B.R. The Parkinson pandemic—A call to action. JAMA Neurol. 2018, 75, 9–10. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharjee, P.; Öhrfelt, A.; Lashley, T.; Blennow, K.; Brinkmalm, A.; Zetterberg, H. Mass Spectrometric Analysis of Lewy Body-Enriched α-Synuclein in Parkinson’s Disease. J. Proteome Res. 2019, 18, 2109–2120. [Google Scholar] [CrossRef] [PubMed]
- Del Tredici, K.; Rüb, U.; De Vos, R.A.; Bohl, J.R.; Braak, H. Where does Parkinson disease pathology begin in the brain? J. Neuropathol. Exp. Neurol. 2002, 61, 413–426. [Google Scholar] [CrossRef]
- Braak, H.; Del Tredici, K.; Rub, U.; de Vos, R.A.; Jansen Steur, E.N.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
- Braak, H.; Rüb, U.; Gai, W.; Del Tredici, K. Idiopathic Parkinson’s disease: Possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm. 2003, 110, 517–536. [Google Scholar] [CrossRef]
- Grunewald, A.; Kumar, K.R.; Sue, C.M. New insights into the complex role of mitochondria in Parkinson’s disease. Prog. Neurobiol. 2019, 177, 73–93. [Google Scholar] [CrossRef]
- Farrand, A.Q.; Helke, K.L.; Gregory, R.A.; Gooz, M.; Hinson, V.K.; Boger, H.A. Vagus nerve stimulation improves locomotion and neuronal populations in a model of Parkinson’s disease. Brain Stimul. 2017, 10, 1045–1054. [Google Scholar] [CrossRef]
- Devos, D.; Lebouvier, T.; Lardeux, B.; Biraud, M.; Rouaud, T.; Pouclet, H.; Coron, E.; Bruley des Varannes, S.; Naveilhan, P.; Nguyen, J.M.; et al. Colonic inflammation in Parkinson’s disease. Neurobiol. Dis. 2013, 50, 42–48. [Google Scholar] [CrossRef]
- Jucker, M.; Walker, L.C. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013, 501, 45–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Volpicelli-Daley, L.A.; Luk, K.C.; Patel, T.P.; Tanik, S.A.; Riddle, D.M.; Stieber, A.; Meaney, D.F.; Trojanowski, J.Q.; Lee, V.M. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 2011, 72, 57–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hawkes, C.H.; Del Tredici, K.; Braak, H. Parkinson’s disease: A dual-hit hypothesis. Neuropathol. Appl. Neurobiol. 2007, 33, 599–614. [Google Scholar] [CrossRef] [PubMed]
- Reichmann, H. View point: Etiology in Parkinson’s disease. Dual hit or spreading intoxication. J. Neurol. Sci. 2011, 310, 9–11. [Google Scholar] [CrossRef] [PubMed]
- Shannon, K.M.; Keshavarzian, A.; Mutlu, E.; Dodiya, H.B.; Daian, D.; Jaglin, J.A.; Kordower, J.H. Alpha-synuclein in colonic submucosa in early untreated Parkinson’s disease. Mov. Disord. 2012, 27, 709–715. [Google Scholar] [CrossRef]
- Wynford-Thomas, R.; Robertson, N.P. The role of skin biopsy in differentiating idiopathic Parkinson’s disease from other types of parkinsonism. J. Neurol. 2015, 262, 2793–2795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Claassen, D.O.; Josephs, K.A.; Ahlskog, J.E.; Silber, M.H.; Tippmann-Peikert, M.; Boeve, B.F. REM sleep behavior disorder preceding other aspects of synucleinopathies by up to half a century. Neurology 2010, 75, 494–499. [Google Scholar] [CrossRef] [Green Version]
- Abbott, R.D.; Petrovitch, H.; White, L.R.; Masaki, K.H.; Tanner, C.M.; Curb, J.D.; Grandinetti, A.; Blanchette, P.L.; Popper, J.S.; Ross, G.W. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology 2001, 57, 456–462. [Google Scholar] [CrossRef]
- Nuzum, N.D.; Loughman, A.; Szymlek-Gay, E.A.; Hendy, A.; Teo, W.P.; Macpherson, H. Gut microbiota differences between healthy older adults and individuals with Parkinson’s disease: A systematic review. Neurosci. Biobehav. Rev. 2020, 112, 227–241. [Google Scholar] [CrossRef]
- Funke, C.; Schneider, S.A.; Berg, D.; Kell, D.B. Genetics and iron in the systems biology of Parkinson’s disease and some related disorders. Neurochem. Int. 2013, 62, 637–652. [Google Scholar] [CrossRef]
- Klein, C.; Westenberger, A. Genetics of Parkinson’s disease. Cold Spring Harbor Perspect. Med. 2012, 2, a008888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Genoud, S.; Senior, A.M.; Hare, D.J.; Double, K.L. Meta-Analysis of Copper and Iron in Parkinson’s Disease Brain and Biofluids. Mov. Disord. 2020, 35, 662–671. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharyya, D.; Mohite, G.M.; Krishnamoorthy, J.; Gayen, N.; Mehra, S.; Navalkar, A.; Kotler, S.A.; Ratha, B.N.; Ghosh, A.; Kumar, R. Lipopolysaccharide from Gut Microbiota Modulates α-Synuclein Aggregation and Alters Its Biological Function. ACS Chem. Neurosci. 2019, 10, 2229–2236. [Google Scholar] [CrossRef] [PubMed]
- Calabrese, V.; Santoro, A.; Monti, D.; Crupi, R.; Di Paola, R.; Latteri, S.; Cuzzocrea, S.; Zappia, M.; Giordano, J.; Calabrese, E.J. Aging and Parkinson’s Disease: Inflammaging, neuroinflammation and biological remodeling as key factors in pathogenesis. Free Radic. Biol. Med. 2018, 115, 80–91. [Google Scholar] [CrossRef]
- Caputi, V.; Giron, M.C. Microbiome-Gut-Brain Axis and Toll-Like Receptors in Parkinson’s Disease. Int. J. Mol. Sci 2018, 19, 1689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mukherjee, A.; Biswas, A.; Das, S.K. Gut dysfunction in Parkinson’s disease. World J. Gastroenterol. 2016, 22, 5742–5752. [Google Scholar] [CrossRef]
- Müller, T.; Palluch, R.; Jackowski, J. Caries and periodontal disease in patients with Parkinson’s disease. Spec. Care Dent. 2011, 31, 178–181. [Google Scholar] [CrossRef]
- Parashar, A.; Udayabanu, M. Gut microbiota: Implications in Parkinson’s disease. Parkinsonism Relat. Disord. 2017, 38, 1–7. [Google Scholar] [CrossRef] [Green Version]
- De Virgilio, A.; Greco, A.; Fabbrini, G.; Inghilleri, M.; Rizzo, M.I.; Gallo, A.; Conte, M.; Rosato, C.; Ciniglio Appiani, M.; de Vincentiis, M. Parkinson’s disease: Autoimmunity and neuroinflammation. Autoimmun. Rev. 2016, 15, 1005–1011. [Google Scholar] [CrossRef] [Green Version]
- Grozdanov, V.; Bousset, L.; Hoffmeister, M.; Bliederhaeuser, C.; Meier, C.; Madiona, K.; Pieri, L.; Kiechle, M.; McLean, P.J.; Kassubek, J.; et al. Increased Immune Activation by Pathologic α-Synuclein in Parkinson’s Disease. Ann. Neurol. 2019, 86, 593–606. [Google Scholar] [CrossRef] [Green Version]
- Butkovich, L.M.; Houser, M.C.; Tansey, M.G. α-Synuclein and Noradrenergic Modulation of Immune Cells in Parkinson’s Disease Pathogenesis. Front. Neurosci. 2018, 12, 626. [Google Scholar] [CrossRef]
- Kannarkat, G.T.; Boss, J.M.; Tansey, M.G. The role of innate and adaptive immunity in Parkinson’s disease. J. Parkinson Dis. 2013, 3, 493–514. [Google Scholar] [CrossRef] [Green Version]
- Kim, R.; Kim, H.J.; Kim, A.; Jang, M.; Kim, A.; Kim, Y.; Yoo, D.; Im, J.H.; Choi, J.H.; Jeon, B. Peripheral blood inflammatory markers in early Parkinson’s disease. J. Clin. Neurosci. 2018, 58, 30–33. [Google Scholar] [CrossRef]
- Williams-Gray, C.H.; Wijeyekoon, R.; Yarnall, A.J.; Lawson, R.A.; Breen, D.P.; Evans, J.R.; Cummins, G.A.; Duncan, G.W.; Khoo, T.K.; Burn, D.J.; et al. Serum immune markers and disease progression in an incident Parkinson’s disease cohort (ICICLE-PD). Mov. Disord. 2016, 31, 995–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eidson, L.N.; Kannarkat, G.T.; Barnum, C.J.; Chang, J.; Chung, J.; Caspell-Garcia, C.; Taylor, P.; Mollenhauer, B.; Schlossmacher, M.G.; Ereshefsky, L.; et al. Candidate inflammatory biomarkers display unique relationships with alpha-synuclein and correlate with measures of disease severity in subjects with Parkinson’s disease. J. Neuroinflamm. 2017, 14, 164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGeer, P.L.; McGeer, E.G. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat. Disord. 2004, 10 (Suppl. 1), S3–S7. [Google Scholar] [CrossRef] [PubMed]
