The Role of Diet as a Modulator of the Inflammatory Process in the Neurological Diseases
Abstract
:1. Introduction
2. Mechanism of Action Associated with Diets and Neurological Disease Development
2.1. Diet, Inflammation, and Its Impact on Neurological Disease Development
2.1.1. Inflammation-Promoting Diet
Dietary Inflammation Index (DII)
2.1.2. The Pyramid of Anti-Inflammatory Nutrition
2.1.3. Antiinflammatory Components of the Diet
Fruits and Vegetables
Herbs and Spices
Zingiber Officinale
Herbs and Spices | Major Bioactive Compounds | Potential Beneficial Effects | Anti-Inflammatory Mechanism | References |
---|---|---|---|---|
Garlic (Allium sativum L.) | allicin, quercetin, kaempferol | anti-inflammatory | suppress the production of inflammatory cytokines | [55] |
Turmeric (Curcuma longa L.) | curcumin, demethoxycurcumin, bisdemethoxycurcumin | anti-inflammatory | inhibits microglial activation and reduces cytokine release | [54] |
Ginger (Zingiber officinale Roscoe) | gingerols, shogaols, paradols | anti-inflammatory antioxidant analgesic | Inhibit cyclooxygenase (COX) and 5- lipoxygenase (LOX), and downregulation of the expression of pro- inflammatory genes suppress the production of inflammatory cytokines | [56,57,58] |
Rosemary (Rosmarinus officinalis L.) | carnosic acid, betulinic acid, carnosol, ursolic acid | anti-inflammatory antioxidant | inhibit the release of pro-inflammatory mediators like NOx, IL-1, and TNF-1α while reducing leukocyte activation | [59,60,61] |
Vegetable Protein: Soybeans and Other Legumes
Vegetable Oils Rich in Unsaturated Fatty Acids
Nuts and Seeds
Tea Beverages
Coffee
Red Wine
Dark Chocolate
2.2. Diet and Microbiome and Its Effects on Neurological Disease Development and Its Outcomes
3. The Significance of Adopting an Anti-Inflammatory Diet for the Prevention and Treatment of Neurodegenerative Disorders
3.1. Alzheimer’s Disease
3.2. Parkinson’s Disease
3.3. Huntington’s Disease
3.4. Multiple Sclerosis
4. Anti-Inflammatory Diet and Its Role in Mental Health
4.1. Depression
4.2. Schizophrenia
4.3. Bipolar Disorder
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Maton, A.; Hopkins, J.; McLaughlin, C.W.; Johnson, S.; Warner, M.Q.; LaHart, D.; Wright, J.D. Human Biology and Health; Prentice Hall: Englewood Cliffs, NJ, USA, 1993; pp. 132–144. [Google Scholar]
- Di Luca, M.; Nutt, D.; Oertel, W.; Boyer, P.; Jaarsma, J.; Destrebecq, F.; Esposito, G.; Quoidbach, V. Towards Earlier Diagnosis and Treatment of Disorders of the Brain. Bull. World Health Organ. 2018, 96, 298–298A. [Google Scholar] [CrossRef]
- Gustavsson, A.; Svensson, M.; Jacobi, F.; Allgulander, C.; Alonso, J.; Beghi, E.; CDBE2010 Study Group. Cost of disorders of the brain in Europe 2010. Eur. Neuropsychopharmacol. 2011, 21, 718–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neurological Disorders Collaborator Group (NDCG). Global, regional, and national burden of neurological disorders during 1990–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet 2017, 16, 877–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gomez-Pinilla, F.; Gomez, A.G. The influence of dietary factors in central nervous system plasticity and injury recovery. Phys. Med. Rehabil. 2011, 3, 111–116. [Google Scholar] [CrossRef] [Green Version]
