The Emerging Scenario of the Gut–Brain Axis: The Therapeutic Actions of the New Actor Kefir against Neurodegenerative Diseases
Abstract
:1. Introduction
2. Gut Microbiota: An Extraordinary Army of Fighters to Defend the Host
3. Gut–Brain Axis: Extraordinary Advances and Opportunities to Improve the Treatment of Dysbiosis
4. The Brain–Heart–Kidney Interconnection: The Role of Kefir
5. Lessons on How to Fight Oxidative Stress
6. Probiotics in ND: Why Would It Be a Useful Tool for Drug Interactions?
7. The Symbiome and Pathobiome: A New Understanding of the Gut in ND
8. Brief History and Biochemical Properties of Kefir: A Basis for Appropriate Supplementation
9. Targeting the Near Future: Milk Kefir against Oxidative Stress and Inflammation
10. ND and Kefir: Promise or Reality?
10.1. What Is New in Dementia?
10.2. Encephalitis and Kefir: A New Insight
10.3. Fighting PD with Kefir: Current Scenario and Future Horizons
11. Conclusions and Perspectives for Future Research Advances
Author Contributions
Funding
Conflicts of Interest
References
- Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stephenson, J.; Nutma, E.; van der Valk, P.; Amor, S. Inflammation in CNS neurodegenerative diseases. Immunology 2018, 154, 204–219. [Google Scholar] [CrossRef] [Green Version]
- Salman, M.M.; Al-Obaidi, Z.; Kitchen, P.; Loreto, A.; Bill, R.M.; Wade-Martins, R. Advances in Applying Computer-Aided Drug Design for Neurodegenerative Diseases. Int. J. Mol. Sci. 2021, 22, 4688. [Google Scholar] [CrossRef] [PubMed]
- Aldewachi, H.; Al-Zidan, R.N.; Conner, M.T.; Salman, M.M. High-Throughput Screening Platforms in the Discovery of Novel Drugs for Neurodegenerative Diseases. Bioengineering 2021, 8, 30. [Google Scholar] [CrossRef]
- Livingston, G.; Huntley, J.; Sommerlad, A.; Ames, D.; Ballard, C.; Banerjee, S.; Brayne, C.; Burns, A.; Cohen-Mansfield, J.; Cooper, C.; et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet 2020, 396, 413–446. [Google Scholar] [CrossRef]
- Rodríguez, J.M.; Murphy, K.; Stanton, C.; Ross, R.P.; Kober, O.I.; Juge, N.; Avershina, E.; Rudi, K.; Narbad, A.; Jenmalm, M.C.; et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 2015, 26, 26050. [Google Scholar] [CrossRef] [PubMed]
- Friques, A.G.; Arpini, C.M.; Kalil, I.C.; Gava, A.L.; Leal, M.A.; Porto, M.L.; Nogueira, B.V.; Dias, A.T.; Andrade, T.U.; Pereira, T.M.; et al. Chronic administration of the probiotic kefir improves the endothelial function in spontaneously hypertensive rats. J. Transl. Med. 2015, 13, 390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- von Mutius, E. The shape of the microbiome in early life. Nat. Med. 2017, 23, 274–275. [Google Scholar] [CrossRef] [PubMed]
- Amorim, F.G.; Coitinho, L.B.; Dias, A.T.; Friques, A.G.F.; Monteiro, B.L.; Rezende, L.C.D.; Pereira, T.M.C.; Campagnaro, B.P.; De Pauw, E.; Vasquez, E.C.; et al. Identification of new bioactive peptides from Kefir milk through proteopeptidomics: Bioprospection of antihypertensive molecules. Food Chem. 2019, 282, 109–119. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [Green Version]
- Tang, W.H.; Kitai, T.; Hazen, S.L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017, 120, 1183–1196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guarner, F.; Malagelada, J.R. Gut flora in health and disease. Lancet 2003, 361, 512–519. [Google Scholar] [CrossRef]
- Koenig, J.E.; Spor, A.; Scalfone, N.; Fricker, A.D.; Stombaugh, J.; Knight, R.; Angenent, L.T.; Ley, R.E. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. 1), 4578–4585. [Google Scholar] [CrossRef] [Green Version]
- Friques, A.G.F.; Santos, F.D.N.; Angeli, D.B.; Silva, F.A.C.; Dias, A.T.; Aires, R.; Leal, M.A.S.; Nogueira, B.V.; Amorim, F.G.; Campagnaro, B.P.; et al. Bisphenol A contamination in infant rats: Molecular, structural, and physiological cardiovascular changes and the protective role of kefir. J. Nutr. Biochem. 2020, 75, 108254. [Google Scholar] [CrossRef] [PubMed]
- Zheng, H.; Xu, P.; Jiang, Q.; Xu, Q.; Zheng, Y.; Yan, J.; Ji, H.; Ning, J.; Zhang, X.; Li, C.; et al. Depletion of acetate-producing bacteria from the gut microbiota facilitates cognitive impairment through the gut-brain neural mechanism in diabetic mice. Microbiome 2021, 9, 145. [Google Scholar] [CrossRef] [PubMed]
- Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef] [PubMed]
- Goswami, C.; Iwasaki, Y.; Yada, T. Short-chain fatty acids suppress food intake by activating vagal afferent neurons. J. Nutr. Biochem. 2018, 57, 130–135. [Google Scholar] [CrossRef] [PubMed]
- Zengeler, K.E.; Lukens, J.R. Innate immunity at the crossroads of healthy brain maturation and neurodevelopmental disorders. Nat. Rev. Immunol. 2021, 21, 454–468. [Google Scholar] [CrossRef]
- Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L.; et al. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [Green Version]
- Kalbermatter, C.; Fernandez Trigo, N.; Christensen, S.; Ganal-Vonarburg, S.C. Maternal Microbiota, Early Life Colonization and Breast Milk Drive Immune Development in the Newborn. Front. Immunol. 2021, 12, 683022. [Google Scholar] [CrossRef] [PubMed]
- Müller, J.E.; Meyer, N.; Santamaria, C.G.; Schumacher, A.; Luque, E.H.; Zenclussen, M.L.; Rodriguez, H.A.; Zenclussen, A.C. Bisphenol A exposure during early pregnancy impairs uterine spiral artery remodeling and provokes intrauterine growth restriction in mice. Sci. Rep. 2018, 8, 9196. [Google Scholar] [CrossRef]
- Nagpal, R.; Tsuji, H.; Takahashi, T.; Nomoto, K.; Kawashima, K.; Nagata, S.; Yamashiro, Y. Gut dysbiosis following C-section instigates higher colonisation of toxigenic Clostridium perfringens in infants. Benef. Microbes 2017, 8, 353–365. [Google Scholar] [CrossRef] [PubMed]
- D’Argenio, V. The Prenatal Microbiome: A New Player for Human Health. High-Throughput 2018, 7, 38. [Google Scholar] [CrossRef] [Green Version]
- Boudry, G.; Charton, E.; Le Huerou-Luron, I.; Ferret-Bernard, S.; Le Gall, S.; Even, S.; Blat, S. The Relationship Between Breast Milk Components and the Infant Gut Microbiota. Front. Nutr. 2021, 8, 629740. [Google Scholar] [CrossRef] [PubMed]
- Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
- Kanasi, E.; Ayilavarapu, S.; Jones, J. The aging population: Demographics and the biology of aging. Periodontol. 2000 2016, 72, 13–18. [Google Scholar] [CrossRef]
- Firoz, C.K.; Jabir, N.R.; Khan, M.S.; Mahmoud, M.; Shakil, S.; Damanhouri, G.A.; Zaidi, S.K.; Tabrez, S.; Kamal, M.A. An overview on the correlation of neurological disorders with cardiovascular disease. Saudi J. Biol. Sci. 2015, 22, 19–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhen, S.; Ma, Y.; Zhao, Z.; Yang, X.; Wen, D. Dietary pattern is associated with obesity in Chinese children and adolescents: Data from China Health and Nutrition Survey (CHNS). Nutr. J. 2018, 17, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lassale, C.; Fezeu, L.; Andreeva, V.A.; Hercberg, S.; Kengne, A.P.; Czernichow, S.; Kesse-Guyot, E. Association between dietary scores and 13-year weight change and obesity risk in a French prospective cohort. Int. J. Obes. 2012, 36, 1455–1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, L.; Stamler, J.; Chan, Q.; Van Horn, L.; Daviglus, M.L.; Dyer, A.R.; Miura, K.; Okuda, N.; Wu, Y.; Ueshima, H.; et al. Salt intake and prevalence of overweight/obesity in Japan, China, the United Kingdom, and the United States: The INTERMAP Study. Am. J. Clin. Nutr. 2019, 110, 34–40. [Google Scholar] [CrossRef]
- Jang, L.G.; Choi, G.; Kim, S.W.; Kim, B.Y.; Lee, S.; Park, H. The combination of sport and sport-specific diet is associated with characteristics of gut microbiota: An observational study. J. Int. Soc. Sports Nutr. 2019, 16, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
- Coman, V.; Vodnar, D.C. Gut microbiota and old age: Modulating factors and interventions for healthy longevity. Exp. Gerontol. 2020, 141, 111095. [Google Scholar] [CrossRef]
- Li, N.; Zuo, B.; Huang, S.; Zeng, B.; Han, D.; Li, T.; Liu, T.; Wu, Z.; Wei, H.; Zhao, J.; et al. Spatial heterogeneity of bacterial colonization across different gut segments following inter-species microbiota transplantation. Microbiome 2020, 8, 161. [Google Scholar] [CrossRef] [PubMed]
- Hemarajata, P.; Versalovic, J. Effects of probiotics on gut microbiota: Mechanisms of intestinal immunomodulation and neuromodulation. Ther. Adv. Gastroenterol. 2013, 6, 39–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Vos, W.M. Microbe Profile: Akkermansia muciniphila: A conserved intestinal symbiont that acts as the gatekeeper of our mucosa. Microbiology 2017, 163, 646–648. [Google Scholar] [CrossRef] [PubMed]
- Fitzgerald, C.B.; Shkoporov, A.N.; Sutton, T.D.S.; Chaplin, A.V.; Velayudhan, V.; Ross, R.P.; Hill, C. Comparative analysis of Faecalibacterium prausnitzii genomes shows a high level of genome plasticity and warrants separation into new species-level taxa. BMC Genom. 2018, 19, 931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Callaghan, A.; van Sinderen, D. Bifidobacteria and Their Role as Members of the Human Gut Microbiota. Front. Microbiol. 2016, 7, 925. [Google Scholar] [CrossRef] [Green Version]
- Mariat, D.; Firmesse, O.; Levenez, F.; Guimarăes, V.; Sokol, H.; Doré, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009, 9, 123. [Google Scholar] [CrossRef]
- Ton, A.M.M.; Campagnaro, B.P.; Alves, G.A.; Aires, R.; Côco, L.Z.; Arpini, C.M.; Guerra, E.O.T.; Campos-Toimil, M.; Meyrelles, S.S.; Pereira, T.M.C.; et al. Oxidative Stress and Dementia in Alzheimer’s Patients: Effects of Synbiotic Supplementation. Oxid. Med. Cell. Longev. 2020, 2020, 2638703. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.H.; Jeong, D.; Kim, H.; Seo, K.H. Modern perspectives on the health benefits of kefir in next generation sequencing era: Improvement of the host gut microbiota. Crit. Rev. Food Sci. Nutr. 2019, 59, 1782–1793. [Google Scholar] [CrossRef] [PubMed]