- Perry, V.H. Innate inflammation in Parkinson’s disease. Cold Spring Harbor Perspect. Med. 2012, 2, a009373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stojkovska, I.; Wagner, B.M.; Morrison, B.E. Parkinson’s disease and enhanced inflammatory response. Exp. Biol Med. 2015, 240, 1387–1395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caggiu, E.; Arru, G.; Hosseini, S.; Niegowska, M.; Sechi, G.; Zarbo, I.R.; Sechi, L.A. Inflammation, Infectious Triggers, and Parkinson’s Disease. Front. Neurol. 2019, 10, 122. [Google Scholar] [CrossRef] [Green Version]
- Tansey, M.G.; Romero-Ramos, M. Immune system responses in Parkinson’s disease: Early and dynamic. Eur. J. Neurosci. 2019, 49, 364–383. [Google Scholar] [CrossRef]
- Benakis, C.; Martin-Gallausiaux, C.; Trezzi, J.P.; Melton, P.; Liesz, A.; Wilmes, P. The microbiome-gut-brain axis in acute and chronic brain diseases. Curr. Opin. Neurobiol. 2020, 61, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Mulak, A.; Bonaz, B. Brain-gut-microbiota axis in Parkinson’s disease. World J. Gastroenterol. 2015, 21, 10609–10620. [Google Scholar] [CrossRef] [PubMed]
- Nair, A.T.; Ramachandran, V.; Joghee, N.M.; Antony, S.; Ramalingam, G. Gut Microbiota Dysfunction as Reliable Non-invasive Early Diagnostic Biomarkers in the Pathophysiology of Parkinson’s Disease: A Critical Review. J. Neurogastroenterol. Motil. 2018, 24, 30–42. [Google Scholar] [CrossRef]
- Kell, D.B.; Pretorius, E. No effects without causes: The Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol. Rev. 2018, 93, 1518–1557. [Google Scholar] [CrossRef] [PubMed]
- Adams, B.; Nunes, J.M.; Page, M.J.; Roberts, T.; Carr, J.; Nell, T.A.; Kell, D.B.; Pretorius, E. Parkinson’s Disease: A Systemic Inflammatory Disease Accompanied by Bacterial Inflammagens. Front. Aging Neurosci. 2019, 11, 210. [Google Scholar] [CrossRef] [Green Version]
- Pessoa Rocha, N.; Reis, H.J.; Vanden Berghe, P.; Cirillo, C. Depression and cognitive impairment in Parkinson’s disease: A role for inflammation and immunomodulation? Neuroimmunomodulation 2014, 21, 88–94. [Google Scholar] [CrossRef] [PubMed]
- Deleidi, M.; Gasser, T. The role of inflammation in sporadic and familial Parkinson’s disease. Cell. Mol. Life Sci. CMLS 2013, 70, 4259–4273. [Google Scholar] [CrossRef]
- More, S.V.; Kumar, H.; Kim, I.S.; Song, S.Y.; Choi, D.K. Cellular and molecular mediators of neuroinflammation in the pathogenesis of Parkinson’s disease. Mediat. Inflamm. 2013, 2013, 952375. [Google Scholar] [CrossRef]
- Nolan, Y.M.; Sullivan, A.M.; Toulouse, A. Parkinson’s disease in the nuclear age of neuroinflammation. Trends Mol. Med. 2013, 19, 187–196. [Google Scholar] [CrossRef]
- Taylor, J.M.; Main, B.S.; Crack, P.J. Neuroinflammation and oxidative stress: Co-conspirators in the pathology of Parkinson’s disease. Neurochem. Int. 2013, 62, 803–819. [Google Scholar] [CrossRef]
- Filiou, M.D.; Arefin, A.S.; Moscato, P.; Graeber, M.B. ‘Neuroinflammation’ differs categorically from inflammation: Transcriptomes of Alzheimer’s disease, Parkinson’s disease, schizophrenia and inflammatory diseases compared. Neurogenetics 2014, 15, 201–212. [Google Scholar] [CrossRef] [PubMed]
- Perez, A.; Guan, L.; Sutherland, K.; Cao, C. Immune system and Parkinson’s disease. Arch. Med. 2016, 8, 1–6. [Google Scholar]
- Jiang, H.; Wang, J.; Rogers, J.; Xie, J. Brain Iron Metabolism Dysfunction in Parkinson’s Disease. Mol. Neurobiol. 2017, 54, 3078–3101. [Google Scholar] [CrossRef]
- Pretorius, E.; Swanepoel, A.C.; Buys, A.V.; Vermeulen, N.; Duim, W.; Kell, D.B. Eryptosis as a marker of Parkinson’s disease. Aging 2014, 6, 788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galaris, D.; Barbouti, A.; Pantopoulos, K. Iron homeostasis and oxidative stress: An intimate relationship. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 118535. [Google Scholar] [CrossRef]
- Pretorius, E.; Bester, J.; Vermeulen, N.; Lipinski, B. Oxidation inhibits iron-induced blood coagulation. Curr. Drug Targets 2013, 14, 13–19. [Google Scholar] [CrossRef] [Green Version]
- Pretorius, E.; Vermeulen, N.; Bester, J.; Lipinski, B.; Kell, D.B. A novel method for assessing the role of iron and its functional chelation in fibrin fibril formation: The use of scanning electron microscopy. Toxicol. Mech. Methods 2013, 23, 352–359. [Google Scholar] [CrossRef]
- Kell, D.B.; Pretorius, E. Serum ferritin is an important inflammatory disease marker, as it is mainly a leakage product from damaged cells. Metallomics 2014, 6, 748–773. [Google Scholar] [CrossRef] [Green Version]
- Pretorius, E.; Kell, D.B. Diagnostic morphology: Biophysical indicators for iron-driven inflammatory diseases. Integr. Biol. 2014, 6, 486–510. [Google Scholar] [CrossRef] [Green Version]
- Madenci, G.; Bilen, S.; Arli, B.; Saka, M.; Ak, F. Serum iron, vitamin B12 and folic acid levels in Parkinson’s disease. Neurochem. Res. 2012, 37, 1436–1441. [Google Scholar] [CrossRef]
- Berg, D.; Youdim, M.B. Role of iron in neurodegenerative disorders. Top. Magn. Reason Imaging 2006, 17, 5–17. [Google Scholar] [CrossRef] [PubMed]
- Altamura, S.; Muckenthaler, M.U. Iron toxicity in diseases of aging: Alzheimer’s disease, Parkinson’s disease and atherosclerosis. J. Alzheimer Dis. JAD 2009, 16, 879–895. [Google Scholar] [CrossRef] [PubMed]
- Kell, D.B. Iron behaving badly: Inappropriate iron chelation as a major contributor to the aetiology of vascular and other progressive inflammatory and degenerative diseases. BMC Med. Genom. 2009, 2, 2. [Google Scholar]
- Schneider, S.A.; Hardy, J.; Bhatia, K.P. Syndromes of neurodegeneration with brain iron accumulation (NBIA): An update on clinical presentations, histological and genetic underpinnings, and treatment considerations. Mov. Disord. 2012, 27, 42–53. [Google Scholar] [CrossRef] [PubMed]
- Qiu, Y.; Cao, Y.; Cao, W.; Jia, Y.; Lu, N. The Application of Ferroptosis in Diseases. Pharmacol. Res. 2020, 159, 104919. [Google Scholar] [CrossRef] [PubMed]
- Weiland, A.; Wang, Y.; Wu, W.; Lan, X.; Han, X.; Li, Q.; Wang, J. Ferroptosis and Its Role in Diverse Brain Diseases. Mol. Neurobiol. 2019, 56, 4880–4893. [Google Scholar] [CrossRef]
- Do Van, B.; Gouel, F.; Jonneaux, A.; Timmerman, K.; Gelé, P.; Pétrault, M.; Bastide, M.; Laloux, C.; Moreau, C.; Bordet, R.; et al. Ferroptosis, a newly characterized form of cell death in Parkinson’s disease that is regulated by PKC. Neurobiol. Dis. 2016, 94, 169–178. [Google Scholar] [CrossRef]
- Cao, J.Y.; Dixon, S.J. Mechanisms of ferroptosis. Cell. Mol. Life Sci. CMLS 2016, 73, 2195–2209. [Google Scholar] [CrossRef] [Green Version]
- Yu, H.; Guo, P.; Xie, X.; Wang, Y.; Chen, G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J. Cell Mol. Med. 2017, 21, 648–657. [Google Scholar] [CrossRef]
- Guiney, S.J.; Adlard, P.A.; Bush, A.I.; Finkelstein, D.I.; Ayton, S. Ferroptosis and cell death mechanisms in Parkinson’s disease. Neurochem. Int. 2017, 104, 34–48. [Google Scholar] [CrossRef]
- Hare, D.J.; Double, K.L. Iron and dopamine: A toxic couple. Brain 2016, 139 Pt 4, 1026–1035. [Google Scholar] [CrossRef]
- Plum, S.; Steinbach, S.; Attems, J.; Keers, S.; Riederer, P.; Gerlach, M.; May, C.; Marcus, K. Proteomic characterization of neuromelanin granules isolated from human substantia nigra by laser-microdissection. Sci. Rep. 2016, 6, 37139. [Google Scholar] [CrossRef] [Green Version]
- Knorle, R. Neuromelanin in Parkinson’s Disease: From Fenton Reaction to Calcium Signaling. Neurotox Res. 2018, 33, 515–522. [Google Scholar] [CrossRef]