- Hanamsagar, R.; Bilbo, S.D. Sex differences in neurodevelopmental and neurodegenerative disorders: Focus on microglial function and neuroinflammation during development. J. Steroid Biochem. Mol. Biol. 2016, 160, 127–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, G.H.; Kim, J.E.; Rhie, S.J.; Yoon, S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef] [Green Version]
- Estrada, J.A.; Contreras, I. Nutritional modulation of immune and central nervous system homeostasis: The role of diet in development of neuroinflammation and neurological disease. Nutrients 2019, 11, 1076. [Google Scholar] [CrossRef] [Green Version]
- Mirza, S.K.; Asema, U.K.; Kasim, S.S. To study the harmful effects of food preservatives on human health. J. Med. Chem. Drug Des. 2017, 2, 610–616. [Google Scholar]
- Gupta, R.; Yadav, R.K. Impact Of Chemical Food Preservatives on Human Health. PalArch’s J. Archaeol. Egypt Egyptol. 2021, 18, 811–818. [Google Scholar]
- Pascale, A.; Marchesi, N.; Govoni, S.; Barbieri, A. Targeting the microbiota in pharmacology of psychiatric disorders. Pharm. Res. 2020, 157, 104856. [Google Scholar] [CrossRef]
- Fond, G.; Boukouaci, W.; Chevalier, G.; Regnault, A.; Eberl, G.; Hamdani, N.; Leboyer, M. The “psychomicrobiotic”: Targeting microbiota in major psychiatric disorders: A systematic review. Pathol. Biol. 2015, 63, 35–42. [Google Scholar] [CrossRef]
- Slyepchenko, A.; Maes, M.; Jacka, F.N.; Kohler, C.A.; Barichello, T.; McIntyre, R.S.; Berk, M. Gut microbiota, bacterial translocation, and interactions with diet: Pathophysiological links between major depressive disorder and non-communicable medical comorbidities. Psychother. Psychosom. 2016, 85, 31–46. [Google Scholar] [CrossRef] [Green Version]
- Kelly, J.R.; Borre, Y.; O’Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P.J.; Beers, S.; Scott, K.; Moloney, G.; et al. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109–118. [Google Scholar] [CrossRef]
- Soilu-Hänninen, M.; Åivo, J.; Lindström, B.M.; Elovaara, I.; Sumelahti, M.L.; Färkkilä, M.; Tienari, P.; Atula, S.; Sarasoja, T.; Herrala, L.; et al. A randomised, double blind, placebo controlled trial with vitamin D3 as an add on treatment to interferon β-1b in patients with multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2012, 83, 565–571. [Google Scholar] [CrossRef]
- Noble, E.E.; Kanoski, S.E. Early life exposure to obesogenic diets and learning and memory dysfunction. Curr. Opin. Behav. Sci. 2016, 9, 7–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bingham, M. Obesity-Related Altered Brain Responses to Simple Sugars: Implications for Weight Gain. Diabetes 2016, 65, 1868–1882. [Google Scholar]
- DeSalvo, K.B.; Olson, R.; Casavale, K.O. Dietary guidelines for Americans. JAMA 2016, 315, 457–458. [Google Scholar] [CrossRef] [Green Version]
- Martínez Leo, E.E.; Peñafiel, A.M.; Escalante, V.M.H.; Cabrera Araujo, Z.M. Ultra-processed diet, systemic oxidative stress, and breach of immunologic tolerance. Nutrition 2021, 91–92, 111419. [Google Scholar] [CrossRef] [PubMed]
- Zmora, N.; Suez, J.; Elinav, E. You are what you eat: Diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef] [Green Version]