- Ahmadi, S.; Razazan, A.; Nagpal, R.; Jain, S.; Wang, B.; Mishra, S.P.; Wang, S.; Justice, J.; Ding, J.; McClain, D.A.; et al. Metformin Reduces Aging-Related Leaky Gut and Improves Cognitive Function by Beneficially Modulating Gut Microbiome/Goblet Cell/Mucin Axis. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, e9–e21. [Google Scholar] [CrossRef] [PubMed]
- Bonaz, B.; Sinniger, V.; Pellissier, S. The Vagus Nerve in the Neuro-Immune Axis: Implications in the Pathology of the Gastrointestinal Tract. Front. Immunol. 2017, 8, 1452. [Google Scholar] [CrossRef] [PubMed]
- Vasquez, E.C.; Aires, R.; Ton, A.M.M.; Amorim, F.G. New Insights on the Beneficial Effects of the Probiotic Kefir on Vascular Dysfunction in Cardiovascular and Neurodegenerative Diseases. Curr. Pharm. Des. 2020, 26, 3700–3710. [Google Scholar] [CrossRef]
- Yue, Q.; Cai, M.; Xiao, B.; Zhan, Q.; Zeng, C. The Microbiota-Gut-Brain Axis and Epilepsy. Cell. Mol. Neurobiol. 2021. [Google Scholar] [CrossRef] [PubMed]
- Howland, R.H. Vagus Nerve Stimulation. Curr. Behav. Neurosci. Rep. 2014, 1, 64–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vasquez, E.C.; Lewis, S.J.; Varner, K.J.; Brody, M.J. Chronic lesion of rostral ventrolateral medulla in spontaneously hypertensive rats. Hypertension 1992, 19, Ii154–Ii158. [Google Scholar] [CrossRef] [Green Version]
- Fülling, C.; Dinan, T.G.; Cryan, J.F. Gut Microbe to Brain Signaling: What Happens in Vagus. Neuron 2019, 101, 998–1002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schenberg, L.C.; Brandão, C.A.; Vasquez, E.C. Role of periaqueductal gray matter in hypertension in spontaneously hypertensive rats. Hypertension 1995, 26, 1125–1128. [Google Scholar] [CrossRef]
- Sampaio, K.N.; Mauad, H.; Vasquez, E.C.; Schenberg, L.C. Role of pulmonary stretch receptors and sympathetic system in the inhibition of reflex bradycardia produced by chemical stimulation of the periaqueductal gray matter of the rat. Neuroscience 2012, 210, 222–233. [Google Scholar] [CrossRef] [PubMed]
- Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr. Med. 2018, 17, 28–32. [Google Scholar]
- Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef] [PubMed]
- Crumeyrolle-Arias, M.; Jaglin, M.; Bruneau, A.; Vancassel, S.; Cardona, A.; Daugé, V.; Naudon, L.; Rabot, S. Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 2014, 42, 207–217. [Google Scholar] [CrossRef]
- Schmidt, K.; Cowen, P.J.; Harmer, C.J.; Tzortzis, G.; Errington, S.; Burnet, P.W. Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology 2015, 232, 1793–1801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, N.; Zhang, Y.; Li, M.; Wang, W.; Liu, Z.; Xi, C.; Huang, X.; Liu, J.; Huang, J.; Tian, D.; et al. Efficacy of probiotics on stress in healthy volunteers: A systematic review and meta-analysis based on randomized controlled trials. Brain Behav. 2020, 10, e01699. [Google Scholar] [CrossRef]
- Tsavkelova, E.A.; Botvinko, I.V.; Kudrin, V.S.; Oleskin, A.V. Detection of neurotransmitter amines in microorganisms with the use of high-performance liquid chromatography. Dokl. Biochem. 2000, 372, 115–117. [Google Scholar] [PubMed]
- Strandwitz, P.; Kim, K.H.; Terekhova, D.; Liu, J.K.; Sharma, A.; Levering, J.; McDonald, D.; Dietrich, D.; Ramadhar, T.R.; Lekbua, A.; et al. GABA-modulating bacteria of the human gut microbiota. Nat. Microbiol. 2019, 4, 396–403. [Google Scholar] [CrossRef] [PubMed]
- Muller, P.A.; Schneeberger, M.; Matheis, F.; Wang, P.; Kerner, Z.; Ilanges, A.; Pellegrino, K.; Del Mármol, J.; Castro, T.B.R.; Furuichi, M.; et al. Microbiota modulate sympathetic neurons via a gut-brain circuit. Nature 2020, 583, 441–446. [Google Scholar] [CrossRef]
- Karbach, S.H.; Schönfelder, T.; Brandão, I.; Wilms, E.; Hörmann, N.; Jäckel, S.; Schüler, R.; Finger, S.; Knorr, M.; Lagrange, J.; et al. Gut Microbiota Promote Angiotensin II-Induced Arterial Hypertension and Vascular Dysfunction. J. Am. Heart Assoc. 2016, 5, e003698. [Google Scholar] [CrossRef] [Green Version]
- Lu, C.C.; Ma, K.L.; Ruan, X.Z.; Liu, B.C. Intestinal dysbiosis activates renal renin-angiotensin system contributing to incipient diabetic nephropathy. Int. J. Med. Sci. 2018, 15, 816–822. [Google Scholar] [CrossRef] [Green Version]
- Lu, C.C.; Hu, Z.B.; Wang, R.; Hong, Z.H.; Lu, J.; Chen, P.P.; Zhang, J.X.; Li, X.Q.; Yuan, B.Y.; Huang, S.J.; et al. Gut microbiota dysbiosis-induced activation of the intrarenal renin-angiotensin system is involved in kidney injuries in rat diabetic nephropathy. Acta Pharmacol. Sin. 2020, 41, 1111–1118. [Google Scholar] [CrossRef] [PubMed]
- Madison, A.; Kiecolt-Glaser, J.K. Stress, depression, diet, and the gut microbiota: Human-bacteria interactions at the core of psychoneuroimmunology and nutrition. Curr. Opin. Behav. Sci. 2019, 28, 105–110. [Google Scholar] [CrossRef] [PubMed]
- Monteiro, B.L.; Dias, A.T.; Wanderkoke, S.C.; Yokota, R.; Casarini, D.E.; Leal, M.A.S.; Nogueira, B.V.; Meyrelles, S.S.; Campos-Toimil, M.; Campagnaro, B.P.; et al. Protective effects of kefir in the angiotensin II-dependent hypertension. J. Funct. Foods 2020, 75, 104260. [Google Scholar] [CrossRef]
- Klippel, B.F.; Duemke, L.B.; Leal, M.A.; Friques, A.G.; Dantas, E.M.; Dalvi, R.F.; Gava, A.L.; Pereira, T.M.; Andrade, T.U.; Meyrelles, S.S.; et al. Effects of Kefir on the Cardiac Autonomic Tones and Baroreflex Sensitivity in Spontaneously Hypertensive Rats. Front. Physiol. 2016, 7, 211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brasil, G.A.; Silva-Cutini, M.A.; Moraes, F.S.A.; Pereira, T.M.C.; Vasquez, E.C.; Lenz, D.; Bissoli, N.S.; Endringer, D.C.; de Lima, E.M.; Biancardi, V.C.; et al. The benefits of soluble non-bacterial fraction of kefir on blood pressure and cardiac hypertrophy in hypertensive rats are mediated by an increase in baroreflex sensitivity and decrease in angiotensin-converting enzyme activity. Nutrition 2018, 51–52, 66–72. [Google Scholar] [CrossRef] [PubMed]
- Vasquez, E.C.; Johnson, R.F.; Beltz, T.G.; Haskell, R.E.; Davidson, B.L.; Johnson, A.K. Replication-deficient adenovirus vector transfer of gfp reporter gene into supraoptic nucleus and subfornical organ neurons. Exp. Neurol. 1998, 154, 353–365. [Google Scholar] [CrossRef] [PubMed]
- Basso, N.; Terragno, N.A. History about the discovery of the renin-angiotensin system. Hypertension 2001, 38, 1246–1249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pereira, T.M.; Balarini, C.M.; Silva, I.V.; Cabral, A.M.; Vasquez, E.C.; Meyrelles, S.S. Endogenous angiotensin II modulates nNOS expression in renovascular hypertension. Braz. J. Med. Biol. Res. 2009, 42, 685–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nogueira, B.V.; Palomino, Z.; Porto, M.L.; Balarini, C.M.; Pereira, T.M.; Baldo, M.P.; Casarini, D.E.; Meyrelles, S.S.; Vasquez, E.C. Granulocyte colony stimulating factor prevents kidney infarction and attenuates renovascular hypertension. Cell. Physiol. Biochem. 2012, 29, 143–152. [Google Scholar] [CrossRef] [Green Version]
- Pereira, T.M.; Pimenta, F.S.; Porto, M.L.; Baldo, M.P.; Campagnaro, B.P.; Gava, A.L.; Meyrelles, S.S.; Vasquez, E.C. Coadjuvants in the Diabetic Complications: Nutraceuticals and Drugs with Pleiotropic Effects. Int. J. Mol. Sci. 2016, 17, 1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toral, M.; Robles-Vera, I.; de la Visitación, N.; Romero, M.; Yang, T.; Sánchez, M.; Gómez-Guzmán, M.; Jiménez, R.; Raizada, M.K.; Duarte, J. Critical Role of the Interaction Gut Microbiota—Sympathetic Nervous System in the Regulation of Blood Pressure. Front. Physiol. 2019, 10, 231. [Google Scholar] [CrossRef] [Green Version]
- Dias, A.T.; Cintra, A.S.; Frossard, J.C.; Palomino, Z.; Casarini, D.E.; Gomes, I.B.; Balarini, C.M.; Gava, A.L.; Campagnaro, B.P.; Pereira, T.M.; et al. Inhibition of phosphodiesterase 5 restores endothelial function in renovascular hypertension. J. Transl. Med. 2014, 12, 250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campagnaro, B.P.; Gava, A.L.; Meyrelles, S.S.; Vasquez, E.C. Cardiac-autonomic imbalance and baroreflex dysfunction in the renovascular Angiotensin-dependent hypertensive mouse. Int. J. Hypertens. 2012, 2012, 968123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campagnaro, B.P.; Tonini, C.L.; Nogueira, B.V.; Casarini, D.E.; Vasquez, E.C.; Meyrelles, S.S. DNA damage and augmented oxidative stress in bone marrow mononuclear cells from Angiotensin-dependent hypertensive mice. Int. J. Hypertens. 2013, 2013, 305202. [Google Scholar] [CrossRef] [PubMed]
- Nishi, E.E.; Martins, B.S.; Milanez, M.I.; Lopes, N.R.; de Melo, J.F., Jr.; Pontes, R.B.; Girardi, A.C.; Campos, R.R.; Bergamaschi, C.T. Stimulation of renal afferent fibers leads to activation of catecholaminergic and non-catecholaminergic neurons in the medulla oblongata. Auton. Neurosci. 2017, 204, 48–56. [Google Scholar] [CrossRef] [PubMed]
- Carmichael, C.Y.; Kuwabara, J.T.; Pascale, C.L.; Moreira, J.D.; Mahne, S.E.; Kapusta, D.R.; Rosene, D.L.; Williams, J.S.; Cunningham, J.T.; Wainford, R.D. Hypothalamic Paraventricular Nucleus Gαi(2) (Guanine Nucleotide-Binding Protein Alpha Inhibiting Activity Polypeptide 2) Protein-Mediated Neural Control of the Kidney and the Salt Sensitivity of Blood Pressure. Hypertension 2020, 75, 1002–1011. [Google Scholar] [CrossRef] [PubMed]