- Stojakovic, A.; Paz-Filho, G.; Arcos-Burgos, M.; Licinio, J.; Wong, M.L.; Mastronardi, C.A. Role of the IL-1 Pathway in Dopaminergic Neurodegeneration and Decreased Voluntary Movement. Mol. Neurobiol 2017, 54, 4486–4495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alam, Q.; Alam, M.Z.; Mushtaq, G.; Damanhouri, G.A.; Rasool, M.; Kamal, M.A.; Haque, A. Inflammatory Process in Alzheimer’s and Parkinson’s Diseases: Central Role of Cytokines. Curr. Pharm. Des. 2016, 22, 541–548. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Guajardo, V.; Tentillier, N.; Romero-Ramos, M. The relation between α-synuclein and microglia in Parkinson’s disease: Recent developments. Neuroscience 2015, 302, 47–58. [Google Scholar] [CrossRef] [PubMed]
- Sutachan, J.J.; Casas, Z.; Albarracin, S.L.; Stab, B.R., 2nd; Samudio, I.; Gonzalez, J.; Morales, L.; Barreto, G.E. Cellular and molecular mechanisms of antioxidants in Parkinson’s disease. Nutr. Neurosci. 2012, 15, 120–126. [Google Scholar] [CrossRef]
- Chang, K.H.; Chen, C.M. The Role of Oxidative Stress in Parkinson’s Disease. Antioxidants (Basel) 2020, 9. [Google Scholar]
- Sohmiya, M.; Tanaka, M.; Tak, N.W.; Yanagisawa, M.; Tanino, Y.; Suzuki, Y.; Okamoto, K.; Yamamoto, Y. Redox status of plasma coenzyme Q10 indicates elevated systemic oxidative stress in Parkinson’s disease. J. Neurol. Sci. 2004, 223, 161–166. [Google Scholar] [CrossRef]
- Nicoletti, G.; Crescibene, L.; Scornaienchi, M.; Bastone, L.; Bagalà, A.; Napoli, I.D.; Caracciolo, M.; Quattrone, A. Plasma levels of vitamin E in Parkinson’s disease. Arch. Gerontol. Geriatr. 2001, 33, 7–12. [Google Scholar] [CrossRef]
- Mochizuki, H.; Choong, C.J.; Baba, K. Parkinson’s disease and iron. J. Neural. Transm. 2020, 127, 181–187. [Google Scholar] [CrossRef] [PubMed]
- Medeiros, M.S.; Schumacher-Schuh, A.; Cardoso, A.M.; Bochi, G.V.; Baldissarelli, J.; Kegler, A.; Santana, D.; Chaves, C.M.; Schetinger, M.R.; Moresco, R.N.; et al. Iron and Oxidative Stress in Parkinson’s Disease: An Observational Study of Injury Biomarkers. PLoS ONE 2016, 11, e0146129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Joers, V.; Tansey, M.G.; Mulas, G.; Carta, A.R. Microglial phenotypes in Parkinson’s disease and animal models of the disease. Prog. Neurobiol. 2017, 155, 57–75. [Google Scholar] [CrossRef] [PubMed]
- Olanow, C.W. The pathogenesis of cell death in Parkinson’s disease—2007. Mov. Disord. 2007, 22 (Suppl. 17), S335–S342. [Google Scholar] [CrossRef] [PubMed]
- Andersen, J.K. Oxidative stress in neurodegeneration: Cause or consequence? Nat. Med. 2004, 10, S18–S25. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Guajardo, V.; Barnum, C.J.; Tansey, M.G.; Romero-Ramos, M. Neuroimmunological processes in Parkinson’s disease and their relation to α-synuclein: Microglia as the referee between neuronal processes and peripheral immunity. ASN Neurol. 2013, 5, AN20120066. [Google Scholar] [CrossRef] [Green Version]
- Castro, A.; Macedo-de la Concha, L.; Pantoja-Meléndez, C. Low-grade inflammation and its relation to obesity and chronic degenerative diseases. Rev. Médica Hosp. Gen. México 2017, 80, 101–105. [Google Scholar] [CrossRef]
- Barcia, C.; Ros, C.M.; Annese, V.; Gomez, A.; Ros-Bernal, F.; Aguado-Yera, D.; Martinez-Pagan, M.E.; de Pablos, V.; Fernandez-Villalba, E.; Herrero, M.T. IFN-gamma signaling, with the synergistic contribution of TNF-alpha, mediates cell specific microglial and astroglial activation in experimental models of Parkinson’s disease. Cell Death Dis. 2011, 2, e142. [Google Scholar] [CrossRef] [Green Version]
- Boka, G.; Anglade, P.; Wallach, D.; Javoy-Agid, F.; Agid, Y.; Hirsch, E.C. Immunocytochemical analysis of tumor necrosis factor and its receptors in Parkinson’s disease. Neurosci. Lett. 1994, 172, 151–154. [Google Scholar] [CrossRef]
- Depino, A.M.; Earl, C.; Kaczmarczyk, E.; Ferrari, C.; Besedovsky, H.; del Rey, A.; Pitossi, F.J.; Oertel, W.H. Microglial activation with atypical proinflammatory cytokine expression in a rat model of Parkinson’s disease. Eur. J. Neurosci. 2003, 18, 2731–2742. [Google Scholar] [CrossRef]
- Song, I.-U.; Chung, S.-W.; Kim, J.-S.; Lee, K.-S. Association between high-sensitivity C-reactive protein and risk of early idiopathic Parkinson’s disease. Neurol. Sci. 2011, 32, 31–34. [Google Scholar] [CrossRef] [PubMed]
- Rathnayake, D.; Chang, T.; Udagama, P. Selected serum cytokines and nitric oxide as potential multi-marker biosignature panels for Parkinson disease of varying durations: A case-control study. BMC Neurol. 2019, 19, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qin, X.Y.; Zhang, S.P.; Cao, C.; Loh, Y.P.; Cheng, Y. Aberrations in Peripheral Inflammatory Cytokine Levels in Parkinson Disease: A Systematic Review and Meta-analysis. JAMA Neurol. 2016, 73, 1316–1324. [Google Scholar] [CrossRef] [PubMed]
- Fiszer, U.; Mix, E.; Fredrikson, S.; Kostulas, V.; Olsson, T.; Link, H. γδ+ T cells are increased in patients with Parkinson’s disease. J. Neurol. Sci. 1994, 121, 39–45. [Google Scholar] [CrossRef]
- Bas, J.; Calopa, M.; Mestre, M.; Mollevi, D.G.; Cutillas, B.; Ambrosio, S.; Buendia, E. Lymphocyte populations in Parkinson’s disease and in rat models of parkinsonism. J. Neuroimmunol. 2001, 113, 146–152. [Google Scholar] [CrossRef]
- Hunot, S.; Boissiere, F.; Faucheux, B.; Brugg, B.; Mouatt-Prigent, A.; Agid, Y.; Hirsch, E. Nitric oxide synthase and neuronal vulnerability in Parkinson’s disease. Neuroscience 1996, 72, 355–363. [Google Scholar] [CrossRef]
- Knott, C.; Stern, G.; Wilkin, G. Inflammatory regulators in Parkinson’s disease: iNOS, lipocortin-1, and cyclooxygenases-1 and-2. Mol. Cell. Neurosci. 2000, 16, 724–739. [Google Scholar] [CrossRef]
- Houser, M.C.; Tansey, M.G. The gut-brain axis: Is intestinal inflammation a silent driver of Parkinson’s disease pathogenesis? NPJ Parkinsons Dis. 2017, 3, 3. [Google Scholar] [CrossRef]
- Perez-Pardo, P.; Hartog, M.; Garssen, J.; Kraneveld, A.D. Microbes Tickling Your Tummy: The Importance of the Gut-Brain Axis in Parkinson’s Disease. Curr. Behav. Neurosci. Rep. 2017, 4, 361–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Houser, M.C.; Chang, J.; Factor, S.A.; Molho, E.S.; Zabetian, C.P.; Hill-Burns, E.M.; Payami, H.; Hertzberg, V.S.; Tansey, M.G. Stool Immune Profiles Evince Gastrointestinal Inflammation in Parkinson’s Disease. Mov. Disord. 2018, 33, 793–804. [Google Scholar] [CrossRef] [PubMed]
- Schwiertz, A.; Spiegel, J.; Dillmann, U.; Grundmann, D.; Bürmann, J.; Faßbender, K.; Schäfer, K.H.; Unger, M.M. Fecal markers of intestinal inflammation and intestinal permeability are elevated in Parkinson’s disease. Parkinsonism Relat. Disord. 2018, 50, 104–107. [Google Scholar] [CrossRef] [PubMed]
- de Waal, G.M.; Engelbrecht, L.; Davis, T.; de Villiers, W.J.; Kell, D.B.; Pretorius, E. Correlative Light-Electron Microscopy detects lipopolysaccharide and its association with fibrin fibres in Parkinson’s Disease, Alzheimer’s Disease and Type 2 Diabetes Mellitus. Sci. Rep. 2018, 8, 16798. [Google Scholar] [CrossRef] [PubMed]
- Hasegawa, S.; Goto, S.; Tsuji, H.; Okuno, T.; Asahara, T.; Nomoto, K.; Shibata, A.; Fujisawa, Y.; Minato, T.; Okamoto, A.; et al. Intestinal Dysbiosis and Lowered Serum Lipopolysaccharide-Binding Protein in Parkinson’s Disease. PLoS ONE 2015, 10, e0142164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wijeyekoon, R.S.; Kronenberg-Versteeg, D.; Scott, K.M.; Hayat, S.; Kuan, W.L.; Evans, J.R.; Breen, D.P.; Cummins, G.; Jones, J.L.; Clatworthy, M.R.; et al. Peripheral innate immune and bacterial signals relate to clinical heterogeneity in Parkinson’s disease. Brain Behav. Immun. 2020, 87, 473–488. [Google Scholar] [CrossRef] [PubMed]