- Velloso, L.A. The brain is the conductor: Diet-induced inflammation overlapping physiological control of body mass and metabolism. Arq. Bras. Endocrinol. Metabol. 2009, 53, 151–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christ, A.; Lauterbach, M.; Latz, E. Western Diet and the Immune System: An Inflammatory Connection. Immunity 2019, 51, 794–811. [Google Scholar] [CrossRef] [PubMed]
- Hyatt, H.; Deminice, R.; Yoshihara, T.; Powers, S.K. Mitochondrial dysfunction induces muscle atrophy during prolonged inactivity: A review of the causes and effects. Arch. Biochem. Biophys. 2019, 662, 49–60. [Google Scholar] [CrossRef]
- Sears, B.; Ricordi, C. Anti-inflammatory nutrition as a pharmacological approach to treat obesity. J. Obes. 2011, 2011, 431985. [Google Scholar] [CrossRef] [PubMed]
- Galland, L. Diet and inflammation. Nutr. Clin. Pract. 2010, 25, 634–640. [Google Scholar] [CrossRef]
- Freeman, L.R.; Haley-Zitlin, V.; Rosenberger, D.S.; Granholm, A.C. Damaging effects of a high-fat diet to the brain and cognition: A review of proposed mechanisms. Nutr. Neurosci. 2014, 17, 241–251. [Google Scholar] [CrossRef]
- Saha, S.K.; Lee, S.B.; Won, J.; Choi, H.Y.; Kim, K.; Yang, G.M.; Dayem, A.A.; Cho, S. Correlation between Oxidative Stress, Nutrition, and Cancer Initiation. Int. J. Mol. Sci. 2017, 18, 1544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freeman, L.R.; Granholm, A.C. Vascular changes in rat hippo-campus following a high saturated fat and cholesterol diet. J. Cereb. Blood Flow Metab. 2012, 32, 643–653. [Google Scholar] [CrossRef] [Green Version]
- Silveira, B.K.S.; Oliveira, T.M.S.; Andrade, P.A.; Hermsdorff, H.H.M.; Rosa, C.D.O.B.; Franceschini, S.D.C.C. Dietary pattern and macronutrients profile on the variation of inflammatory biomarkers: Scientific Update. Cardiol. Res. Pract. 2018, 2018, 4762575. [Google Scholar] [CrossRef] [Green Version]
- Linecker, M.; Limani, P.; Botea, F.; Popescu, I.; Alikhanov, R.; Efanov, M.; Kim, P.; Khatkov, I.; Raptis, D.A.; Tschuor, C.; et al. A randomized, double-blind study of the effects of omega-3 fatty acids (Omegaven™) on outcome after major liver resection. BMC Gastroenterol. 2015, 15, 102. [Google Scholar] [CrossRef] [Green Version]
- Montonen, J.; Boeing, H.; Fritsche, A.; Schleicher, E.; Joost, H.G.; Schulze, M.B.; Pischon, T. Consumption of red meat and whole-grain bread in relation to biomarkers of obesity, inflammation, glucose metabolism and oxidative stress. Eur. J. Nutr. 2013, 52, 337–345. [Google Scholar] [CrossRef] [Green Version]
- Zhong, X.; Guo, L.; Zhang, L.; Li, Y.; He, R.; Cheng, G. Inflammatory potential of diet and risk of cardiovascular disease or mortality: A meta-analysis. Sci. Rep. 2017, 7, 6367. [Google Scholar] [CrossRef] [PubMed]
- Galas, A.; Kulig, P.; Kulig, P. Dietary inflammatory index as a potential determinant of a length of hospitalization among surgical patients treated for colorectal cancer. Eur. J. Clin. Nutr. 2014, 68, 1168–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weil, A.W. Anti Inflammatory Diet and Pyramid. Anti-Inflammatory Diet. 2012. Available online: https://www.drweil.com/diet-nutrition/anti-inflammatory-diet-pyramid/dr-weils-anti-inflammatory-food-pyramid/ (accessed on 11 March 2023).