- Nakamura-Palacios, E.M.; Caldas, C.K.; Fiorini, A.; Chagas, K.D.; Chagas, K.N.; Vasquez, E.C. Deficits of spatial learning and working memory in spontaneously hypertensive rats. Behav. Brain Res. 1996, 74, 217–227. [Google Scholar] [CrossRef]
- Cabral, A.M.; Vasquez, E.C. Time course of cardiac sympathetic and vagal tone changes in renovascular hypertensive rats. Am. J. Hypertens. 1991, 4, 815–819. [Google Scholar] [CrossRef]
- Sharma, R.K.; Yang, T.; Oliveira, A.C.; Lobaton, G.O.; Aquino, V.; Kim, S.; Richards, E.M.; Pepine, C.J.; Sumners, C.; Raizada, M.K. Microglial Cells Impact Gut Microbiota and Gut Pathology in Angiotensin II-Induced Hypertension. Circ. Res. 2019, 124, 727–736. [Google Scholar] [CrossRef]
- Santanna, A.F.; Filete, P.F.; Lima, E.M.; Porto, M.L.; Meyrelles, S.S.; Vasquez, E.C.; Endringer, D.C.; Lenz, D.; Abdalla, D.S.P.; Pereira, T.M.C.; et al. Chronic administration of the soluble, nonbacterial fraction of kefir attenuates lipid deposition in LDLr(-/-) mice. Nutrition 2017, 35, 100–105. [Google Scholar] [CrossRef]
- Moyses, M.R.; Cabral, A.M.; Marçal, D.; Vasquez, E.C. Sigmoidal curve-fitting of baroreceptor sensitivity in renovascular 2K1C hypertensive rats. Braz. J. Med. Biol. Res. 1994, 27, 1419–1424. [Google Scholar]
- Cunha, R.S.; Cabral, A.M.; Vasquez, E.C. Evidence that the autonomic nervous system plays a major role in the L-NAME-induced hypertension in conscious rats. Am. J. Hypertens. 1993, 6, 806–809. [Google Scholar] [CrossRef]
- Mell, B.; Jala, V.R.; Mathew, A.V.; Byun, J.; Waghulde, H.; Zhang, Y.; Haribabu, B.; Vijay-Kumar, M.; Pennathur, S.; Joe, B. Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol. Genom. 2015, 47, 187–197. [Google Scholar] [CrossRef] [Green Version]
- Yang, T.; Santisteban, M.M.; Rodriguez, V.; Li, E.; Ahmari, N.; Carvajal, J.M.; Zadeh, M.; Gong, M.; Qi, Y.; Zubcevic, J.; et al. Gut dysbiosis is linked to hypertension. Hypertension 2015, 65, 1331–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miners, J.S.; Schulz, I.; Love, S. Differing associations between Aβ accumulation, hypoperfusion, blood-brain barrier dysfunction and loss of PDGFRB pericyte marker in the precuneus and parietal white matter in Alzheimer’s disease. J. Cereb. Blood Flow Metab. 2018, 38, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Šiarnik, P.; Klobučníková, K.; Mucska, I.; Hlucháňová, A.; Hanus, O.; Turčáni, P.; Kollár, B. Obstructive sleep apnea and hypertension: The role of gut microbiome. Vnitr. Lek. 2020, 66, 415–419. [Google Scholar] [CrossRef]
- Dumitrescu, L.; Popescu-Olaru, I.; Cozma, L.; Tulbă, D.; Hinescu, M.E.; Ceafalan, L.C.; Gherghiceanu, M.; Popescu, B.O. Oxidative Stress and the Microbiota-Gut-Brain Axis. Oxid. Med. Cell. Longev. 2018, 2018, 2406594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sies, H.; Cadenas, E. Oxidative stress: Damage to intact cells and organs. Philos. Trans. R. Soc. Lond. B Biol. Sci. 1985, 311, 617–631. [Google Scholar] [CrossRef] [PubMed]
- Sies, H. Oxidative Stress: Concept and Some Practical Aspects. Antioxidants 2020, 9, 852. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.H.; Kim, K.; Ahn, J.H.; Chang, H.K. Increased expression of tumor necrosis factor-alpha in the rat hippocampus after acute homocysteine administration. J. Epilepsy Res. 2011, 1, 6–12. [Google Scholar] [CrossRef] [PubMed]
- Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salim, S. Oxidative Stress and the Central Nervous System. J. Pharmacol. Exp. Ther. 2017, 360, 201–205. [Google Scholar] [CrossRef] [PubMed]
- Arnal, M.E.; Lallès, J.P. Gut epithelial inducible heat-shock proteins and their modulation by diet and the microbiota. Nutr. Rev. 2016, 74, 181–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Obrenovich, M.E.M. Leaky Gut, Leaky Brain? Microorganisms 2018, 6, 107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luca, M.; Di Mauro, M.; Luca, A. Gut Microbiota in Alzheimer’s Disease, Depression, and Type 2 Diabetes Mellitus: The Role of Oxidative Stress. Oxid. Med. Cell. Longev. 2019, 2019, 4730539. [Google Scholar] [CrossRef]
- Pollack, M.; Leeuwenburgh, C. Molecular mechanisms of oxidative stress in aging: Free radicals, aging, antioxidants and disease. Handb. Oxid. Antioxid. Exerc. 1999, 30, 881–926. [Google Scholar]
- Phaniendra, A.; Jestadi, D.B.; Periyasamy, L. Free radicals: Properties, sources, targets, and their implication in various diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [Green Version]
- Viña, D.; Seoane, N.; Vasquez, E.C.; Campos-Toimil, M. cAMP Compartmentalization in Cerebrovascular Endothelial Cells: New Therapeutic Opportunities in Alzheimer’s Disease. Cells 2021, 10, 1951. [Google Scholar] [CrossRef]
- Salman, M.M.; Kitchen, P.; Woodroofe, M.N.; Bill, R.M.; Conner, A.C.; Heath, P.R.; Conner, M.T. Transcriptome Analysis of Gene Expression Provides New Insights into the Effect of Mild Therapeutic Hypothermia on Primary Human Cortical Astrocytes Cultured under Hypoxia. Front. Cell. Neurosci. 2017, 11, 386. [Google Scholar] [CrossRef] [Green Version]
- Vasquez, E.C.; Pereira, T.M.C.; Campos-Toimil, M.; Baldo, M.P.; Peotta, V.A. Gut Microbiota, Diet, and Chronic Diseases: The Role Played by Oxidative Stress. Oxid. Med. Cell. Longev. 2019, 2019, 7092032. [Google Scholar] [CrossRef]
- Barboza, K.R.M.; Coco, L.Z.; Alves, G.M.; Peters, B.; Vasquez, E.C.; Pereira, T.M.C.; Meyrelles, S.S.; Campagnaro, B.P. Gastroprotective effect of oral kefir on indomethacin-induced acute gastric lesions in mice: Impact on oxidative stress. Life Sci. 2018, 209, 370–376. [Google Scholar] [CrossRef] [PubMed]
- de Souza Santos, V.; Peters, B.; Côco, L.Z.; Alves, G.M.; de Assis, A.; Nogueira, B.V.; Meyrelles, S.S.; Porto, M.L.; Vasquez, E.C.; Campagnaro, B.P.; et al. Silymarin protects against radiocontrast-induced nephropathy in mice. Life Sci. 2019, 228, 305–315. [Google Scholar] [CrossRef] [PubMed]
- Chisté, L.A.; Pereira, B.P.; Porto, M.L.; de Oliveira, J.P.; de Assis, A.; Nogueira, B.V.; Meyrelles, S.S.; de Andrade, T.U.; Campos-Toimil, M.; Vasquez, E.C.; et al. Worsening of Oxidative Stress, DNA Damage, and Atherosclerotic Lesions in Aged LDLr(-/-) Mice after Consumption of Guarana Soft Drinks. Oxid. Med. Cell. Longev. 2019, 2019, 9042526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ravcheev, D.A.; Thiele, I. Systematic genomic analysis reveals the complementary aerobic and anaerobic respiration capacities of the human gut microbiota. Front. Microbiol. 2014, 5, 674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, Y.; Chen, D.; Zheng, P.; Yu, J.; He, J.; Mao, X.; Yu, B. The Bidirectional Interactions between Resveratrol and Gut Microbiota: An Insight into Oxidative Stress and Inflammatory Bowel Disease Therapy. Biomed. Res. Int. 2019, 2019, 5403761. [Google Scholar] [CrossRef]
- Gustaw-Rothenberg, K.; Kowalczuk, K.; Stryjecka-Zimmer, M. Lipids’ peroxidation markers in Alzheimer’s disease and vascular dementia. Geriatr. Gerontol. Int. 2010, 10, 161–166. [Google Scholar] [CrossRef] [PubMed]
- Akbari, E.; Asemi, Z.; Daneshvar Kakhaki, R.; Bahmani, F.; Kouchaki, E.; Tamtaji, O.R.; Hamidi, G.A.; Salami, M. Effect of Probiotic Supplementation on Cognitive Function and Metabolic Status in Alzheimer’s Disease: A Randomized, Double-Blind and Controlled Trial. Front. Aging Neurosci. 2016, 8, 256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McGrath, L.T.; McGleenon, B.M.; Brennan, S.; McColl, D.; Mc, I.S.; Passmore, A.P. Increased oxidative stress in Alzheimer’s disease as assessed with 4-hydroxynonenal but not malondialdehyde. QJM 2001, 94, 485–490. [Google Scholar] [CrossRef]
- Leandro, G.S.; Lobo, R.R.; Oliveira, D.V.; Moriguti, J.C.; Sakamoto-Hojo, E.T. Lymphocytes of patients with Alzheimer’s disease display different DNA damage repair kinetics and expression profiles of DNA repair and stress response genes. Int. J. Mol. Sci. 2013, 14, 12380–12400. [Google Scholar] [CrossRef] [Green Version]
- Møller, P.; Stopper, H.; Collins, A.R. Measurement of DNA damage with the comet assay in high-prevalence diseases: Current status and future directions. Mutagenesis 2020, 35, 5–18. [Google Scholar] [CrossRef] [PubMed]
- Tonini, C.L.; Campagnaro, B.P.; Louro, L.P.; Pereira, T.M.; Vasquez, E.C.; Meyrelles, S.S. Effects of Aging and Hypercholesterolemia on Oxidative Stress and DNA Damage in Bone Marrow Mononuclear Cells in Apolipoprotein E-deficient Mice. Int. J. Mol. Sci. 2013, 14, 3325–3342. [Google Scholar] [CrossRef] [PubMed]
- Dikalov, S.I.; Harrison, D.G. Methods for detection of mitochondrial and cellular reactive oxygen species. Antioxid. Redox Signal. 2014, 20, 372–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, A.; Chen, S.H.; Kadiiska, M.B.; Hong, J.S.; Zielonka, J.; Kalyanaraman, B.; Mason, R.P. Inducible nitric oxide synthase is key to peroxynitrite-mediated, LPS-induced protein radical formation in murine microglial BV2 cells. Free Radic. Biol. Med. 2014, 73, 51–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, B.; Deng, X.; Lim, G.G.Y.; Xie, S.; Zhou, Z.D.; Lim, K.L.; Tan, E.K. Superoxide drives progression of Parkin/PINK1-dependent mitophagy following translocation of Parkin to mitochondria. Cell Death Dis. 2017, 8, e3097. [Google Scholar] [CrossRef] [PubMed]
- Lemos, V.R.; Aires, R.; Côco, L.Z.; Domingues, R.B.; Meyrelles, S.S.; Vasquez, E.C.; Pereira, T.M.C.; Campagnaro, B.P. Benefits of multi-day supplementation with probiotic kefir in Rasmussen encephalitis: The first case report. Nutr. Neurosci. 2021, 1–8. [Google Scholar] [CrossRef]
- Bôa, I.S.; Porto, M.L.; Pereira, A.C.; Ramos, J.P.; Scherer, R.; Oliveira, J.P.; Nogueira, B.V.; Meyrelles, S.S.; Vasquez, E.C.; Endringer, D.C.; et al. Resin from Virola oleifera Protects Against Radiocontrast-Induced Nephropathy in Mice. PLoS ONE 2015, 10, e0144329. [Google Scholar] [CrossRef]