- Barnham, K.J.; Bush, A.I. Metals in Alzheimer’s and Parkinson’s diseases. Curr. Opin. Chem. Biol. 2008, 12, 222–228. [Google Scholar] [CrossRef] [PubMed]
- Bartzokis, G.; Tishler, T.A.; Lu, P.H.; Villablanca, P.; Altshuler, L.L.; Carter, M.; Huang, D.; Edwards, N.; Mintz, J. Brain ferritin iron may influence age- and gender-related risks of neurodegeneration. Neurobiol. Aging 2007, 28, 414–423. [Google Scholar] [CrossRef]
- Berg, D.; Hochstrasser, H. Iron metabolism in Parkinsonian syndromes. Mov. Disord. 2006, 21, 1299–1310. [Google Scholar] [CrossRef]
- Boelmans, K.; Holst, B.; Hackius, M.; Finsterbusch, J.; Gerloff, C.; Fiehler, J.; Munchau, A. Brain iron deposition fingerprints in Parkinson’s disease and progressive supranuclear palsy. Mov. Disord. 2012, 27, 421–427. [Google Scholar] [CrossRef]
- Gotz, M.E.; Double, K.; Gerlach, M.; Youdim, M.B.; Riederer, P. The relevance of iron in the pathogenesis of Parkinson’s disease. Ann. N. Y. Acad. Sci. 2004, 1012, 193–208. [Google Scholar] [CrossRef]
- Jomova, K.; Vondrakova, D.; Lawson, M.; Valko, M. Metals, oxidative stress and neurodegenerative disorders. Mol. Cell. Biochem. 2010, 345, 91–104. [Google Scholar] [CrossRef]
- Lewis, M.M.; Du, G.; Kidacki, M.; Patel, N.; Shaffer, M.L.; Mailman, R.B.; Huang, X. Higher iron in the red nucleus marks Parkinson’s dyskinesia. Neurobiol. Aging 2013, 34, 1497–1503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lingor, P.; Carboni, E.; Koch, J.C. Alpha-synuclein and iron: Two keys unlocking Parkinson’s disease. J. Neural. Transm. 2017, 124, 973–981. [Google Scholar] [CrossRef] [PubMed]
- Logroscino, G.; Marder, K.; Graziano, J.; Freyer, G.; Slavkovich, V.; LoIacono, N.; Cote, L.; Mayeux, R. Altered systemic iron metabolism in Parkinson’s disease. Neurology 1997, 49, 714–717. [Google Scholar] [CrossRef] [PubMed]
- Thomas, M.; Jankovic, J. Neurodegenerative disease and iron storage in the brain. Curr. Opin. Neurol. 2004, 17, 437–442. [Google Scholar] [CrossRef] [PubMed]
- Genoud, S.; Roberts, B.R.; Gunn, A.P.; Halliday, G.M.; Lewis, S.J.G.; Ball, H.J.; Hare, D.J.; Double, K.L. Subcellular compartmentalisation of copper, iron, manganese, and zinc in the Parkinson’s disease brain. Metallomics 2017, 9, 1447–1455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sampson, T.R.; Challis, C.; Jain, N.; Moiseyenko, A.; Ladinsky, M.S.; Shastri, G.G.; Thron, T.; Needham, B.D.; Horvath, I.; Debelius, J.W.; et al. A gut bacterial amyloid promotes α-synuclein aggregation and motor impairment in mice. Elife 2020, 9, e53111. [Google Scholar] [CrossRef]
- Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
- Borghammer, P.; Van Den Berge, N. Brain-First versus Gut-First Parkinson’s Disease: A Hypothesis. J. Parkinson Dis. 2019, 9, S281–S295. [Google Scholar] [CrossRef] [Green Version]
- Asahina, M.; Vichayanrat, E.; Low, D.A.; Iodice, V.; Mathias, C.J. Autonomic dysfunction in parkinsonian disorders: Assessment and pathophysiology. J. Neurol. Neurosurg. Psychiatry 2013, 84, 674–680. [Google Scholar] [CrossRef]
- Merola, A.; Coon, E.A. Dysautonomia in early Parkinson disease: A window into the determinants of functional disability and an opportunity for early intervention. Clin. Auton. Res. Off. J. Clin. Auton. Res. Soc. 2020. [Google Scholar] [CrossRef]
- Anang, J.B.; Gagnon, J.F.; Bertrand, J.A.; Romenets, S.R.; Latreille, V.; Panisset, M.; Montplaisir, J.; Postuma, R.B. Predictors of dementia in Parkinson disease: A prospective cohort study. Neurology 2014, 83, 1253–1260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mendoza-Velasquez, J.J.; Flores-Vazquez, J.F.; Barron-Velazquez, E.; Sosa-Ortiz, A.L.; Illigens, B.W.; Siepmann, T. Autonomic Dysfunction in alpha-Synucleinopathies. Front. Neurol. 2019, 10, 363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, A.; Kurek, J.; Morgan, J.C.; Wakade, C.; Rao, S.S.C. Constipation in Parkinson’s Disease: A Nuisance or Nuanced Answer to the Pathophysiological Puzzle? Curr. Gastroenterol. Rep. 2018, 20, 1. [Google Scholar] [CrossRef] [PubMed]
- Hilton, D.; Stephens, M.; Kirk, L.; Edwards, P.; Potter, R.; Zajicek, J.; Broughton, E.; Hagan, H.; Carroll, C. Accumulation of α-synuclein in the bowel of patients in the pre-clinical phase of Parkinson’s disease. Acta Neuropathol. 2014, 127, 235–241. [Google Scholar] [CrossRef] [PubMed]
- Corbillé, A.G.; Preterre, C.; Rolli-Derkinderen, M.; Coron, E.; Neunlist, M.; Lebouvier, T.; Derkinderen, P. Biochemical analysis of α-synuclein extracted from control and Parkinson’s disease colonic biopsies. Neurosci. Lett. 2017, 641, 81–86. [Google Scholar] [CrossRef]
- Kaufmann, H.; Norcliffe-Kaufmann, L.; Palma, J.A.; Biaggioni, I.; Low, P.A.; Singer, W.; Goldstein, D.S.; Peltier, A.C.; Shibao, C.A.; Gibbons, C.H.; et al. Natural history of pure autonomic failure: A United States prospective cohort. Ann. Neurol. 2017, 81, 287–297. [Google Scholar] [CrossRef] [Green Version]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480. [Google Scholar] [CrossRef] [Green Version]
- Bonaz, B.; Bazin, T.; Pellissier, S. The vagus nerve at the interface of the microbiota-gut-brain axis. Front. Neurosci. 2018, 12, 49. [Google Scholar] [CrossRef] [Green Version]
- Ueki, A.; Otsuka, M. Life style risks of Parkinson’s disease: Association between decreased water intake and constipation. J. Neurol. 2004, 251, vii18–vii23. [Google Scholar] [CrossRef]
- Braak, H.; de Vos, R.A.; Bohl, J.; Del Tredici, K. Gastric alpha-synuclein immunoreactive inclusions in Meissner’s and Auerbach’s plexuses in cases staged for Parkinson’s disease-related brain pathology. Neurosci. Lett. 2006, 396, 67–72. [Google Scholar] [CrossRef]
- Verbaan, D.; Marinus, J.; Visser, M.; van Rooden, S.M.; Stiggelbout, A.M.; van Hilten, J.J. Patient-reported autonomic symptoms in Parkinson disease. Neurology 2007, 69, 333–341. [Google Scholar] [CrossRef]
- Scheperjans, F.; Aho, V.; Pereira, P.A.; Koskinen, K.; Paulin, L.; Pekkonen, E.; Haapaniemi, E.; Kaakkola, S.; Eerola-Rautio, J.; Pohja, M.; et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov. Disord. 2015, 30, 350–358. [Google Scholar] [CrossRef] [PubMed]
- Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. 2015, 30, 1351–1360. [Google Scholar] [CrossRef] [PubMed]
- Cersosimo, M.G.; Benarroch, E.E. Pathological correlates of gastrointestinal dysfunction in Parkinson’s disease. Neurobiol. Dis. 2012, 46, 559–564. [Google Scholar] [CrossRef] [PubMed]
- Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS ONE 2011, 6, e28032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shannon, K.M.; Keshavarzian, A.; Dodiya, H.B.; Jakate, S.; Kordower, J.H. Is alpha-synuclein in the colon a biomarker for premotor Parkinson’s disease? Evidence from 3 cases. Mov. Disord. 2012, 27, 716–719. [Google Scholar] [CrossRef]
- Holmqvist, S.; Chutna, O.; Bousset, L.; Aldrin-Kirk, P.; Li, W.; Bjorklund, T.; Wang, Z.Y.; Roybon, L.; Melki, R.; Li, J.Y. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 2014, 128, 805–820. [Google Scholar] [CrossRef] [Green Version]
- Svensson, E.; Horvath-Puho, E.; Thomsen, R.W.; Djurhuus, J.C.; Pedersen, L.; Borghammer, P.; Sorensen, H.T. Vagotomy and subsequent risk of Parkinson’s disease. Ann. Neurol. 2015, 78, 522–529. [Google Scholar] [CrossRef]