- Rondanelli, M.; Faliva, M.A.; Miccono, A.; Naso, M.; Nichetti, M.; Riva, A.; Guerriero, F.; De Gregori, M.; Peroni, G.; Perna, S. Food pyramid for subjects with chronic pain: Foods and dietary constituents as anti-inflammatory and antioxidant agents. Nutr. Res. Rev. 2018, 31, 131–151. [Google Scholar] [CrossRef]
- McGrattan, A.M.; McGuinness, B.; McKinley, M.C.; Kee, F.; Passmore, P.; Woodside, J.V.; McEvoy, C.T. Diet and Inflammation in Cognitive Ageing and Alzheimer’s Disease. Curr. Nutr. Rep. 2019, 8, 53–65. [Google Scholar] [CrossRef] [Green Version]
- Kamat, C.D.; Gadal, S.; Mhatre, M.; Williamson, K.S.; Pye, Q.N.; Hensley, K. Antioxidants in central nervous system diseases: Preclinical promise and translational challenges. J. Alzheimer’s Dis. 2008, 15, 473–493. [Google Scholar] [CrossRef] [Green Version]
- Román, G.C.; Jackson, R.E.; Gadhia, R.; Román, A.N.; Reis, J. Mediterranean diet: The role of long-chain ω-3 fatty acids in fish, polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine, probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Nat. Rev. Neurol. 2019, 175, 724–741. [Google Scholar] [CrossRef]
- Liang, S.; Wu, X.; Hu, X.; Wang, T.; Jin, F. Recognizing Depression from the Microbiota–Gut–Brain Axis. Int. J. Mol. Sci. 2018, 19, 1592. [Google Scholar] [CrossRef] [Green Version]
- Mousavi-Shirazi-Fard, Z.; Mazloom, Z.; Izadi, S.; Fararouei, M. The effects of modified anti-inflammatory diet on fatigue, quality of life, and inflammatory biomarkers in relapsing-remitting multiple sclerosis patients: A randomized clinical trial. Int. J. Neurosci. 2021, 131, 657–665. [Google Scholar] [CrossRef]
- Akbari, M.; Ostadmohammadi, V.; Lankarani, K.B.; Tabrizi, R.; Kolahdooz, F.; Khatibi, S.R.; Asemi, Z. The effects of alpha-lipoic acid supplementation on glucose control and lipid profiles among patients with metabolic diseases: A systematic review and meta-analysis of randomized controlled trials. Metabolism 2018, 87, 56–69. [Google Scholar] [CrossRef] [PubMed]
- Phillips, M.C.; Murtagh, D.K.; Gilbertson, L.J.; Asztely, F.J.; Lynch, C.D. Low-fat versus ketogenic diet in Parkinson’s disease: A pilot randomized controlled trial. Mov. Disord. 2018, 33, 1306–1314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, B.; Parsaik, A.K.; Mielke, M.M.; Erwin, P.J.; Knopman, D.S.; Petersen, R.C.; Roberts, R.O. Association of Mediterranean Diet with Mild Cognitive Impairment and Alzheimer’s Disease: A Systematic Review and Meta-Analysis. J. Alzheimer’s Dis. 2014, 39, 271–282. [Google Scholar] [CrossRef] [Green Version]
- Paknahad, Z.; Sheklabadi, E.; Moravejolahkami, A.R.; Chitsaz, A.; Hassanzadeh, A. The effects of Mediterranean diet on severity of disease and serum Total Antioxidant Capacity (TAC) in patients with Parkinson’s disease: A single center, randomized controlled trial. Nutr. Neurosci. 2020, 25, 313–320. [Google Scholar] [CrossRef]