- Porto, M.L.; Lírio, L.M.; Dias, A.T.; Batista, A.T.; Campagnaro, B.P.; Mill, J.G.; Meyrelles, S.S.; Baldo, M.P. Increased oxidative stress and apoptosis in peripheral blood mononuclear cells of fructose-fed rats. Toxicol. Vitr. 2015, 29, 1977–1981. [Google Scholar] [CrossRef] [PubMed]
- Jacinto, T.A.; Meireles, G.S.; Dias, A.T.; Aires, R.; Porto, M.L.; Gava, A.L.; Vasquez, E.C.; Pereira, T.M.C.; Campagnaro, B.P.; Meyrelles, S.S. Increased ROS production and DNA damage in monocytes are biomarkers of aging and atherosclerosis. Biol. Res. 2018, 51, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Magistrelli, L.; Amoruso, A.; Mogna, L.; Graziano, T.; Cantello, R.; Pane, M.; Comi, C. Probiotics May Have Beneficial Effects in Parkinson’s Disease: In vitro Evidence. Front. Immunol. 2019, 10, 969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castelli, V.; d’Angelo, M.; Quintiliani, M.; Benedetti, E.; Cifone, M.G.; Cimini, A. The emerging role of probiotics in neurodegenerative diseases: New hope for Parkinson’s disease? Neural Regen. Res. 2021, 16, 628–634. [Google Scholar] [CrossRef]
- Cassani, E.; Privitera, G.; Pezzoli, G.; Pusani, C.; Madio, C.; Iorio, L.; Barichella, M. Use of probiotics for the treatment of constipation in Parkinson’s disease patients. Minerva Gastroenterol. Dietol. 2011, 57, 117–121. [Google Scholar] [PubMed]
- Liu, J.; Xu, F.; Nie, Z.; Shao, L. Gut Microbiota Approach-A New Strategy to Treat Parkinson’s Disease. Front. Cell. Infect. Microbiol. 2020, 10, 570658. [Google Scholar] [CrossRef] [PubMed]
- Leblhuber, F.; Steiner, K.; Schuetz, B.; Fuchs, D.; Gostner, J.M. Probiotic Supplementation in Patients with Alzheimer’s Dementia—An Explorative Intervention Study. Curr. Alzheimer Res. 2018, 15, 1106–1113. [Google Scholar] [CrossRef] [PubMed]
- Hsieh, T.H.; Kuo, C.W.; Hsieh, K.H.; Shieh, M.J.; Peng, C.W.; Chen, Y.C.; Chang, Y.L.; Huang, Y.Z.; Chen, C.C.; Chang, P.K.; et al. Probiotics Alleviate the Progressive Deterioration of Motor Functions in a Mouse Model of Parkinson’s Disease. Brain Sci. 2020, 10, 206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Agahi, A.; Hamidi, G.A.; Daneshvar, R.; Hamdieh, M.; Soheili, M.; Alinaghipour, A.; Esmaeili Taba, S.M.; Salami, M. Does Severity of Alzheimer’s Disease Contribute to Its Responsiveness to Modifying Gut Microbiota? A Double Blind Clinical Trial. Front. Neurol. 2018, 9, 662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Den, H.; Dong, X.; Chen, M.; Zou, Z. Efficacy of probiotics on cognition, and biomarkers of inflammation and oxidative stress in adults with Alzheimer’s disease or mild cognitive impairment—A meta-analysis of randomized controlled trials. Aging 2020, 12, 4010–4039. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Eguílaz, M.; Ramón-Trapero, J.L.; Pérez-Martínez, L.; Blanco, J.R. The beneficial effect of probiotics as a supplementary treatment in drug-resistant epilepsy: A pilot study. Benef. Microbes 2018, 9, 875–881. [Google Scholar] [CrossRef] [PubMed]
- Vezzani, A. Inflammation and epilepsy. Epilepsy Curr. 2005, 5, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Walker, L.; Sills, G.J. Inflammation and epilepsy: The foundations for a new therapeutic approach in epilepsy? Epilepsy Curr. 2012, 12, 8–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bagheri, S.; Heydari, A.; Alinaghipour, A.; Salami, M. Effect of probiotic supplementation on seizure activity and cognitive performance in PTZ-induced chemical kindling. Epilepsy Behav. 2019, 95, 43–50. [Google Scholar] [CrossRef] [PubMed]
- Kilinc, E.; Ankarali, S.; Ayhan, D.; Ankarali, H.; Torun, I.E.; Cetinkaya, A. Protective effects of long-term probiotic mixture supplementation against pentylenetetrazole-induced seizures, inflammation and oxidative stress in rats. J. Nutr. Biochem. 2021, 98, 108830. [Google Scholar] [CrossRef] [PubMed]
- O’Reilly, S.; Loncin, M.; Cooksey, B. Dopamine and basal ganglia disorders. Neurology 1965, 15, 980–984. [Google Scholar] [CrossRef] [PubMed]
- Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
- Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. NPJ Parkinsons Dis. 2021, 7, 27. [Google Scholar] [CrossRef]
- DeMaagd, G.; Philip, A. Parkinson’s Disease and Its Management: Part 1: Disease Entity, Risk Factors, Pathophysiology, Clinical Presentation, and Diagnosis. Pharm. Ther. 2015, 40, 504–532. [Google Scholar]
- Gazerani, P. Probiotics for Parkinson’s Disease. Int. J. Mol. Sci. 2019, 20, 4121. [Google Scholar] [CrossRef] [Green Version]
- Shen, T.; Yue, Y.; He, T.; Huang, C.; Qu, B.; Lv, W.; Lai, H.Y. The Association between the Gut Microbiota and Parkinson’s Disease, a Meta-Analysis. Front. Aging Neurosci. 2021, 13, 636545. [Google Scholar] [CrossRef]
- Georgescu, D.; Ancusa, O.E.; Georgescu, L.A.; Ionita, I.; Reisz, D. Nonmotor gastrointestinal disorders in older patients with Parkinson’s disease: Is there hope? Clin. Interv. Aging 2016, 11, 1601–1608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tamtaji, O.R.; Taghizadeh, M.; Daneshvar Kakhaki, R.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z. Clinical and metabolic response to probiotic administration in people with Parkinson’s disease: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2019, 38, 1031–1035. [Google Scholar] [CrossRef] [PubMed]
- van Kessel, S.P.; Frye, A.K.; El-Gendy, A.O.; Castejon, M.; Keshavarzian, A.; van Dijk, G.; El Aidy, S. Gut bacterial tyrosine decarboxylases restrict levels of levodopa in the treatment of Parkinson’s disease. Nat. Commun. 2019, 10, 310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perez-Pardo, P.; Kliest, T.; Dodiya, H.B.; Broersen, L.M.; Garssen, J.; Keshavarzian, A.; Kraneveld, A.D. The gut-brain axis in Parkinson’s disease: Possibilities for food-based therapies. Eur. J. Pharmacol. 2017, 817, 86–95. [Google Scholar] [CrossRef] [PubMed]
- Bass, D.; Stentiford, G.D.; Wang, H.C.; Koskella, B.; Tyler, C.R. The Pathobiome in Animal and Plant Diseases. Trends Ecol. Evol. 2019, 34, 996–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marsh, A.J.; O’Sullivan, O.; Hill, C.; Ross, R.P.; Cotter, P.D. Sequencing-based analysis of the bacterial and fungal composition of kefir grains and milks from multiple sources. PLoS ONE 2013, 8, e69371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Etienne-Mesmin, L.; Livrelli, V.; Privat, M.; Denis, S.; Cardot, J.M.; Alric, M.; Blanquet-Diot, S. Effect of a new probiotic Saccharomyces cerevisiae strain on survival of Escherichia coli O157:H7 in a dynamic gastrointestinal model. Appl. Environ. Microbiol. 2011, 77, 1127–1131. [Google Scholar] [CrossRef] [Green Version]
- Barnes, L.L.; Capuano, A.W.; Aiello, A.E.; Turner, A.D.; Yolken, R.H.; Torrey, E.F.; Bennett, D.A. Cytomegalovirus infection and risk of Alzheimer disease in older black and white individuals. J. Infect. Dis. 2015, 211, 230–237. [Google Scholar] [CrossRef] [Green Version]
- Itzhaki, R.F. Herpes simplex virus type 1 and Alzheimer’s disease: Possible mechanisms and signposts. FASEB J. 2017, 31, 3216–3226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Angelucci, F.; Cechova, K.; Amlerova, J.; Hort, J. Antibiotics, gut microbiota, and Alzheimer’s disease. J. Neuroinflamm. 2019, 16, 108. [Google Scholar] [CrossRef] [PubMed]
- Gallucci, A.; Patel, D.C.; Thai, K.; Trinh, J.; Gude, R.; Shukla, D.; Campbell, S.L. Gut metabolite S-equol ameliorates hyperexcitability in entorhinal cortex neurons following Theiler murine encephalomyelitis virus-induced acute seizures. Epilepsia 2021, 62, 1829–1841. [Google Scholar] [CrossRef]
- Pimenta, F.S.; Luaces-Regueira, M.; Ton, A.M.; Campagnaro, B.P.; Campos-Toimil, M.; Pereira, T.M.; Vasquez, E.C. Mechanisms of Action of Kefir in Chronic Cardiovascular and Metabolic Diseases. Cell. Physiol. Biochem. 2018, 48, 1901–1914. [Google Scholar] [CrossRef] [PubMed]
- Irigoyen, A.; Arana, I.; Castiella, M.; Torre, P.; Ibanez, F.C. Microbiological, physicochemical, and sensory characteristics of kefir during storage. Food Chem. 2005, 90, 613–620. [Google Scholar] [CrossRef]
- Alves, E.; Ntungwe, E.N.; Gregório, J.; Rodrigues, L.M.; Pereira-Leite, C.; Caleja, C.; Pereira, E.; Barros, L.; Aguilar-Vilas, M.V.; Rosado, C.; et al. Characterization of Kefir Produced in Household Conditions: Physicochemical and Nutritional Profile, and Storage Stability. Foods 2021, 10, 1057. [Google Scholar] [CrossRef]
- Wouters, J.T.M.; Ayad, E.H.E.; Hugenholtz, J.; Smit, G. Microbes from raw milk for fermented dairy products. Int. Dairy J. 2002, 12, 91–109. [Google Scholar] [CrossRef]
- Bourrie, B.C.; Willing, B.P.; Cotter, P.D. The Microbiota and Health Promoting Characteristics of the Fermented Beverage Kefir. Front. Microbiol. 2016, 7, 647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kemp, N. Kefir, the champagne of cultured dairy products. Cult. Dairy Prod. J. 1984, 19, 29–30. [Google Scholar]
- Fiorda, F.A.; de Melo Pereira, G.V.; Thomaz-Soccol, V.; Rakshit, S.K.; Pagnoncelli, M.G.B.; Vandenberghe, L.P.S.; Soccol, C.R. Microbiological, biochemical, and functional aspects of sugary kefir fermentation—A review. Food Microbiol. 2017, 66, 86–95. [Google Scholar] [CrossRef] [PubMed]
- Abraham, A.G.; De Antoni, G.L. Characterization of kefir grains grown in cows’ milk and in soya milk. J. Dairy Res. 1999, 66, 327–333. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, Z.; Wang, Y.; Ahmad, A.; Khan, S.T.; Nisa, M.; Ahmad, H.; Afreen, A. Kefir and health: A contemporary perspective. Crit. Rev. Food Sci. Nutr. 2013, 53, 422–434. [Google Scholar] [CrossRef] [PubMed]