- Clarke, G.; Grenham, S.; Scully, P.; Fitzgerald, P.; Moloney, R.D.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol. Psychiatry 2013, 18, 666–673. [Google Scholar] [CrossRef] [Green Version]
- Van Den Berge, N.; Ferreira, N.; Gram, H.; Mikkelsen, T.W.; Alstrup, A.K.O.; Casadei, N.; Tsung-Pin, P.; Riess, O.; Nyengaard, J.R.; Tamgüney, G.; et al. Evidence for bidirectional and trans-synaptic parasympathetic and sympathetic propagation of alpha-synuclein in rats. Acta Neuropathol. 2019, 138, 535–550. [Google Scholar] [CrossRef] [Green Version]
- Manfredsson, F.P.; Luk, K.C.; Benskey, M.J.; Gezer, A.; Garcia, J.; Kuhn, N.C.; Sandoval, I.M.; Patterson, J.R.; O’Mara, A.; Yonkers, R.; et al. Induction of alpha-synuclein pathology in the enteric nervous system of the rat and non-human primate results in gastrointestinal dysmotility and transient CNS pathology. Neurobiol. Dis. 2018, 112, 106–118. [Google Scholar] [CrossRef] [PubMed]
- Challis, C.; Hori, A.; Sampson, T.R.; Yoo, B.B.; Challis, R.C.; Hamilton, A.M.; Mazmanian, S.K.; Volpicelli-Daley, L.A.; Gradinaru, V. Gut-seeded α-synuclein fibrils promote gut dysfunction and brain pathology specifically in aged mice. Nat. Neurosci. 2020, 23, 327–336. [Google Scholar] [CrossRef] [PubMed]
- Breen, D.P.; Halliday, G.M.; Lang, A.E. Gut-brain axis and the spread of α-synuclein pathology: Vagal highway or dead end? Mov. Disord. 2019, 34, 307–316. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.-J.; Chung, W.C.; Kim, B.W.; Kim, S.S.; Kim, J.I.; Kim, N.J.; Yoo, J.; Kim, S.H. Is Helicobacter pylori associated functional dyspepsia correlated with dysbiosis? J. Neurogastroenterol. Motil. 2017, 23, 504. [Google Scholar] [CrossRef] [Green Version]
- Choi, V.M.; Herrou, J.; Hecht, A.L.; Teoh, W.P.; Turner, J.R.; Crosson, S.; Bubeck Wardenburg, J. Activation of Bacteroides fragilis toxin by a novel bacterial protease contributes to anaerobic sepsis in mice. Nat. Med. 2016, 22, 563–567. [Google Scholar] [CrossRef] [Green Version]
- Seong, K.-J.; Lee, H.-G.; Kook, M.S.; Ko, H.-M.; Jung, J.-Y.; Kim, W.-J. Epigallocatechin-3-gallate rescues LPS-impaired adult hippocampal neurogenesis through suppressing the TLR4-NF-κB signaling pathway in mice. Korean J. Physiol. Pharmacol. 2016, 20, 41–51. [Google Scholar] [CrossRef]
- Pretorius, L.; Kell, D.B.; Pretorius, E. Iron Dysregulation and Dormant Microbes as Causative Agents for Impaired Blood Rheology and Pathological Clotting in Alzheimer’s Type Dementia. Front. Neurosci. 2018, 12, 851. [Google Scholar] [CrossRef]
- Kell, D.B.; Pretorius, E. Proteins behaving badly. Substoichiometric molecular control and amplification of the initiation and nature of amyloid fibril formation: Lessons from and for blood clotting. Prog. Biophys. Mol. Biol. 2017, 123, 16–41. [Google Scholar] [CrossRef]
- Furuta, N.; Takeuchi, H.; Amano, A. Entry of Porphyromonas gingivalis outer membrane vesicles into epithelial cells causes cellular functional impairment. Infect. Immun. 2009, 77, 4761–4770. [Google Scholar] [CrossRef] [Green Version]
- Poole, S.; Singhrao, S.K.; Kesavalu, L.; Curtis, M.A.; Crean, S. Determining the presence of periodontopathic virulence factors in short-term postmortem Alzheimer’s disease brain tissue. J. Alzheimer Dis. JAD 2013, 36, 665–677. [Google Scholar] [CrossRef]
- Del Prete, S.; De Luca, V.; Scozzafava, A.; Carginale, V.; Supuran, C.T.; Capasso, C. Biochemical properties of a new alpha-carbonic anhydrase from the human pathogenic bacterium, Vibrio cholerae. J. Enzym. Inhib. Med. Chem. 2014, 29, 23–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bielecka, E.; Scavenius, C.; Kantyka, T.; Jusko, M.; Mizgalska, D.; Szmigielski, B.; Potempa, B.; Enghild, J.J.; Prossnitz, E.R.; Blom, A.M.; et al. Peptidyl arginine deiminase from Porphyromonas gingivalis abolishes anaphylatoxin C5a activity. J. Biol. Chem. 2014, 289, 32481–32487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, S.G.; Stribinskis, V.; Rane, M.J.; Demuth, D.R.; Gozal, E.; Roberts, A.M.; Jagadapillai, R.; Liu, R.; Choe, K.; Shivakumar, B.; et al. Exposure to the Functional Bacterial Amyloid Protein Curli Enhances Alpha-Synuclein Aggregation in Aged Fischer 344 Rats and Caenorhabditis elegans. Sci. Rep. 2016, 6, 34477. [Google Scholar] [CrossRef] [PubMed]
- Evans, M.L.; Gichana, E.; Zhou, Y.; Chapman, M.R. Bacterial Amyloids. Methods Mol. Biol. 2018, 1779, 267–288. [Google Scholar]
- Friedland, R.P.; Chapman, M.R. The role of microbial amyloid in neurodegeneration. PLoS Pathog. 2017, 13, e1006654. [Google Scholar] [CrossRef]
- Pretorius, E.; Bester, J.; Mbotwa, S.; Robinson, C.; Kell, D.B. Acute induction of anomalous blood clotting by molecular amplification of highly substoichiometric levels of bacterial lipopolysaccharide (LPS). BioRxiv 2016, 053538. [Google Scholar] [CrossRef] [Green Version]
- Holst, O. The structures of core regions from enterobacterial lipopolysaccharides–an update. FEMS Microbiol. Let. 2007, 271, 3–11. [Google Scholar] [CrossRef]
- Steimle, A.; Autenrieth, I.B.; Frick, J.S. Structure and function: Lipid A modifications in commensals and pathogens. Int. J. Med. Microbiol 2016, 306, 290–301. [Google Scholar] [CrossRef] [Green Version]
- Ege, M.J. The Hygiene Hypothesis in the Age of the Microbiome. Ann. Am. Thorac Soc. 2017, 14 (Suppl. 5), S348–S353. [Google Scholar] [CrossRef]
- Vatanen, T.; Kostic, A.D.; d’Hennezel, E.; Siljander, H.; Franzosa, E.A.; Yassour, M.; Kolde, R.; Vlamakis, H.; Arthur, T.D.; Hämäläinen, A.M.; et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 2016, 165, 1551. [Google Scholar] [CrossRef]
- Park, B.S.; Lee, J.O. Recognition of lipopolysaccharide pattern by TLR4 complexes. Exp. Mol. Med. 2013, 45, e66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kell, D.B.; Pretorius, E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: The central roles of LPS and LPS-induced cell death. Integr. Biol. Q. Biosci. Nano Macro 2015, 7, 1339–1377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raetz, C.R.; Whitfield, C. Lipopolysaccharide endotoxins. Annu Rev. Biochem. 2002, 71, 635–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pugin, J.; Schürer-Maly, C.C.; Leturcq, D.; Moriarty, A.; Ulevitch, R.J.; Tobias, P.S. Lipopolysaccharide activation of human endothelial and epithelial cells is mediated by lipopolysaccharide-binding protein and soluble CD14. Proc. Natl. Acad. Sci. USA 1993, 90, 2744–2748. [Google Scholar] [CrossRef] [Green Version]
- Chow, J.C.; Young, D.W.; Golenbock, D.T.; Christ, W.J.; Gusovsky, F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J. Biol. Chem. 1999, 274, 10689–10692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cohen, J. The immunopathogenesis of sepsis. Nature 2002, 420, 885–891. [Google Scholar] [CrossRef]
- Takeuchi, O.; Hoshino, K.; Kawai, T.; Sanjo, H.; Takada, H.; Ogawa, T.; Takeda, K.; Akira, S. Differential roles of TLR2 and TLR4 in recognition of gram-negative and gram-positive bacterial cell wall components. Immunity 1999, 11, 443–451. [Google Scholar] [CrossRef] [Green Version]
- Muhammad, T.; Ikram, M.; Ullah, R.; Rehman, S.U.; Kim, M.O. Hesperetin, a Citrus Flavonoid, Attenuates LPS-Induced Neuroinflammation, Apoptosis and Memory Impairments by Modulating TLR4/NF-κB Signaling. Nutrients 2019, 11, 648. [Google Scholar] [CrossRef] [Green Version]
- Perez-Pardo, P.; Dodiya, H.B.; Engen, P.A.; Forsyth, C.B.; Huschens, A.M.; Shaikh, M.; Voigt, R.M.; Naqib, A.; Green, S.J.; Kordower, J.H.; et al. Role of TLR4 in the gut-brain axis in Parkinson’s disease: A translational study from men to mice. Gut 2019, 68, 829–843. [Google Scholar] [CrossRef]