- Zhang, Y.; Chen, J.; Qiu, J.; Li, Y.; Wang, J.; Jiao, J. Intakes of fish and polyunsaturated fatty acids and mild-to-severe cognitive impairment risks: A dose-response meta-analysis of 21 cohort studies1–3. Am. J. Clin. Nutr. 2016, 103, 330–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, X.; Chen, H.; Fung, T.T.; Logroscino, G.; Schwarzschild, M.A.; Hu, F.B.; Ascherio, A. Prospective study of dietary pattern and risk of Parkinson disease. Am. J. Clin. Nutr. 2007, 86, 1486–1494. [Google Scholar] [CrossRef] [Green Version]
- Lai, J.S.; Hiles, S.; Bisquera, A.; Hure, A.J.; McEvoy, M.; Attia, J. A systematic review and meta-analysis of dietary patterns and depression in community-dwelling adults. Am. J. Clin. Nutr. 2014, 99, 181–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- El-Mallakh, R.; Paskitti, M. The ketogenic diet may have mood-stabilizing properties. Med. Hypotheses 2001, 57, 724–726. [Google Scholar] [CrossRef] [PubMed]
- Saita, E.; Kondo, K.; Momiyama, Y. Anti-inflammatory diet for atherosclerosis and coronary artery disease: Antioxidant foods. Clin. Med. Insights Cardiol. 2014, 8, CMC-S17071. [Google Scholar] [CrossRef] [Green Version]
- Assmann, K.E.; Lassale, C.; Andreeva, V.A.; Jeandel, C.; Hercberg, S.; Galan, P.; Kesse-Guyot, E. A healthy dietary pattern at midlife, combined with a regulated energy intake, is related to increased odds for healthy aging. J. Nutr. 2015, 145, 2139–2145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holt, E.M.; Steffen, L.M.; Moran, A.; Basu, S.; Steinberger, J.; Ross, J.A.; Hong, C.P.; Sinaiko, A.R. Fruit and vegetable consumption and its relation to markers of inflammation and oxidative stress in adolescents. J. Am. Diet. Assoc. 2009, 109, 414–421. [Google Scholar] [CrossRef] [Green Version]
- Aggarwal, B.B.; Van Kuiken, M.E.; Iyer, L.H.; Harikumar, K.B.; Sung, B. Molecular targets of nutraceuticals derived from dietary spices: Potential role in suppression of inflammation and tumorigenesis. Exp. Biol. Med. 2009, 234, 825–849. [Google Scholar] [CrossRef] [Green Version]
- Serafini, M.; Peluso, I. Functional foods for health: The interrelated antioxidant and anti-inflammatory role of fruits, vegetables, herbs, spices and cocoa in humans. Curr. Pharm. Des. 2016, 22, 6701–6715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rubió, L.; Motilva, M.J.; Romero, M.P. Recent advances in biologically active compounds in herbs and spices: A review of the most effective antioxidant and anti-inflammatory active principles. Crit. Rev. Food Sci. Nutr. 2013, 53, 943–953. [Google Scholar] [CrossRef] [PubMed]
- Lee, D.Y.; Li, H.; Lim, H.J.; Lee, H.J.; Jeon, R.; Ryu, J.H. Anti-inflammatory activity of sulfur-containing compounds from garlic. J. Med. Food 2012, 15, 992–999. [Google Scholar] [CrossRef] [Green Version]
- Thomson, M.; Al-Qattan, K.K.; Al-Sawan, S.M.; Alnaqeeb, M.A.; Khan, I.; Ali, M. The use of ginger (Zingiber officinale Rosc.) as a potential anti-inflammatory and antithrombotic agent. Prostaglandins Leukot. Essent. Fat. Acids 2002, 67, 475–478. [Google Scholar] [CrossRef]
- Habib, S.H.M.; Makpol, S.; Hamid, N.A.A.; Das, S.; Ngah, W.Z.W.; Yusof, Y.A.M. Ginger extract (Zingiber officinale) has anti-cancer and anti-inflammatory effects on ethionine-induced hepatoma rats. Clinics 2008, 63, 807–813. [Google Scholar] [CrossRef] [Green Version]