- Vinderola, C.G.; Duarte, J.; Thangavel, D.; Perdigón, G.; Farnworth, E.; Matar, C. Immunomodulating capacity of kefir. J. Dairy Res. 2005, 72, 195–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodrigues, K.L.; Caputo, L.R.; Carvalho, J.C.; Evangelista, J.; Schneedorf, J.M. Antimicrobial and healing activity of kefir and kefiran extract. Int. J. Antimicrob. Agents 2005, 25, 404–408. [Google Scholar] [CrossRef] [PubMed]
- Marquina, D.; Santos, A.; Corpas, I.; Muñoz, J.; Zazo, J.; Peinado, J.M. Dietary influence of kefir on microbial activities in the mouse bowel. Lett. Appl. Microbiol. 2002, 35, 136–140. [Google Scholar] [CrossRef] [PubMed]
- Sharifi, M.; Moridnia, A.; Mortazavi, D.; Salehi, M.; Bagheri, M.; Sheikhi, A. Kefir: A powerful probiotics with anticancer properties. Med. Oncol. 2017, 34, 183. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.L.; Hung, K.F.; Yen, C.C.; Laio, C.H.; Wang, J.L.; Lan, Y.W.; Chong, K.Y.; Fan, H.C.; Chen, C.M. Kefir peptides alleviate particulate matter <4 μm (PM(4.0))-induced pulmonary inflammation by inhibiting the NF-κB pathway using luciferase transgenic mice. Sci. Rep. 2019, 9, 11529. [Google Scholar] [CrossRef] [PubMed]
- Garofalo, C.; Ferrocino, I.; Reale, A.; Sabbatini, R.; Milanović, V.; Alkić-Subašić, M.; Boscaino, F.; Aquilanti, L.; Pasquini, M.; Trombetta, M.F.; et al. Study of kefir drinks produced by backslopping method using kefir grains from Bosnia and Herzegovina: Microbial dynamics and volatilome profile. Food. Res. Int. 2020, 137, 109369. [Google Scholar] [CrossRef]
- Prado, M.R.; Blandón, L.M.; Vandenberghe, L.P.; Rodrigues, C.; Castro, G.R.; Thomaz-Soccol, V.; Soccol, C.R. Milk kefir: Composition, microbial cultures, biological activities, and related products. Front. Microbiol. 2015, 6, 1177. [Google Scholar] [CrossRef] [Green Version]
- Magalhães, K.T.; de Melo Pereira, G.V.; Campos, C.R.; Dragone, G.; Schwan, R.F. Brazilian kefir: Structure, microbial communities and chemical composition. Braz. J. Microbiol. 2011, 42, 693–702. [Google Scholar] [CrossRef] [Green Version]
- Wiszolek, M.; Tamime, A.Y.; Muir, D.D.; Barcley, M.N.I. Properties of kefir made in Scothland and Poland using bovine, caprine and ovine milk with different starter culture. LWT-Food Sci. Technol. 2001, 34, 251–261. [Google Scholar] [CrossRef]
- Liutkevičius, A.; Šarkinas, A. Studies on the growth conditions and composition of kefir grains—As a food and forage biomass. Vet. Zootec. 2004, 25, 64–70. [Google Scholar]
- Wang, H.; Wang, C.; Wang, M.; Guo, M. Chemical, Physiochemical, and Microstructural Properties, and Probiotic Survivability of Fermented Goat Milk Using Polymerized Whey Protein and Starter Culture Kefir Mild 01. J. Food Sci. 2017, 82, 2650–2658. [Google Scholar] [CrossRef]
- de Lima, M.; da Silva, R.A.; da Silva, M.F.; da Silva, P.A.B.; Costa, R.; Teixeira, J.A.C.; Porto, A.L.F.; Cavalcanti, M.T.H. Brazilian Kefir-Fermented Sheep’s Milk, a Source of Antimicrobial and Antioxidant Peptides. Probiotics Antimicrob. Proteins 2018, 10, 446–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cais-Sokolińska, D.; Wójtowski, J.; Pikul, J.; Danków, R.; Majcher, M.; Teichert, J.; Bagnicka, E. Formation of volatile compounds in kefir made of goat and sheep milk with high polyunsaturated fatty acid content. J. Dairy Sci. 2015, 98, 6692–6705. [Google Scholar] [CrossRef] [Green Version]
- Micheli, L.; Uccelletti, D.; Palleschi, C.; Crescenzi, V. Isolation and characterisation of a ropy Lactobacillus strain producing the exopolysaccharide kefiran. Appl. Microbiol. Biotechnol. 1999, 53, 69–74. [Google Scholar] [CrossRef] [PubMed]
- Maeda, H.; Zhu, X.; Suzuki, S.; Suzuki, K.; Kitamura, S. Structural characterization and biological activities of an exopolysaccharide kefiran produced by Lactobacillus kefiranofaciens WT-2B(T). J. Agric. Food Chem. 2004, 52, 5533–5538. [Google Scholar] [CrossRef] [PubMed]
- Duarte, J.; Vinderola, G.; Ritz, B.; Perdigón, G.; Matar, C. Immunomodulating capacity of commercial fish protein hydrolysate for diet supplementation. Immunobiology 2006, 211, 341–350. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.-R.; Wang, S.-Y.; Lin, Y.-Y.; Lin, C.-W. Antitumor activity of milk kefir and soy milk kefir in tumor-bearing mice. Nutr. Cancer 2002, 44, 183–187. [Google Scholar] [CrossRef]
- Sadewa, A.H. Kefir properties prepared with goat milk and black rice (Oryza sativa L.) extract and its influence on the improvement of pancreatic β-cells in diabetic rats. Emir. J. Food Agric. 2015, 727–735. [Google Scholar] [CrossRef]
- Yilmaz-Ersan, L.; Ozcan, T.; Akpinar-Bayizit, A.; Sahin, S. Comparison of antioxidant capacity of cow and ewe milk kefirs. J. Dairy Sci. 2018, 101, 3788–3798. [Google Scholar] [CrossRef] [PubMed]
- Alimi, D.; Rekik, M.; Akkari, H. Comparative in vitro efficacy of kefir produced from camel, goat, ewe and cow milk on Haemonchus contortus. J. Helminthol. 2019, 93, 440–446. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.L.; Tsai, T.C.; Tsai, Y.C.; Liao, J.W.; Yen, C.C.; Chen, C.M. Kefir peptides prevent high-fructose corn syrup-induced non-alcoholic fatty liver disease in a murine model by modulation of inflammation and the JAK2 signaling pathway. Nutr. Diabetes 2016, 6, e237. [Google Scholar] [CrossRef] [PubMed]
- Tung, Y.T.; Chen, H.L.; Wu, H.S.; Ho, M.H.; Chong, K.Y.; Chen, C.M. Kefir peptides prevent hyperlipidemia and obesity in high-fat-diet-induced obese rats via lipid metabolism modulation. Mol. Nutr. Food Res. 2018, 62, 1700505. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.-L.; Lan, Y.-W.; Tu, M.-Y.; Tung, Y.-T.; Chan, M.N.-Y.; Wu, H.-S.; Yen, C.-C.; Chen, C.-M. Kefir peptides exhibit antidepressant-like activity in mice through the BDNF/TrkB pathway. J. Dairy Sci. 2021, 104, 6415–6430. [Google Scholar] [CrossRef]
- Tung, M.C.; Lan, Y.W.; Li, H.H.; Chen, H.L.; Chen, S.Y.; Chen, Y.H.; Lin, C.C.; Tu, M.Y.; Chen, C.M. Kefir peptides alleviate high-fat diet-induced atherosclerosis by attenuating macrophage accumulation and oxidative stress in ApoE knockout mice. Sci. Rep. 2020, 10, 8802. [Google Scholar] [CrossRef] [PubMed]
- Tu, M.-Y.; Han, K.-Y.; Chang, G.R.-L.; Lai, G.-D.; Chang, K.-Y.; Chen, C.-F.; Lai, J.-C.; Lai, C.-Y.; Chen, H.-L.; Chen, C.-M. Kefir peptides prevent estrogen deficiency-induced bone loss and modulate the structure of the gut microbiota in ovariectomized mice. Nutrients 2020, 12, 3432. [Google Scholar] [CrossRef]
- Quirós, A.; Hernández-Ledesma, B.; Ramos, M.; Amigo, L.; Recio, I. Angiotensin-converting enzyme inhibitory activity of peptides derived from caprine kefir. J. Dairy Sci. 2005, 88, 3480–3487. [Google Scholar] [CrossRef] [Green Version]
- Izquierdo-González, J.J.; Amil-Ruiz, F.; Zazzu, S.; Sánchez-Lucas, R.; Fuentes-Almagro, C.A.; Rodríguez-Ortega, M.J. Proteomic analysis of goat milk kefir: Profiling the fermentation-time dependent protein digestion and identification of potential peptides with biological activity. Food Chem. 2019, 295, 456–465. [Google Scholar] [CrossRef]
- Ebner, J.; Arslan, A.A.; Fedorova, M.; Hoffmann, R.; Küçükçetin, A.; Pischetsrieder, M. Peptide profiling of bovine kefir reveals 236 unique peptides released from caseins during its production by starter culture or kefir grains. J. Proteom. 2015, 117, 41–57. [Google Scholar] [CrossRef] [PubMed]
- Otles, S.; Cagindi, O. Kefir: A probiotic dairy-composition, nutritional and therapeutic aspects. Pak. J. Nutr. 2003, 2, 54–59. [Google Scholar]
- Biadała, A.; Adzahan, N.M. Storage Stability of Antioxidant in Milk Products Fermented with Selected Kefir Grain Microflora. Molecules 2021, 26, 3307. [Google Scholar] [CrossRef] [PubMed]
- Satir, G.; Guzel-Seydim, Z.B. Influence of Kefir fermentation on the bioactive substances of different breed goat milks. LWT-Food Sci. Technol. 2015, 63, 852–858. [Google Scholar] [CrossRef]
- Vieira, C.P.; Rosario, A.I.L.S.; Lelis, C.A.; Rekowsky, B.S.S.; Carvalho, A.P.A.; Rosário, D.K.A.; Elias, T.A.; Costa, M.P.; Foguel, D.; Conte-Junior, C.A. Bioactive Compounds from Kefir and Their Potential Benefits on Health: A Systematic Review and Meta-Analysis. Oxid. Med. Cell. Longev. 2021, 2021, 9081738. [Google Scholar] [CrossRef]
- Setyawardani, T.; Rahardjo, A.H.D.; Sulistyowati, M.; Wasito, S. Physiochemical and organoleptic features of goat milk kefir made of different kefir grain concentration on controlled fermentation. Anim. Prod. 2014, 16, 48–54. [Google Scholar]
- Seydim, Z.G.; Wyffels, J.T.; Seydim, A.C.; Greene, A.K. Turkish kefir and kefir grains: Microbial enumeration and electron microscopic observation. Int. J. Dairy Technol. 2005, 58, 25–29. [Google Scholar] [CrossRef]
- Simova, E.; Beshkova, D.; Angelov, A.; Hristozova, T.; Frengova, G.; Spasov, Z. Lactic acid bacteria and yeasts in kefir grains and kefir made from them. J. Ind. Microbiol. Biotechnol. 2002, 28, 1–6. [Google Scholar] [CrossRef]
- Farag, M.A.; Jomaa, S.A.; El-Wahed, A.A.; El-Seedi, A.H.R. The Many Faces of Kefir Fermented Dairy Products: Quality Characteristics, Flavour Chemistry, Nutritional Value, Health Benefits, and Safety. Nutrients 2020, 12, 346. [Google Scholar] [CrossRef] [Green Version]
- Sarkar, S. Potential of kefir as a dietetic beverage—A review. Br. Food J. 2007, 109, 280–290. [Google Scholar] [CrossRef]
- Garrote, G.L.; Abraham, A.G.; De Antoni, G.L. Chemical and microbiological characterisation of kefir grains. J. Dairy Res. 2001, 68, 639–652. [Google Scholar] [CrossRef] [PubMed]
- Ferreira, I.M.; Pinho, O.; Monteiro, D.; Faria, S.; Cruz, S.; Perreira, A.; Roque, A.C.; Tavares, P. Short communication: Effect of kefir grains on proteolysis of major milk proteins. J. Dairy Sci. 2010, 93, 27–31. [Google Scholar] [CrossRef] [Green Version]