- Lu, Y.C.; Yeh, W.C.; Ohashi, P.S. LPS/TLR4 signal transduction pathway. Cytokine 2008, 42, 145–151. [Google Scholar] [CrossRef]
- Patel, H.; Fellowes, R.; Coade, S.; Woo, P. Human serum amyloid A has cytokine-like properties. Scand. J. Immunol. 1998, 48, 410–418. [Google Scholar] [CrossRef] [PubMed]
- Man, S.M.; Kanneganti, T.D. Regulation of inflammasome activation. Immun. Rev. 2015, 265, 6–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, W.-G.; Mohney, R.P.; Wilson, B.; Jeohn, G.-H.; Liu, B.; Hong, J.-S. Regional difference in susceptibility to lipopolysaccharide-induced neurotoxicity in the rat brain: Role of microglia. J. Neurosci. 2000, 20, 6309–6316. [Google Scholar] [CrossRef] [PubMed]
- Qin, L.; Liu, Y.; Wang, T.; Wei, S.J.; Block, M.L.; Wilson, B.; Liu, B.; Hong, J.S. NADPH oxidase mediates lipopolysaccharide-induced neurotoxicity and proinflammatory gene expression in activated microglia. J. Biol. Chem. 2004, 279, 1415–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sumi, N.; Nishioku, T.; Takata, F.; Matsumoto, J.; Watanabe, T.; Shuto, H.; Yamauchi, A.; Dohgu, S.; Kataoka, Y. Lipopolysaccharide-activated microglia induce dysfunction of the blood-brain barrier in rat microvascular endothelial cells co-cultured with microglia. Cell Mol. Neurobiol. 2010, 30, 247–253. [Google Scholar] [CrossRef]
- di Penta, A.; Moreno, B.; Reix, S.; Fernandez-Diez, B.; Villanueva, M.; Errea, O.; Escala, N.; Vandenbroeck, K.; Comella, J.X.; Villoslada, P. Oxidative stress and proinflammatory cytokines contribute to demyelination and axonal damage in a cerebellar culture model of neuroinflammation. PLoS ONE 2013, 8, e54722. [Google Scholar] [CrossRef] [Green Version]
- Gao, H.-M.; Zhang, F.; Zhou, H.; Kam, W.; Wilson, B.; Hong, J.-S. Neuroinflammation and α-synuclein dysfunction potentiate each other, driving chronic progression of neurodegeneration in a mouse model of Parkinson’s disease. Environ. Health Perspect. 2011, 119, 807–814. [Google Scholar] [CrossRef] [Green Version]
- Rutherford, N.J.; Sacino, A.N.; Brooks, M.; Ceballos-Diaz, C.; Ladd, T.B.; Howard, J.K.; Golde, T.E.; Giasson, B.I. Studies of lipopolysaccharide effects on the induction of alpha-synuclein pathology by exogenous fibrils in transgenic mice. Mol. Neurodegener. 2015, 10, 32. [Google Scholar] [CrossRef] [Green Version]
- Dutta, G.; Zhang, P.; Liu, B. The lipopolysaccharide Parkinson’s disease animal model: Mechanistic studies and drug discovery. Fundam. Clin. Pharmacol. 2008, 22, 453–464. [Google Scholar] [CrossRef]
- Nava Catorce, M.; Gevorkian, G. LPS-induced murine neuroinflammation model: Main features and suitability for pre-clinical assessment of nutraceuticals. Curr. Neuropharmacol. 2016, 14, 155–164. [Google Scholar] [CrossRef] [Green Version]
- Sheng, J.G.; Bora, S.H.; Xu, G.; Borchelt, D.R.; Price, D.L.; Koliatsos, V.E. Lipopolysaccharide-induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid beta peptide in APPswe transgenic mice. Neurobiol. Dis. 2003, 14, 133–145. [Google Scholar] [CrossRef]
- Faas, M.M.; Schuiling, G.A.; Baller, J.F.; Visscher, C.A.; Bakker, W.W. A new animal model for human preeclampsia: Ultra-lowdose endotoxin infusion in pregnant rats. Am. J. Obstet. Gynecol. 1994, 171, 158–164. [Google Scholar] [CrossRef]
- Kell, D.B.; Kenny, L.C. A Dormant Microbial Component in the Development of Preeclampsia. Front. Med. 2016, 3, 60. [Google Scholar] [CrossRef] [Green Version]
- Kenny, L.C.; Kell, D.B. Immunological Tolerance, Pregnancy, and Preeclampsia: The Roles of Semen Microbes and the Father. Front. Med. 2017, 4, 239. [Google Scholar] [CrossRef] [Green Version]
- Wispelwey, B.; Lesse, A.J.; Hansen, E.J.; Scheld, W.M. Haemophilus influenzae lipopolysaccharide-induced blood brain barrier permeability during experimental meningitis in the rat. J. Clin. Investig. 1988, 82, 1339–1346. [Google Scholar] [CrossRef] [PubMed]
- Tomas-Camardiel, M.; Rite, I.; Herrera, A.J.; de Pablos, R.M.; Cano, J.; Machado, A.; Venero, J.L. Minocycline reduces the lipopolysaccharide-induced inflammatory reaction, peroxynitrite-mediated nitration of proteins, disruption of the blood-brain barrier, and damage in the nigral dopaminergic system. Neurobiol. Dis. 2004, 16, 190–201. [Google Scholar] [CrossRef] [PubMed]
- Bohatschek, M.; Werner, A.; Raivich, G. Systemic LPS injection leads to granulocyte influx into normal and injured brain: Effects of ICAM-1 deficiency. Exp. Neurol. 2001, 172, 137–152. [Google Scholar] [CrossRef]
- Koprich, J.B.; Reske-Nielsen, C.; Mithal, P.; Isacson, O. Neuroinflammation mediated by IL-1beta increases susceptibility of dopamine neurons to degeneration in an animal model of Parkinson’s disease. J. Neuroinflamm. 2008, 5, 8. [Google Scholar] [CrossRef] [Green Version]
- Sui, Y.T.; Bullock, K.M.; Erickson, M.A.; Zhang, J.; Banks, W.A. Alpha synuclein is transported into and out of the brain by the blood-brain barrier. Peptides 2014, 62, 197–202. [Google Scholar] [CrossRef] [Green Version]
- Vekilov, P.G. Phase diagrams and kinetics of phase transitions in protein solutions. J. Phys. Condens Matter 2012, 24, 193101. [Google Scholar] [CrossRef]
- Oosawa, F.; Kasai, M. A theory of linear and helical aggregations of macromolecules. J. Mol. Biol 1962, 4, 10–21. [Google Scholar] [CrossRef]
- Ruschak, A.M.; Miranker, A.D. Fiber-dependent amyloid formation as catalysis of an existing reaction pathway. Proc. Natl. Acad. Sci. USA 2007, 104, 12341–12346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cohen, S.I.; Linse, S.; Luheshi, L.M.; Hellstrand, E.; White, D.A.; Rajah, L.; Otzen, D.E.; Vendruscolo, M.; Dobson, C.M.; Knowles, T.P. Proliferation of amyloid-β42 aggregates occurs through a secondary nucleation mechanism. Proc. Natl. Acad. Sci. USA 2013, 110, 9758–9763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cohen, S.I.; Vendruscolo, M.; Dobson, C.M.; Knowles, T.P. Nucleated polymerization with secondary pathways. II. Determination of self-consistent solutions to growth processes described by non-linear master equations. J. Chem. Phys. 2011, 135, 065106. [Google Scholar] [CrossRef] [Green Version]
- Luhrs, T.; Ritter, C.; Adrian, M.; Riek-Loher, D.; Bohrmann, B.; Dobeli, H.; Schubert, D.; Riek, R. 3D structure of Alzheimer’s amyloid-beta(1–42) fibrils. Proc. Natl. Acad. Sci. USA 2005, 102, 17342–17347. [Google Scholar] [CrossRef] [Green Version]
- Galvagnion, C.; Buell, A.K.; Meisl, G.; Michaels, T.C.; Vendruscolo, M.; Knowles, T.P.; Dobson, C.M. Lipid vesicles trigger α-synuclein aggregation by stimulating primary nucleation. Nat. Chem. Biol. 2015, 11, 229–234. [Google Scholar] [CrossRef] [Green Version]
- Pretorius, E.; Page, M.J.; Hendricks, L.; Nkosi, N.B.; Benson, S.R.; Kell, D.B. Both lipopolysaccharide and lipoteichoic acids potently induce anomalous fibrin amyloid formation: Assessment with novel Amytracker™ stains. J. R. Soc. Interface 2018, 15, 20170941. [Google Scholar] [CrossRef] [Green Version]
- Kaur, T.; Uppoor, A.; Naik, D. Parkinson’s disease and periodontitis—The missing link? A review. Gerodontology 2016, 33, 434–438. [Google Scholar] [CrossRef]
- Pradeep, A.R.; Singh, S.P.; Martande, S.S.; Raju, A.P.; Rustagi, T.; Suke, D.K.; Naik, S.B. Clinical evaluation of the periodontal health condition and oral health awareness in Parkinson’s disease patients. Gerodontology 2015, 32, 100–106. [Google Scholar] [CrossRef]
- Cicciu, M.; Risitano, G.; Lo Giudice, G.; Bramanti, E. Periodontal health and caries prevalence evaluation in patients affected by Parkinson’s disease. Parkinsons Dis. 2012, 2012, 541908. [Google Scholar] [CrossRef]