- Pagano, E.; Souto, E.B.; Durazzo, A.; Sharifi-Rad, J.; Lucarini, M.; Souto, S.B.; Romano, B. Ginger (Zingiber officinale Roscoe) as a nutraceutical: Focus on the metabolic, analgesic, and antiinflammatory effects. Phytother. Res. 2021, 35, 2403–2417. [Google Scholar] [CrossRef]
- Altinier, G.; Sosa, S.; Aquino, R.P.; Mencherini, T.; Loggia, R.D.; Tubaro, A. Characterization of topical antiinflammatory compounds in Rosmarinus officinalis L. J. Agric. Food Chem. 2007, 55, 1718–1723. [Google Scholar] [CrossRef]
- Benincá, J.P.; Dalmarco, J.B.; Pizzolatti, M.G.; Fröde, T.S. Analysis of the anti-inflammatory properties of Rosmarinus officinalis L. in mice. Food Chem. 2011, 124, 468–475. [Google Scholar] [CrossRef]
- Juhás, Š.; Bukovská, A.; Čikoš, Š.; Czikková, S.; Fabian, D.; Koppel, J. Anti-inflammatory effects of Rosmarinus officinalis essential oil in mice. Acta Vet. Brno 2009, 78, 121–127. [Google Scholar] [CrossRef] [Green Version]
- Hirohata, M.; Ono, K.; Takasaki, J.; Takahashi, R.; Ikeda, T.; Morinaga, A.; Yamada, M. Anti-amyloidogenic effects of soybean isoflavones in vitro: Fluorescence spectroscopy demonstrating direct binding to Aβ monomers, oligomers and fibrils. Biochim. Biophys. Acta 2012, 1822, 1316–1324. [Google Scholar] [CrossRef] [Green Version]
- Yamagata, K. Soy Isoflavones Inhibit Endothelial Cell Dysfunction and Prevent Cardiovascular Disease. J. Cardiovasc. Pharmacol. 2019, 74, 201–209. [Google Scholar] [CrossRef]
- Zhang, T.; Dou, W.; Zhang, X.; Zhao, Y.; Zhang, Y.; Jiang, L.; Sui, X. The development history and recent updates on soy protein-based meat alternatives. Trends Food Sci. Technol. 2021, 109, 702–710. [Google Scholar] [CrossRef]
- Sekikawa, A.; Higashiyama, A.; Lopresti, B.J.; Ihara, M.; Aizenstein, H.; Watanabe, M.; Chang, Y.; Kakuta, C.; Yu, Z.; Mathis, C.; et al. Associations of equol-producing status with white matter lesion and amyloid-β deposition in cognitively normal elderly Japanese. Alzheimer’s Dement. 2020, 6, 12089–12098. [Google Scholar] [CrossRef]
- Volpe, R.; Stefano, P.; Massimiliano, M.; Francesca, M.; Gianluca, S.; Federica, R. Healthy fats for healthy nutrition. An educational approach in the workplace to regulate food choices and improve prevention of non-communicable diseases. High Blood Press. Cardiovasc. Prev. 2015, 22, 395–401. [Google Scholar] [CrossRef] [PubMed]
- Fernandes, J.; Fialho, M.; Santos, R.; Peixoto-Placido, C.; Madeira, T.; Sousa-Santos, N.; Carneiro, A.V. Is olive oil good for you? A systematic review and meta-analysis on anti-inflammatory benefits from regular dietary intake. Nutrition 2020, 69, 110559. [Google Scholar] [CrossRef] [PubMed]
- Mazza, E.; Fava, A.; Ferro, Y.; Rotundo, S.; Romeo, S.; Bosco, D.; Montalcini, T. Effect of the replacement of dietary vegetable oils with a low dose of extravirgin olive oil in the Mediterranean Diet on cognitive functions in the elderly. J. Transl. Med. 2018, 16, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santangelo, C.; Vari, R.; Scazzocchio, B.; De Sanctis, P.; Giovannini, C.; D’Archivio, M.; Masella, R. Anti-inflammatory activity of extra virgin olive oil polyphenols: Which role in the prevention and treatment of immune-mediated inflammatory diseases? Endocr. Metab. Immun. Disord. Drug Targets 2018, 18, 36–50. [Google Scholar] [CrossRef]