- Ertekin, B.; Guzel-Seydim, Z.B. Effect of fat replacers on kefir quality. J. Sci. Food Agric. 2010, 90, 543–548. [Google Scholar] [CrossRef] [PubMed]
- Kazazi, H.; Khodaiyan, F.; Rezaei, K.; Pishvaei, M.; Mohammadifar, M.A.; Moieni, S. Rheology and microstructure of kefiran and whey protein mixed gels. J. Food Sci. Technol. 2017, 54, 1168–1174. [Google Scholar] [CrossRef] [Green Version]
- Salari, A.; Hashemi, M.; Afshari, A. Functional Properties of Kefiran in Medical field and food industry. Curr. Pharm. Biotechnol. 2021. [Google Scholar] [CrossRef]
- Rosa, D.D.; Dias, M.M.S.; Grześkowiak, Ł.M.; Reis, S.A.; Conceição, L.L.; Peluzio, M. Milk kefir: Nutritional, microbiological and health benefits. Nutr. Res. Rev. 2017, 30, 82–96. [Google Scholar] [CrossRef] [PubMed]
- Peluzio, M.; Dias, M.M.E.; Martinez, J.A.; Milagro, F.I. Kefir and Intestinal Microbiota Modulation: Implications in Human Health. Front. Nutr. 2021, 8, 638740. [Google Scholar] [CrossRef]
- Chen, Z.; Shi, J.; Yang, X.; Nan, B.; Liu, Y.; Wang, Z. Chemical and physical characteristics and antioxidant activities of the exopolysaccharide produced by Tibetan kefir grains during milk fermentation. Int. Dairy J. 2015, 43, 15–21. [Google Scholar] [CrossRef]
- Carasi, P.; Racedo, S.M.; Jacquot, C.; Romanin, D.E.; Serradell, M.A.; Urdaci, M.C. Impact of kefir derived Lactobacillus kefiri on the mucosal immune response and gut microbiota. J. Immunol. Res. 2015, 2015, 361604. [Google Scholar] [CrossRef] [Green Version]
- Carullo, G.; Governa, P.; Spizzirri, U.G.; Biagi, M.; Sciubba, F.; Giorgi, G.; Loizzo, M.R.; Di Cocco, M.E.; Aiello, F.; Restuccia, D. Sangiovese cv Pomace Seeds Extract-Fortified Kefir Exerts Anti-Inflammatory Activity in an In Vitro Model of Intestinal Epithelium Using Caco-2 Cells. Antioxidants 2020, 9, 54. [Google Scholar] [CrossRef] [Green Version]
- Radhouani, H.; Gonçalves, C.; Maia, F.R.; Oliveira, J.M.; Reis, R.L. Biological performance of a promising Kefiran-biopolymer with potential in regenerative medicine applications: A comparative study with hyaluronic acid. J. Mater. Sci. Mater. Med. 2018, 29, 124. [Google Scholar] [CrossRef]
- Fahmy, H.A.; Ismail, A.F.M. Gastroprotective effect of kefir on ulcer induced in irradiated rats. J. Photochem. Photobiol. B Biol. 2015, 144, 85–93. [Google Scholar] [CrossRef]
- Ghoneum, M.; Abdulmalek, S.; Pan, D. Reversal of age-associated oxidative stress in mice by PFT, a novel kefir product. Int. J. Immunopathol. Pharmacol. 2020, 34, 2058738420950149. [Google Scholar] [CrossRef]
- Ali, O.S.M.; Amin, N.E.; Abdel Fattah, S.M.; Abd El-Rahman, O. Ameliorative effect of kefir against γ-irradiation induced liver injury in male rats: Impact on oxidative stress and inflammation. Environ. Sci. Pollut. Res. Int. 2020, 27, 35161–35173. [Google Scholar] [CrossRef]
- Punaro, G.R.; Maciel, F.R.; Rodrigues, A.M.; Rogero, M.M.; Bogsan, C.S.; Oliveira, M.N.; Ihara, S.S.; Araujo, S.R.; Sanches, T.R.; Andrade, L.C.; et al. Kefir administration reduced progression of renal injury in STZ-diabetic rats by lowering oxidative stress. Nitric Oxide 2014, 37, 53–60. [Google Scholar] [CrossRef] [Green Version]
- Rosa, D.D.; Grześkowiak, Ł.M.; Ferreira, C.L.; Fonseca, A.C.; Reis, S.A.; Dias, M.M.; Siqueira, N.P.; Silva, L.L.; Neves, C.A.; Oliveira, L.L.; et al. Kefir reduces insulin resistance and inflammatory cytokine expression in an animal model of metabolic syndrome. Food Funct. 2016, 7, 3390–3401. [Google Scholar] [CrossRef]
- El Golli-Bennour, E.; Timoumi, R.; Annaibi, E.; Mokni, M.; Omezzine, A.; Bacha, H.; Abid-Essefi, S. Protective effects of kefir against deltamethrin-induced hepatotoxicity in rats. Environ. Sci. Pollut. Res. Int. 2019, 26, 18856–18865. [Google Scholar] [CrossRef]
- Guven, M.; Akman, T.; Yener, A.U.; Sehitoglu, M.H.; Yuksel, Y.; Cosar, M. The Neuroprotective Effect of Kefir on Spinal Cord Ischemia/Reperfusion Injury in Rats. J. Korean Neurosurg. Soc. 2015, 57, 335–341. [Google Scholar] [CrossRef] [Green Version]
- El Sayed, N.S.; Kandil, E.A.; Ghoneum, M.H. Probiotics Fermentation Technology, a Novel Kefir Product, Ameliorates Cognitive Impairment in Streptozotocin-Induced Sporadic Alzheimer’s Disease in Mice. Oxid. Med. Cell. Longev. 2021, 2021, 5525306. [Google Scholar] [CrossRef]
- Raz, L.; Knoefel, J.; Bhaskar, K. The neuropathology and cerebrovascular mechanisms of dementia. J. Cereb. Blood Flow Metab. 2016, 36, 172–186. [Google Scholar] [CrossRef] [Green Version]
- Seward, M.E.; Swanson, E.; Norambuena, A.; Reimann, A.; Cochran, J.N.; Li, R.; Roberson, E.D.; Bloom, G.S. Amyloid-β signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer’s disease. J. Cell Sci. 2013, 126, 1278–1286. [Google Scholar] [CrossRef] [Green Version]
- Tönnies, E.; Trushina, E. Oxidative Stress, Synaptic Dysfunction, and Alzheimer’s Disease. J. Alzheimers Dis. 2017, 57, 1105–1121. [Google Scholar] [CrossRef] [Green Version]
- Trist, B.G.; Hare, D.J.; Double, K.L. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging Cell 2019, 18, e13031. [Google Scholar] [CrossRef] [Green Version]
- Gonzalez-Hunt, C.P.; Sanders, L.H. DNA damage and repair in Parkinson’s disease: Recent advances and new opportunities. J. Neurosci. Res. 2021, 99, 180–189. [Google Scholar] [CrossRef]
- Lin, X.; Kapoor, A.; Gu, Y.; Chow, M.J.; Peng, J.; Zhao, K.; Tang, D. Contributions of DNA Damage to Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 1666. [Google Scholar] [CrossRef] [Green Version]
- Ghavami, S.; Shojaei, S.; Yeganeh, B.; Ande, S.R.; Jangamreddy, J.R.; Mehrpour, M.; Christoffersson, J.; Chaabane, W.; Moghadam, A.R.; Kashani, H.H.; et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog. Neurobiol. 2014, 112, 24–49. [Google Scholar] [CrossRef] [Green Version]
- Degterev, A.; Huang, Z.; Boyce, M.; Li, Y.; Jagtap, P.; Mizushima, N.; Cuny, G.D.; Mitchison, T.J.; Moskowitz, M.A.; Yuan, J. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 2005, 1, 112–119. [Google Scholar] [CrossRef] [PubMed]
- Ofengeim, D.; Ito, Y.; Najafov, A.; Zhang, Y.; Shan, B.; DeWitt, J.P.; Ye, J.; Zhang, X.; Chang, A.; Vakifahmetoglu-Norberg, H.; et al. Activation of necroptosis in multiple sclerosis. Cell Rep. 2015, 10, 1836–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pickford, F.; Masliah, E.; Britschgi, M.; Lucin, K.; Narasimhan, R.; Jaeger, P.A.; Small, S.; Spencer, B.; Rockenstein, E.; Levine, B.; et al. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J. Clin. Investig. 2008, 118, 2190–2199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hara, T.; Nakamura, K.; Matsui, M.; Yamamoto, A.; Nakahara, Y.; Suzuki-Migishima, R.; Yokoyama, M.; Mishima, K.; Saito, I.; Okano, H.; et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 2006, 441, 885–889. [Google Scholar] [CrossRef] [PubMed]
- Kovacs, G.G.; Robinson, J.L.; Xie, S.X.; Lee, E.B.; Grossman, M.; Wolk, D.A.; Irwin, D.J.; Weintraub, D.; Kim, C.F.; Schuck, T.; et al. Evaluating the Patterns of Aging-Related Tau Astrogliopathy Unravels Novel Insights into Brain Aging and Neurodegenerative Diseases. J. Neuropathol. Exp. Neurol. 2017, 76, 270–288. [Google Scholar] [CrossRef] [Green Version]
- Liu, Z.; Li, T.; Li, P.; Wei, N.; Zhao, Z.; Liang, H.; Ji, X.; Chen, W.; Xue, M.; Wei, J. The Ambiguous Relationship of Oxidative Stress, Tau Hyperphosphorylation, and Autophagy Dysfunction in Alzheimer’s Disease. Oxid. Med. Cell. Longev. 2015, 2015, 352723. [Google Scholar] [CrossRef]
- Silva, I.; Silva, J.; Ferreira, R.; Trigo, D. Glymphatic system, AQP4, and their implications in Alzheimer’s disease. Neurol. Res. Pract. 2021, 3, 5. [Google Scholar] [CrossRef]
- Angelova, P.R.; Abramov, A.Y. Role of mitochondrial ROS in the brain: From physiology to neurodegeneration. FEBS Lett. 2018, 592, 692–702. [Google Scholar] [CrossRef]
- Dawson, T.M.; Dawson, V.L. Nitric Oxide Signaling in Neurodegeneration and Cell Death. Adv. Pharmacol. 2018, 82, 57–83. [Google Scholar] [CrossRef]
- Marogianni, C.; Sokratous, M.; Dardiotis, E.; Hadjigeorgiou, G.M.; Bogdanos, D.; Xiromerisiou, G. Neurodegeneration and Inflammation-An Interesting Interplay in Parkinson’s Disease. Int. J. Mol. Sci. 2020, 21, 8421. [Google Scholar] [CrossRef] [PubMed]
- Iliff, J.J.; Wang, M.; Liao, Y.; Plogg, B.A.; Peng, W.; Gundersen, G.A.; Benveniste, H.; Vates, G.E.; Deane, R.; Goldman, S.A.; et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 2012, 4, 147ra111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reddy, O.C.; van der Werf, Y.D. The Sleeping Brain: Harnessing the Power of the Glymphatic System through Lifestyle Choices. Brain Sci. 2020, 10, 868. [Google Scholar] [CrossRef]
- Salman, M.M.; Kitchen, P.; Iliff, J.J.; Bill, R.M. Aquaporin 4 and glymphatic flow have central roles in brain fluid homeostasis. Nat. Rev. Neurosci. 2021, 22, 650–651. [Google Scholar] [CrossRef] [PubMed]
- Salman, M.M.; Kitchen, P.; Halsey, A.; Wang, M.X.; Tornroth-Horsefield, S.; Conner, A.C.; Badaut, J.; Iliff, J.J.; Bill, R.M. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis. Brain 2021. [Google Scholar] [CrossRef] [PubMed]
- Giacoppo, S.; Galuppo, M.; Calabrò, R.S.; D’Aleo, G.; Marra, A.; Sessa, E.; Bua, D.G.; Potortì, A.G.; Dugo, G.; Bramanti, P.; et al. Heavy metals and neurodegenerative diseases: An observational study. Biol. Trace Elem. Res. 2014, 161, 151–160. [Google Scholar] [CrossRef]