- Schwarz, J.; Heimhilger, E.; Storch, A. Increased periodontal pathology in Parkinson’s disease. J. Neurol. 2006, 253, 608–611. [Google Scholar] [CrossRef] [PubMed]
- Liu, T.C.; Sheu, J.J.; Lin, H.C.; Jensen, D.A. Increased risk of parkinsonism following chronic periodontitis: A retrospective cohort study. Mov. Disord. 2013, 28, 1307–1308. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.-K.; Wu, Y.-T.; Chang, Y.-C. Periodontal inflammatory disease is associated with the risk of Parkinson’s disease: A population-based retrospective matched-cohort study. PeerJ 2017, 5, e3647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shanker, J.; Setty, P.; Arvind, P.; Nair, J.; Bhasker, D.; Balakrishna, G.; Kakkar, V.V. Relationship between periodontal disease, Porphyromonas gingivalis, peripheral vascular resistance markers and coronary artery disease in Asian Indians. Thromb. Res. 2013, 132, e8–e14. [Google Scholar] [CrossRef] [PubMed]
- Amar, S.; Engelke, M. Periodontal Innate Immune Mechanisms Relevant to Atherosclerosis. Mol. Oral Microbiol. 2014. [Google Scholar]
- Singhrao, S.K.; Harding, A.; Simmons, T.; Robinson, S.; Kesavalu, L.; Crean, S. Oral inflammation, tooth loss, risk factors, and association with progression of Alzheimer’s disease. J. Alzheimer Dis. JAD 2014, 42, 723–737. [Google Scholar] [CrossRef] [Green Version]
- Dominy, S.S.; Lynch, C.; Ermini, F.; Benedyk, M.; Marczyk, A.; Konradi, A.; Nguyen, M.; Haditsch, U.; Raha, D.; Griffin, C.; et al. Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci. Adv. 2019, 5, eaau3333. [Google Scholar] [CrossRef] [Green Version]
- Olsen, I.; Singhrao, S.K. Is there a link between genetic defects in the complement cascade and Porphyromonas gingivalis in Alzheimer’s disease? J. Oral Microbiol. 2020, 12, 1676486. [Google Scholar] [CrossRef] [Green Version]
- Nakajima, M.; Arimatsu, K.; Kato, T.; Matsuda, Y.; Minagawa, T.; Takahashi, N.; Ohno, H.; Yamazaki, K. Oral Administration of P. gingivalis Induces Dysbiosis of Gut Microbiota and Impaired Barrier Function Leading to Dissemination of Enterobacteria to the Liver. PLoS ONE 2015, 10, e0134234. [Google Scholar] [CrossRef] [Green Version]
- Arimatsu, K.; Yamada, H.; Miyazawa, H.; Minagawa, T.; Nakajima, M.; Ryder, M.I.; Gotoh, K.; Motooka, D.; Nakamura, S.; Iida, T.; et al. Oral pathobiont induces systemic inflammation and metabolic changes associated with alteration of gut microbiota. Sci. Rep. 2014, 4, 4828. [Google Scholar] [CrossRef] [Green Version]
- Kato, T.; Yamazaki, K.; Nakajima, M.; Date, Y.; Kikuchi, J.; Hase, K.; Ohno, H.; Yamazaki, K. Oral Administration of Porphyromonas gingivalis Alters the Gut Microbiome and Serum Metabolome. mSphere 2018, 3, e0046018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakajima, Y.; Ito, K.; Yoshimoto, T. Prolyl Tripeptidylpeptidase. In Handbook of Proteolytic Enzymes; Academic Press: Cambridge, MA, USA, 2013; Volume 3, pp. 3371–3374. [Google Scholar]
- Devos, D.; Moreau, C.; Devedjian, J.C.; Kluza, J.; Petrault, M.; Laloux, C.; Jonneaux, A.; Ryckewaert, G.; Garçon, G.; Rouaix, N. Targeting chelatable iron as a therapeutic modality in Parkinson’s disease. Antioxid. Redox Signal. 2014, 21, 195–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jomova, K.; Valko, M. Importance of iron chelation in free radical-induced oxidative stress and human disease. Curr. Pharm. Des. 2011, 17, 3460–3473. [Google Scholar] [CrossRef] [PubMed]
- Masaldan, S.; Bush, A.I.; Devos, D.; Rolland, A.S.; Moreau, C. Striking while the iron is hot: Iron metabolism and ferroptosis in neurodegeneration. Free Radic. Biol. Med. 2019, 133, 221–233. [Google Scholar] [CrossRef]
- Zecca, L.; Casella, L.; Albertini, A.; Bellei, C.; Zucca, F.A.; Engelen, M.; Zadlo, A.; Szewczyk, G.; Zareba, M.; Sarna, T. Neuromelanin can protect against iron-mediated oxidative damage in system modeling iron overload of brain aging and Parkinson’s disease. J. Neurochem. 2008, 106, 1866–1875. [Google Scholar]
- 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]
- Youdim, M.B.H.; Stephenson, G.; Shachar, D.B. Ironing iron out in Parkinson’s disease and other neurodegenerative diseases with iron chelators: A lesson from 6-hydroxydopamine and iron chelators, desferal and VK-28. Ann. N. Y. Acad. Sci. 2004, 1012, 306–325. [Google Scholar] [CrossRef]
- Bar-Am, O.; Amit, T.; Kupershmidt, L.; Aluf, Y.; Mechlovich, D.; Kabha, H.; Danovitch, L.; Zurawski, V.R.; Youdim, M.B.; Weinreb, O. Neuroprotective and neurorestorative activities of a novel iron chelator-brain selective monoamine oxidase-A/monoamine oxidase-B inhibitor in animal models of Parkinson’s disease and aging. Neurobiol. Aging 2015, 36, 1529–1542. [Google Scholar] [CrossRef]
- Nuñez, M.T.; Urrutia, P.; Mena, N.; Aguirre, P.; Tapia, V.; Salazar, J. Iron toxicity in neurodegeneration. Biometals 2012, 25, 761–776. [Google Scholar] [CrossRef]
- Nuñez, M.T.; Chana-Cuevas, P. New Perspectives in Iron Chelation Therapy for the Treatment of Neurodegenerative Diseases. Pharmaceuticals 2018, 11, 109. [Google Scholar] [CrossRef] [Green Version]
- Youdim, M.; Fridkin, M.; Zheng, H. Novel bifunctional drugs targeting monoamine oxidase inhibition and iron chelation as an approach to neuroprotection in Parkinson’s disease and other neurodegenerative diseases. J. Neural. Transm. 2004, 111, 1455–1471. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Wan, J.; Lan, X.; Han, X.; Wang, Z.; Wang, J. Neuroprotection of brain-permeable iron chelator VK-28 against intracerebral hemorrhage in mice. J. Cereb. Blood Flow Metab. 2017, 37, 3110–3123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin-Bastida, A.; Ward, R.J.; Newbould, R.; Piccini, P.; Sharp, D.; Kabba, C.; Patel, M.C.; Spino, M.; Connelly, J.; Tricta, F.; et al. Brain iron chelation by deferiprone in a phase 2 randomised double-blinded placebo controlled clinical trial in Parkinson’s disease. Sci. Rep. 2017, 7, 1398. [Google Scholar] [CrossRef] [PubMed]
- Klopstock, T.; Tricta, F.; Neumayr, L.; Karin, I.; Zorzi, G.; Fradette, C.; Kmieć, T.; Büchner, B.; Steele, H.E.; Horvath, R.; et al. Safety and efficacy of deferiprone for pantothenate kinase-associated neurodegeneration: A randomised, double-blind, controlled trial and an open-label extension study. Lancet Neurol. 2019, 18, 631–642. [Google Scholar] [CrossRef]
- 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]
- Ghosh, B.; Antonio, T.; Reith, M.E.; Dutta, A.K. Discovery of 4-(4-(2-((5-Hydroxy-1, 2, 3, 4-tetrahydronaphthalen-2-yl)(propyl) amino) ethyl) piperazin-1-yl) quinolin-8-ol and its analogues as highly potent dopamine D2/D3 agonists and as iron chelator: In vivo activity indicates potential application in symptomatic and neuroprotective therapy for Parkinson’s disease. J. Med. Chem. 2010, 53, 2114–2125. [Google Scholar]
- Aguirre, P.; Garcia-Beltran, O.; Tapia, V.; Munoz, Y.; Cassels, B.K.; Nunez, M.T. Neuroprotective Effect of a New 7,8-Dihydroxycoumarin-Based Fe(2+)/Cu(2+) Chelator in Cell and Animal Models of Parkinson’s Disease. ACS Chem. Neurosci. 2017, 8, 178–185. [Google Scholar] [CrossRef]
- Garcia-Beltran, O.; Mena, N.P.; Aguirre, P.; Barriga-Gonzalez, G.; Galdamez, A.; Nagles, E.; Adasme, T.; Hidalgo, C.; Nunez, M.T. Development of an iron-selective antioxidant probe with protective effects on neuronal function. PLoS ONE 2017, 12, e0189043. [Google Scholar] [CrossRef] [Green Version]