- Materac, E.; Marczyński, Z.; Bodek, K.H. Rola kwasów tłuszczowych omega-3 i omega-6 w organizmie człowieka. [The role of omega-3 and omega-6 fatty acids in the human body]. Bromat. Chem. Toksykol. 2013, 46, 225–233. [Google Scholar]
- Gorji, N.; Moeini, R.; Memariani, Z. Almond, hazelnut and walnut, three nuts for neuroprotection in Alzheimer’s disease: A neuropharmacological review of their bioactive constituents. Pharmacol. Res. 2018, 129, 115–127. [Google Scholar] [CrossRef] [PubMed]
- Hu, N.; Yu, J.T.; Tan, L.; Wang, Y.-L.; Sun, L.; Tan, L. Nutrition and the Risk of Alzheimer’s Disease. BioMed. Res. Int. 2013, 2013, 524820. [Google Scholar] [CrossRef] [Green Version]
- Eskelinen, M.H.; Kivipelto, M. Caffeine as a Protective Factor in Dementia and Alzheimer’s Disease. J. Alzheimer’s Dis. 2010, 20, 167–174. [Google Scholar] [CrossRef] [Green Version]
- Barranco Quintana, J.L.; Allam, M.F.; Del Castillo, A.S.; Navajas, R.F.-C. Alzheimer’s disease and coffee: A quantitative review. Neurol. Res. 2007, 29, 91–95. [Google Scholar] [CrossRef]
- Zale, E.L.; Powers, J.M.; Ditre, J.W. Cognitive-affective transdiagnostic factors associated with vulnerability to alcohol and prescription opioid use in the context of pain. Alcohol Res. Curr. Rev. 2021, 41, 8. [Google Scholar] [CrossRef]
- Zaw, J.J.T.; Howe, P.R.; Wong, R.H. Long-term effects of resveratrol on cognition, cerebrovascular function and cardio-metabolic markers in postmenopausal women: A 24-month randomised, double-blind, placebo-controlled, crossover study. Clin. Nutr. 2021, 40, 820–829. [Google Scholar]
- Makkar, R.; Behl, T.; Bungau, S.; Zengin, G.; Mehta, V.; Kumar, A. Nutraceuticals in neurological disorders. Int. J. Mol. Sci. 2020, 21, 4424. [Google Scholar] [CrossRef]
- Haß, U.; Herpich, C.; Norman, K. Anti-inflammatory diets and fatigue. Nutrients 2019, 11, 2315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Mello, C.; Ronaghan, N.; Zaheer, R.; Dicay, M.; Le, T.; MacNaughton, W.K.; Surrette, M.G.; Swain, M.G. Probiotics improve inflammation-associated sickness behavior by altering communication between the peripheral immune system and the brain. J. Neurosci. 2015, 35, 10821–10830. [Google Scholar] [CrossRef] [Green Version]
- Paraskevakos, G. Probiotics and the Brain. Agro Food Ind. Hi-Tech 2022, 33, 4–5. [Google Scholar]
- Leta, V.; Chaudhuril, K.R.; Milner, O.; Chung-Faye, G.; Metta, V.; Pariante, C.M.; Borsini, C.M.A. Neurogenic and anti-inflammatory effects of probiotics in Parkinson’s disease: A systematic review of preclinical and clinical evidence. Brain Behav. Immun. 2021, 98, 59–73. [Google Scholar] [CrossRef]