- Chin-Chan, M.; Navarro-Yepes, J.; Quintanilla-Vega, B. Environmental pollutants as risk factors for neurodegenerative disorders: Alzheimer and Parkinson diseases. Front. Cell. Neurosci. 2015, 9, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Milenkovic, T.; Bozhinovska, N.; Macut, D.; Bjekic-Macut, J.; Rahelic, D.; Velija Asimi, Z.; Burekovic, A. Mediterranean Diet and Type 2 Diabetes Mellitus: A Perpetual Inspiration for the Scientific World. A Review. Nutrients 2021, 13, 1307. [Google Scholar] [CrossRef]
- Nuzzo, D.; Amato, A.; Picone, P.; Terzo, S.; Galizzi, G.; Bonina, F.P.; Mulè, F.; Di Carlo, M. A Natural Dietary Supplement with a Combination of Nutrients Prevents Neurodegeneration Induced by a High Fat Diet in Mice. Nutrients 2018, 10, 1130. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.; Kim, D.H.; Lee, D.H.; Jeon, S.J.; Lee, C.H.; Son, K.H.; Jung, J.W.; Shin, C.Y.; Ryu, J.H. Oroxylin A, a flavonoid, stimulates adult neurogenesis in the hippocampal dentate gyrus region of mice. Neurochem. Res. 2010, 35, 1725–1732. [Google Scholar] [CrossRef]
- Burokas, A.; Moloney, R.D.; Dinan, T.G.; Cryan, J.F. Microbiota regulation of the Mammalian gut-brain axis. Adv. Appl. Microbiol. 2015, 91, 1–62. [Google Scholar] [CrossRef]
- Ochoa-Repáraz, J.; Mielcarz, D.W.; Begum-Haque, S.; Kasper, L.H. Gut, bugs, and brain: Role of commensal bacteria in the control of central nervous system disease. Ann. Neurol. 2011, 69, 240–247. [Google Scholar] [CrossRef]
- Luczynski, P.; McVey Neufeld, K.A.; Oriach, C.S.; Clarke, G.; Dinan, T.G.; Cryan, J.F. Growing up in a Bubble: Using Germ-Free Animals to Assess the Influence of the Gut Microbiota on Brain and Behavior. Int. J. Neuropsychopharmacol. 2016, 19, pyw020. [Google Scholar] [CrossRef]
- Ley, R.E.; Hamady, M.; Lozupone, C.; Turnbaugh, P.J.; Ramey, R.R.; Bircher, J.S.; Schlegel, M.L.; Tucker, T.A.; Schrenzel, M.D.; Knight, R.; et al. Evolution of mammals and their gut microbes. Science 2008, 320, 1647–1651. [Google Scholar] [CrossRef] [Green Version]
- Goldman, J.G.; Postuma, R. Premotor and nonmotor features of Parkinson’s disease. Curr. Opin. Neurol. 2014, 27, 434–441. [Google Scholar] [CrossRef] [Green Version]
- Jalili-Firoozinezhad, S.; Gazzaniga, F.S.; Calamari, E.L.; Camacho, D.M.; Fadel, C.W.; Bein, A.; Swenor, B.; Nestor, B.; Cronce, M.J.; Tovaglieri, A.; et al. A complex human gut microbiome cultured in an anaerobic intestine-on-a-chip. Nat. Biomed. Eng. 2019, 3, 520–531. [Google Scholar] [CrossRef] [PubMed]
- Venkataraman, L.; Fair, S.R.; McElroy, C.A.; Hester, M.E.; Fu, H. Modeling neurodegenerative diseases with cerebral organoids and other three-dimensional culture systems: Focus on Alzheimer’s disease. Stem Cell Rev. Rep. 2020, 1–22. [Google Scholar] [CrossRef] [PubMed]
- Yiannopoulou, K.G.; Papageorgiou, S.G. Current and Future Treatments in Alzheimer Disease: An Update. J. Cent. Nerv. Syst. Dis. 2020, 12, 1179573520907397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bondi, M.W.; Edmonds, E.C.; Salmon, D.P. Alzheimer’s Disease: Past, Present, and Future. J. Int. Neuropsychol. Soc. 2017, 23, 818–831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yáñez, M.; Viña, D. Dual inhibitors of monoamine oxidase and cholinesterase for the treatment of Alzheimer disease. Curr. Top. Med. Chem. 2013, 13, 1692–1706. [Google Scholar] [CrossRef]
- Assal, F. History of Dementia. Front. Neurol. Neurosci. 2019, 44, 118–126. [Google Scholar] [CrossRef]
- Su, B.; Wang, X.; Nunomura, A.; Moreira, P.I.; Lee, H.G.; Perry, G.; Smith, M.A.; Zhu, X. Oxidative stress signaling in Alzheimer’s disease. Curr. Alzheimer Res. 2008, 5, 525–532. [Google Scholar] [CrossRef]
- Butterfield, D.A.; Halliwell, B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat. Rev. Neurosci. 2019, 20, 148–160. [Google Scholar] [CrossRef] [PubMed]
- Alasmari, F.; Alshammari, M.A.; Alasmari, A.F.; Alanazi, W.A.; Alhazzani, K. Neuroinflammatory Cytokines Induce Amyloid Beta Neurotoxicity through Modulating Amyloid Precursor Protein Levels/Metabolism. Biomed. Res. Int. 2018, 2018, 3087475. [Google Scholar] [CrossRef] [PubMed]
- Doecke, J.D.; Laws, S.M.; Faux, N.G.; Wilson, W.; Burnham, S.C.; Lam, C.P.; Mondal, A.; Bedo, J.; Bush, A.I.; Brown, B.; et al. Blood-based protein biomarkers for diagnosis of Alzheimer disease. Arch. Neurol. 2012, 69, 1318–1325. [Google Scholar] [CrossRef] [Green Version]
- Zheng, C.; Zhou, X.W.; Wang, J.Z. The dual roles of cytokines in Alzheimer’s disease: Update on interleukins, TNF-α, TGF-β and IFN-γ. Transl. Neurodegener. 2016, 5, 7. [Google Scholar] [CrossRef] [Green Version]
- Saresella, M.; La Rosa, F.; Piancone, F.; Zoppis, M.; Marventano, I.; Calabrese, E.; Rainone, V.; Nemni, R.; Mancuso, R.; Clerici, M. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease. Mol. Neurodegener. 2016, 11, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- King, E.; O’Brien, J.T.; Donaghy, P.; Morris, C.; Barnett, N.; Olsen, K.; Martin-Ruiz, C.; Taylor, J.P.; Thomas, A.J. Peripheral inflammation in prodromal Alzheimer’s and Lewy body dementias. J. Neurol. Neurosurg. Psychiatry 2018, 89, 339–345. [Google Scholar] [CrossRef] [PubMed]
- Zuroff, L.; Daley, D.; Black, K.L.; Koronyo-Hamaoui, M. Clearance of cerebral Aβ in Alzheimer’s disease: Reassessing the role of microglia and monocytes. Cell. Mol. Life Sci. 2017, 74, 2167–2201. [Google Scholar] [CrossRef] [Green Version]
- Kinney, J.W.; Bemiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef]
- Zhan, X.; Stamova, B.; Jin, L.W.; DeCarli, C.; Phinney, B.; Sharp, F.R. Gram-negative bacterial molecules associate with Alzheimer disease pathology. Neurology 2016, 87, 2324–2332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Sun, G.; Feng, T.; Zhang, J.; Huang, X.; Wang, T.; Xie, Z.; Chu, X.; Yang, J.; Wang, H.; et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res. 2019, 29, 787–803. [Google Scholar] [CrossRef] [PubMed]
- Vendrik, K.E.W.; Ooijevaar, R.E.; de Jong, P.R.C.; Laman, J.D.; van Oosten, B.W.; van Hilten, J.J.; Ducarmon, Q.R.; Keller, J.J.; Kuijper, E.J.; Contarino, M.F. Fecal Microbiota Transplantation in Neurological Disorders. Front. Cell. Infect. Microbiol. 2020, 10, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhattacharjee, S.; Lukiw, W.J. Alzheimer’s disease and the microbiome. Front. Cell. Neurosci. 2013, 7, 153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salami, M. Interplay of Good Bacteria and Central Nervous System: Cognitive Aspects and Mechanistic Considerations. Front. Neurosci. 2021, 15, 613120. [Google Scholar] [CrossRef]
- Bloemendaal, M.; Szopinska-Tokov, J.; Belzer, C.; Boverhoff, D.; Papalini, S.; Michels, F.; van Hemert, S.; Arias Vasquez, A.; Aarts, E. Probiotics-induced changes in gut microbial composition and its effects on cognitive performance after stress: Exploratory analyses. Transl. Psychiatry 2021, 11, 300. [Google Scholar] [CrossRef]
- Sun, M.; Ma, K.; Wen, J.; Wang, G.; Zhang, C.; Li, Q.; Bao, X.; Wang, H. A Review of the Brain-Gut-Microbiome Axis and the Potential Role of Microbiota in Alzheimer’s Disease. J. Alzheimers Dis. 2020, 73, 849–865. [Google Scholar] [CrossRef] [PubMed]
- Valentini, L.; Pinto, A.; Bourdel-Marchasson, I.; Ostan, R.; Brigidi, P.; Turroni, S.; Hrelia, S.; Hrelia, P.; Bereswill, S.; Fischer, A.; et al. Impact of personalized diet and probiotic supplementation on inflammation, nutritional parameters and intestinal microbiota—The “RISTOMED project”: Randomized controlled trial in healthy older people. Clin. Nutr. 2015, 34, 593–602. [Google Scholar] [CrossRef] [Green Version]
- Westfall, S.; Lomis, N.; Kahouli, I.; Dia, S.Y.; Singh, S.P.; Prakash, S. Microbiome, probiotics and neurodegenerative diseases: Deciphering the gut brain axis. Cell. Mol. Life Sci. 2017, 74, 3769–3787. [Google Scholar] [CrossRef]
- Kowalski, K.; Mulak, A. Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J. Neurogastroenterol. Motil. 2019, 25, 48–60. [Google Scholar] [CrossRef] [Green Version]
- Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef]
- Martins, R.N.; Villemagne, V.; Sohrabi, H.R.; Chatterjee, P.; Shah, T.M.; Verdile, G.; Fraser, P.; Taddei, K.; Gupta, V.B.; Rainey-Smith, S.R.; et al. Alzheimer’s Disease: A Journey from Amyloid Peptides and Oxidative Stress, to Biomarker Technologies and Disease Prevention Strategies-Gains from AIBL and DIAN Cohort Studies. J. Alzheimers Dis. 2018, 62, 965–992. [Google Scholar] [CrossRef] [Green Version]
- Baj, A.; Moro, E.; Bistoletti, M.; Orlandi, V.; Crema, F.; Giaroni, C. Glutamatergic Signaling along the Microbiota-Gut-Brain Axis. Int. J. Mol. Sci. 2019, 20, 1482. [Google Scholar] [CrossRef] [Green Version]
- Harris, L.; Griem, J.; Gummery, A.; Marsh, L.; Defres, S.; Bhojak, M.; Das, K.; Easton, A.; Solomon, T.; Kopelman, M. Neuropsychological and psychiatric outcomes in encephalitis: A multi-centre case-control study. PLoS ONE 2020, 15, e0230436. [Google Scholar] [CrossRef] [Green Version]
- Faria, A.V.; Reis, F.; Dabus, G.C.; Zanardi, V.A.; Guerreiro, M.M.; Cendes, F. MRI findings in the diagnosis and monitoring of Rasmussen’s encephalitis. Arq. Neuropsiquiatr. 2009, 67, 792–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, D.; Blümcke, I.; Gui, Q.; Zhou, W.; Zuo, H.; Lin, J.; Luo, Y. Clinico-pathological investigations of Rasmussen encephalitis suggest multifocal disease progression and associated focal cortical dysplasia. Epileptic Disord. 2013, 15, 32–43. [Google Scholar] [CrossRef]
- Pardo, C.A.; Vining, E.P.; Guo, L.; Skolasky, R.L.; Carson, B.S.; Freeman, J.M. The pathology of Rasmussen syndrome: Stages of cortical involvement and neuropathological studies in 45 hemispherectomies. Epilepsia 2004, 45, 516–526. [Google Scholar] [CrossRef] [PubMed]
- Hartman, A.L.; Cross, J.H. Timing of surgery in rasmussen syndrome: Is patience a virtue? Epilepsy Curr. 2014, 14, 8–11. [Google Scholar] [CrossRef]