- Borodina, I.; Kenny, L.C.; McCarthy, C.M.; Paramasivan, K.; Pretorius, E.; Roberts, T.J.; van der Hoek, S.A.; Kell, D.B. The biology of ergothioneine, an antioxidant nutraceutical. Nutr. Res. Rev. 2020, 33, 1–28. [Google Scholar] [CrossRef] [Green Version]
- Dutta, S.K.; Verma, S.; Jain, V.; Surapaneni, B.K.; Vinayek, R.; Phillips, L.; Nair, P.P. Parkinson’s Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. J. Neurogastroenterol. Motil. 2019, 25, 363–376. [Google Scholar] [CrossRef] [Green Version]
- Barboza, J.L.; Okun, M.S.; Moshiree, B. The treatment of gastroparesis, constipation and small intestinal bacterial overgrowth syndrome in patients with Parkinson’s disease. Exp. Opin. Pharmacother. 2015, 16, 2449–2464. [Google Scholar] [CrossRef] [PubMed]
- Barichella, M.; Pacchetti, C.; Bolliri, C.; Cassani, E.; Iorio, L.; Pusani, C.; Pinelli, G.; Privitera, G.; Cesari, I.; Faierman, S.A.; et al. Probiotics and prebiotic fiber for constipation associated with Parkinson disease: An RCT. Neurology 2016, 87, 1274–1280. [Google Scholar] [CrossRef]
- Gazerani, P. Probiotics for Parkinson’s Disease. Int. J. Mol. Sci. 2019, 20, 4121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uyar, G.; Yildiran, H. A nutritional approach to microbiota in Parkinson’s disease. Biosci. Microbiota Food Health 2019, 38, 115–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramprasad, C.; Douglas, J.Y.; Moshiree, B. Parkinson’s Disease and Current Treatments for Its Gastrointestinal Neurogastromotility Effects. Curr. Treat. Options Gastroenterol. 2018, 16, 489–510. [Google Scholar] [CrossRef] [PubMed]
- Radad, K.; Moldzio, R.; Rausch, W.D. Minocycline protects dopaminergic neurons against long-term rotenone toxicity. Can. J. Neurol. Sci. 2010, 37, 81–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomas, M.; Le, W.D. Minocycline: Neuroprotective mechanisms in Parkinson’s disease. Curr Pharm Des. 2004, 10, 679–686. [Google Scholar] [CrossRef]
- Kim, H.S.; Suh, Y.H. Minocycline and neurodegenerative diseases. Behav. Brain Res. 2009, 196, 168–179. [Google Scholar] [CrossRef]
- Diguet, E.; Fernagut, P.O.; Wei, X.; Du, Y.; Rouland, R.; Gross, C.; Bezard, E.; Tison, F. Deleterious effects of minocycline in animal models of Parkinson’s disease and Huntington’s disease. Eur. J. Neurosci. 2004, 19, 3266–3276. [Google Scholar] [CrossRef]
- Yulug, B.; Hanoglu, L.; Kilic, E.; Schabitz, W.R. RIFAMPICIN: An antibiotic with brain protective function. Brain Res. Bull. 2014, 107, 37–42. [Google Scholar] [CrossRef]
- Bi, W.; Zhu, L.; Jing, X.; Liang, Y.; Tao, E. Rifampicin and Parkinson’s disease. Neurol. Sci. 2013, 34, 137–141. [Google Scholar] [CrossRef] [PubMed]
- Acuña, L.; Hamadat, S.; Corbalán, N.S.; González-Lizárraga, F.; Dos-Santos-Pereira, M.; Rocca, J.; Díaz, J.S.; Del-Bel, E.; Papy-García, D.; Chehín, R.N.; et al. Rifampicin and Its Derivative Rifampicin Quinone Reduce Microglial Inflammatory Responses and Neurodegeneration Induced In Vitro by α-Synuclein Fibrillary Aggregates. Cells 2019, 8, 776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, D.; Jing, X.; Chen, Y.; Liang, Y.; Lei, M.; Peng, S.; Zhou, T.; Zheng, D.; Zeng, Z.; Wu, X.; et al. Rifampicin pre-treatment inhibits the toxicity of rotenone-induced PC12 cells by enhancing sumoylation modification of α-synuclein. Biochem. Biophys. Res. Commun. 2017, 485, 23–29. [Google Scholar] [CrossRef] [PubMed]
- Henley, J.M.; Carmichael, R.E.; Wilkinson, K.A. Extranuclear SUMOylation in Neurons. Trends Neurosci. 2018, 41, 198–210. [Google Scholar] [CrossRef] [Green Version]
- Guerra de Souza, A.C.; Prediger, R.D.; Cimarosti, H. SUMO-regulated mitochondrial function in Parkinson’s disease. J. Neurochem. 2016, 137, 673–686. [Google Scholar] [CrossRef]
- Mertsalmi, T.H.; Pekkonen, E.; Scheperjans, F. Antibiotic exposure and risk of Parkinson’s disease in Finland: A nationwide case-control study. Mov. Disord. 2020, 35, 431–442. [Google Scholar] [CrossRef]
- Vaiserman, A.M.; Koliada, A.K.; Marotta, F. Gut microbiota: A player in aging and a target for anti-aging intervention. Ageing Res. Rev. 2017, 35, 36–45. [Google Scholar] [CrossRef]
- Endres, K.; Schäfer, K.H. Influence of Commensal Microbiota on the Enteric Nervous System and Its Role in Neurodegenerative Diseases. J. Innate Immun. 2018, 10, 172–180. [Google Scholar] [CrossRef]
- Huang, H.; Xu, H.; Luo, Q.; He, J.; Li, M.; Chen, H.; Tang, W.; Nie, Y.; Zhou, Y. Fecal microbiota transplantation to treat Parkinson’s disease with constipation: A case report. Medicine 2019, 98, e16163. [Google Scholar] [CrossRef]
- Sun, M.F.; Zhu, Y.L.; Zhou, Z.L.; Jia, X.B.; Xu, Y.D.; Yang, Q.; Cui, C.; Shen, Y.Q. Neuroprotective effects of fecal microbiota transplantation on MPTP-induced Parkinson’s disease mice: Gut microbiota, glial reaction and TLR4/TNF-alpha signaling pathway. Brain Behav. Immun. 2018, 70, 48–60. [Google Scholar] [CrossRef]
- Nguyen, S.V.; Nguyen, M.T.H.; Tran, B.C.; Ho, M.T.Q.; Umeda, K.; Rahman, S. Evaluation of lozenges containing egg yolk antibody against Porphyromonas gingivalis gingipains as an adjunct to conventional non-surgical therapy in periodontitis patients: A randomized controlled clinical trial. J. Periodontol. 2018, 89, 1334–1339. [Google Scholar] [CrossRef] [PubMed]
Mediators of Inflammation | References |
---|---|
Presence of activated microglia, dysregulated inflammatory mediators, chemokines, oxidative stress, and both systemic and CNS inflammation | [54,83,84,85,86,87] |
Presence of dysregulated cytokines, including interleukin (IL)-1β, IL-2, IL-6, IL-10, tumour necrosis factor (TNF)-α, interferon (IFN)γ, RANTES), and C-reactive protein (CRP) | [45,88,89,90,91,92,93] |
Increased cluster of differentiation (CD) 4+ T-cells indicating peripheral lymphocyte activation | [94,95] |
Presence of nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) in postmortem PD brains | [96,97] |
Increase in gut–brain axis and intestinal inflammation. An increase in enteric inflammation associated with increased mRNA and mRNA that are associated with glial markers | [10,42,43,98,99] |
Increased presence of stool immune factors | [100,101] |
Dysregulated bacterial inflammagens like LPS (lipopolysaccharides) and bacterial proteases like gingipains | [45,102,103,104] |
Iron dysregulation | [20,50,62,63,105,106,107,108,109,110,111,112,113,114,115] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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
Vuuren, M.J.v.; Nell, T.A.; Carr, J.A.; Kell, D.B.; Pretorius, E. Iron Dysregulation and Inflammagens Related to Oral and Gut Health Are Central to the Development of Parkinson’s Disease. Biomolecules 2021, 11, 30. https://doi.org/10.3390/biom11010030
Vuuren MJv, Nell TA, Carr JA, Kell DB, Pretorius E. Iron Dysregulation and Inflammagens Related to Oral and Gut Health Are Central to the Development of Parkinson’s Disease. Biomolecules. 2021; 11(1):30. https://doi.org/10.3390/biom11010030
Chicago/Turabian StyleVuuren, Marthinus Janse van, Theodore Albertus Nell, Jonathan Ambrose Carr, Douglas B. Kell, and Etheresia Pretorius. 2021. "Iron Dysregulation and Inflammagens Related to Oral and Gut Health Are Central to the Development of Parkinson’s Disease" Biomolecules 11, no. 1: 30. https://doi.org/10.3390/biom11010030
APA StyleVuuren, M. J. v., Nell, T. A., Carr, J. A., Kell, D. B., & Pretorius, E. (2021). Iron Dysregulation and Inflammagens Related to Oral and Gut Health Are Central to the Development of Parkinson’s Disease. Biomolecules, 11(1), 30. https://doi.org/10.3390/biom11010030