- Matricardi, S.; Farello, G.; Savasta, S.; Verrotti, A. Understanding Childhood Neuroimmune Diseases of the Central Nervous System. Front. Pediatr. 2019, 7, 511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Varadkar, S.; Bien, C.G.; Kruse, C.A.; Jensen, F.E.; Bauer, J.; Pardo, C.A.; Vincent, A.; Mathern, G.W.; Cross, J.H. Rasmussen’s encephalitis: Clinical features, pathobiology, and treatment advances. Lancet Neurol. 2014, 13, 195–205. [Google Scholar] [CrossRef] [Green Version]
- Dericioglu, N.; Soylemezoglu, F.; Gursoy-Ozdemir, Y.; Akalan, N.; Saygi, S.; Dalkara, T. Cell death and survival mechanisms are concomitantly active in the hippocampus of patients with mesial temporal sclerosis. Neuroscience 2013, 237, 56–65. [Google Scholar] [CrossRef] [PubMed]
- Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbe communication in health and disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nigar, S.; Pottoo, F.H.; Tabassum, N.; Verma, S.K.; Javed, M.N. Molecular insights into the role of inflammation and oxidative stress in epilepsy. J. Adv. Med. Pharm. Sci. 2016, 10, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Zhang, Y.; Yang, H.; Rao, Y.; Miao, J.; Lu, X. Intestinal microbiota as an alternative therapeutic target for epilepsy. Can. J. Infect. Dis. Med. Microbiol. 2016, 2016, 9032809. [Google Scholar] [CrossRef] [Green Version]
- Sherwin, E.; Dinan, T.G.; Cryan, J.F. Recent developments in understanding the role of the gut microbiota in brain health and disease. Ann. N. Y. Acad. Sci. 2018, 1420, 5–25. [Google Scholar] [CrossRef]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
- Shreiner, A.B.; Kao, J.Y.; Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol. 2015, 31, 69–75. [Google Scholar] [CrossRef]
- Zhao, W.; Caro, F.; Robins, W.; Mekalanos, J.J. Antagonism toward the intestinal microbiota and its effect on Vibrio cholerae virulence. Science 2018, 359, 210–213. [Google Scholar] [CrossRef] [Green Version]
- Kobylarek, D.; Iwanowski, P.; Lewandowska, Z.; Limphaibool, N.; Szafranek, S.; Labrzycka, A.; Kozubski, W. Advances in the Potential Biomarkers of Epilepsy. Front. Neurol. 2019, 10, 685. [Google Scholar] [CrossRef]
- Hermann, G.E.; Rogers, R.C.; Bresnahan, J.C.; Beattie, M.S. Tumor necrosis factor-alpha induces cFOS and strongly potentiates glutamate-mediated cell death in the rat spinal cord. Neurobiol. Dis. 2001, 8, 590–599. [Google Scholar] [CrossRef] [Green Version]
- Lagarde, S.; Villeneuve, N.; Trébuchon, A.; Kaphan, E.; Lepine, A.; McGonigal, A.; Roubertie, A.; Barthez, M.A.; Trommsdorff, V.; Lefranc, J.; et al. Anti-tumor necrosis factor alpha therapy (adalimumab) in Rasmussen’s encephalitis: An open pilot study. Epilepsia 2016, 57, 956–966. [Google Scholar] [CrossRef]
- Peng, A.; Qiu, X.; Lai, W.; Li, W.; Zhang, L.; Zhu, X.; He, S.; Duan, J.; Chen, L. Altered composition of the gut microbiome in patients with drug-resistant epilepsy. Epilepsy Res. 2018, 147, 102–107. [Google Scholar] [CrossRef] [PubMed]
- Lum, G.R.; Olson, C.A.; Hsiao, E.Y. Emerging roles for the intestinal microbiome in epilepsy. Neurobiol. Dis. 2020, 135, 104576. [Google Scholar] [CrossRef]
- Passos, M.; Moraes-Filho, J.P. INTESTINAL MICROBIOTA IN DIGESTIVE DISEASES. Arq. Gastroenterol. 2017, 54, 255–262. [Google Scholar] [CrossRef] [Green Version]
- Strati, F.; Cavalieri, D.; Albanese, D.; De Felice, C.; Donati, C.; Hayek, J.; Jousson, O.; Leoncini, S.; Renzi, D.; Calabrò, A.; et al. New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome 2017, 5, 24. [Google Scholar] [CrossRef] [Green Version]
- Olsson, E.; Byberg, L.; Höijer, J.; Kilander, L.; Larsson, S.C. Milk and Fermented Milk Intake and Parkinson’s Disease: Cohort Study. Nutrients 2020, 12, 2763. [Google Scholar] [CrossRef]
- Chen, H.; O’Reilly, E.; McCullough, M.L.; Rodriguez, C.; Schwarzschild, M.A.; Calle, E.E.; Thun, M.J.; Ascherio, A. Consumption of dairy products and risk of Parkinson’s disease. Am. J. Epidemiol. 2007, 165, 998–1006. [Google Scholar] [CrossRef]
- Hughes, K.C.; Gao, X.; Kim, I.Y.; Wang, M.; Weisskopf, M.G.; Schwarzschild, M.A.; Ascherio, A. Intake of dairy foods and risk of Parkinson disease. Neurology 2017, 89, 46–52. [Google Scholar] [CrossRef]
- Swanson, K.S.; Gibson, G.R.; Hutkins, R.; Reimer, R.A.; Reid, G.; Verbeke, K.; Scott, K.P.; Holscher, H.D.; Azad, M.B.; Delzenne, N.M.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 687–701. [Google Scholar] [CrossRef]
Chemical Compound | Origin and Amounty of Kefir Animal Milk (Measurement Data) | Biological Activity | |||
---|---|---|---|---|---|
Cow | Goat | Sheep | Camel | ||
Ash | 0.7% [165] 1.1% [167] 1.2 % [168] 0.5 % (w/v) [152] | 0.62% w/w [169] | 0.94g [170] | Inorganic residue | |
Carbohydrates and Lactose | 6.0% [165] 5.0 % (w/v) [152] | 4.6 % (Lactose) [171] 5.9% w/w [169] | 5% (Lactose) [171] 2.92 g (Lactose) [170] 4.11 g [170] | Basic nutrient | |
Grain Exopolysaccharide | 2 g/L Kefiran [172] No measurements have been reported about kefiran concentration in the fermented kefir | Antihypertensive [173] Immunomodulation [40,174] Antitumoral [175] Antimicrobial [160] Antioxidant [176] | |||
Fats | 0.2% [165] 2.3% w/v [166] 1.3 % w/v [152] | 3.4% w/v [171] 2.6% w/w [169] | 5.3% w/v [171] 5.21 g [170] | Basic nutrient | |
Vitamin A and Carotene | B- Carotene 0.005 mg/100 g [177] | B- carotene 0.004 mg/100 g [177] | Antioxidant [177] | ||
Vitamin E | 0.025 mg/100 g [177] | 0.04 mg/100 g [177] | Antioxidant [177] | ||
Vitamin B1 | 10 mg/Kg [168] | Basic nutrient | |||
Vitamin B2 | 5 mg/Kg [168] | Basic nutrient | |||
Vitamin B5 | 3 mg/Kg [168] | Basic nutrient | |||
Protein | 3.0% [165] 3.9% w/v [166] 3.1% w/v [152] | 3.45% w/v [171] | 4.1% w/v [171] 5.71 g [170] | Basic nutrient | |
Total Nitrogen (%) | 0.21 [178] | 0.25 [178] | 0.24 [178] | 0.23 [178] | |
Bioactive Peptides | AVPYPQR [9] Kefir peptides powder—KEFPEP [179,180,181,182,183] | LEIVPK [184] ARHPHPHLSFM [185] LGPVRGPFP [185] TAQVTSTEV [185] VLNENLLR [185] | (Antihypertensive) DKIHPF [170] (Antioxidant) QEPVLGPVRGPFP [170] | “Captopril like effect” [9,184,185] Antihypertensive [170] Antioxidant [9,170,181,185] Lipid metabolism modulation [180] Antifibrotic [181] Antithrombotic [185] Anti-inflammatory [179,181,182] Prevent menopausal osteoporosis [183] Prevent non-alcoholic fatty liver disease [179] Antidepressant [181] Antibacterial [185] | |
Aminoacids (threonine, serine, alanine, lysine, valine, isoleucine, methionine, phenylalanine and tryptophan) | Ranging 60–376 mg/100 g [168,186,187] 6–46 mg/kg [178] | 4–52 mg/ kg [178] | 1–26 mg/kg [178] | 7–56 mg/kg [178] | Basic nutrient |
Phenolic compounds | 67 mg/100mL [177] | 50–70mg/100mL [188] | 78 mg/100mL [177] | Antioxidant [189,190] | |
Lactic acid | 1.0% [165] 1.4–17.4 mg/mL [166] 0.6% w/v [152] | 7.1 g/L [171] | 9.1 g/L [171] | ||
Alcohol | 0.48% [165] 0.5mg/mL [166] | 1% [191] 9.5 ug/g [171] | 12.5 ug/g [171] | ||
CO2 | 201–277 mL/L [165] | ||||
Potassium | 1.65% [168] 1009 ppm [178] | 159 mg/100 g [169] 1049 ppm [178] | 11.2 mg/L [170] 88.3 ppm [178] | 1477 ppm [178] | Basic nutrient |
Calcium | 0.86% [168] 0.22% [166] 88 ppm [178] | 116 mg/100 g [169] 132 ppm [178] | 1765 mg/L [170] 37 ppm [178] | 154 ppm [178] | Basic nutrient |
Phosphorus | 1.45% [168] 733 ppm [178] | 735 ppm [178] | 458 ppm [178] | 783 ppm [178] | Basic nutrient |
Magnesium | 0.3% [168] 100.3 ppm [178] | 10.3 mg/100 g [169] 203 ppm [178] | 206 mg/L [170] 116.7 ppm [178] | 83.4 ppm [178] | Basic nutrient |
Sodium | 15.6 ppm [178] | 17.1 ppm [178] | 20.73 ppm [178] | 20.83 ppm [178] | Basic nutrient |
Copper | 7.32 mg/Kg [168] 0.06 ppm [178] | 0.08 ppm [178] | 0.02 ppm [178] | 0.01 ppm [178] | Basic nutrient |
Zinc | 92.7 mg/Kg [168] 2.17 ppm [178] | 0.34 mg/100 g [169] 2.23 ppm [178] | 0.55 ppm [178] | 2.43 ppm [178] | Basic nutrient |
Iron | 20.3 mg/Kg [168] 4.73 ppm [178] | 2.25 ppm [178] | 3.53 ppm [178] | 4.86 ppm [178] | Basic nutrient |
Manganese | 13 mg/Kg [168] 0.25 ppm [178] | 0.27 ppm [178] | 0.21 ppm [178] | 0.19 ppm [178] | Basic nutrient |
Cobalt | 0.16 mg/Kg [168] | Basic nutrient | |||
Molybdenum | 0.33 mg/Kg [168] 0 ppm [178] | 0.01 ppm [178] | 0.03 ppm [178] | 0 ppm [178] | Basic nutrient |
pH | 4.54 [178] | 4.6 [178] | 4.51 [178] | 4.1 [178] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pereira, T.M.C.; Côco, L.Z.; Ton, A.M.M.; Meyrelles, S.S.; Campos-Toimil, M.; Campagnaro, B.P.; Vasquez, E.C. The Emerging Scenario of the Gut–Brain Axis: The Therapeutic Actions of the New Actor Kefir against Neurodegenerative Diseases. Antioxidants 2021, 10, 1845. https://doi.org/10.3390/antiox10111845
Pereira TMC, Côco LZ, Ton AMM, Meyrelles SS, Campos-Toimil M, Campagnaro BP, Vasquez EC. The Emerging Scenario of the Gut–Brain Axis: The Therapeutic Actions of the New Actor Kefir against Neurodegenerative Diseases. Antioxidants. 2021; 10(11):1845. https://doi.org/10.3390/antiox10111845
Chicago/Turabian StylePereira, Thiago M. C., Larissa Z. Côco, Alyne M. M. Ton, Silvana S. Meyrelles, Manuel Campos-Toimil, Bianca P. Campagnaro, and Elisardo C. Vasquez. 2021. "The Emerging Scenario of the Gut–Brain Axis: The Therapeutic Actions of the New Actor Kefir against Neurodegenerative Diseases" Antioxidants 10, no. 11: 1845. https://doi.org/10.3390/antiox10111845
APA StylePereira, T. M. C., Côco, L. Z., Ton, A. M. M., Meyrelles, S. S., Campos-Toimil, M., Campagnaro, B. P., & Vasquez, E. C. (2021). The Emerging Scenario of the Gut–Brain Axis: The Therapeutic Actions of the New Actor Kefir against Neurodegenerative Diseases. Antioxidants, 10(11), 1845. https://doi.org/10.3390/antiox10111845