- Jangi, S.; Gandhi, R.; Cox, L.M.; Li, N.; von Glehn, F.; Yan, R.; Kivisäkk, P. Alterations of the human gut microbiome in multiple sclerosis. Nat. Commun. 2016, 7, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537. [Google Scholar] [CrossRef] [Green Version]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Mazmanian, S.K. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell 2016, 167, 1469–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacka, F.N.; O’Neil, A.; Opie, R.; Itsiopoulos, C.; Cotton, S.; Mohebbi, M.; Brazionis, L. A randomised controlled trial of dietary improvement for adults with major depression (the ‘SMILES’ trial). BMC Med. 2017, 15, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, B.; Yao, M.; Lv, L.; Ling, Z.; Li, L. The human microbiota in health and disease. Engineering 2017, 3, 71–82. [Google Scholar] [CrossRef]
- Dinan, T.G.; Cryan, J.F. Gut instincts: Microbiota as a key regulator of brain development, ageing and neurodegeneration. J. Physiol. 2017, 595, 489–503. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.N.; Yun, Y.; Ryu, S.; Chang, Y.; Kwon, M.J.; Cho, J.; Kim, H.L. The gut microbiota-derived metabolite trimethylamine N-oxide is elevated in schizophrenia. Brain Behav. Immun. 2020, 87, 516–523. [Google Scholar]
- Wang, H.H.; Li, Y.; Wu, R.R.; Zheng, W.; Ng, C.H.; Ungvari, G.S.; Xiang, Y.T. Effects of prebiotic supplementation on cognitive function and metabolic status in schizophrenia: A randomized, placebo-controlled trial. Eur. Arch. Psychiatry Clin. Neurosci. 2021, 271, 525–535. [Google Scholar]
- Rondanelli, M.; Gasparri, C.; Peroni, G.; Faliva, M.A.; Naso, M.; Perna, S.; Bazire, P.; Sajoux, I.; Maugeri, R.; Rigon, C. The potential roles of very low calorie, very low calorie ketogenic diets and very low carbohydrate diets on the gut microbiota composition. Front. Endocrinol. 2021, 12, 662591. [Google Scholar] [CrossRef] [PubMed]
- Paoli, A.; Rubini, A.; Volek, J.S.; Grimaldi, K.A. Beyond weight loss: A review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets. Eur. J. Clin. Nutr. 2019, 73, 789–796. [Google Scholar]
- Lim, J.-M.; Letchumanan, V.; Tan, L.T.-H.; Hong, K.-W.; Wong, S.-H.; Ab Mutalib, N.-S.; Lee, L.-H.; Law, J.W.-F. Ketogenic Diet: A Dietary Intervention via Gut Microbiome Modulation for the Treatment of Neurological and Nutritional Disorders (a Narrative Review). Nutrients 2022, 14, 3566. [Google Scholar] [CrossRef]
- Phillips, M.C.L.; Deprez, L.M.; Mortimer, G.M.N.; Murtagh, D.K.J.; McCoy, S.; Mylchreest, R.; Gilbertson, L.J.; Clark, K.M.; Simpson, P.V.; McManus, E.J.; et al. Randomized crossover trial of a modified ketogenic diet in Alzheimer’s disease. Alzheimer’s Res. Ther. 2021, 13, 51. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Yang, T.; Leak, R.K.; Chen, J.; Zhang, F. Preventive and protective roles of dietary Nrf2 activators against central nervous system diseases. CNS Neurol. Disord. Drug Targets 2017, 16, 326–338. [Google Scholar] [CrossRef] [Green Version]
- Armstrong, R.A. Risk factors for Alzheimer’s disease. Folia Neuropathol. 2019, 57, 87–105. [Google Scholar] [CrossRef] [Green Version]
- Luchsinger, J.A.; Mayeux, R. Cardiovascular risk factors and Alzheimer’s disease. Curr. Atheroscler. Rep. 2004, 6, 261–266. [Google Scholar] [CrossRef]
- Zigman, W.B.; Im, T.L. Alzheimer’s disease in Down syndrome: Neurobiology and risk. Ment. Retard. Dev. Disabil. Res. Rev. 2007, 13, 237–246. [Google Scholar] [CrossRef]
- Brinton, R.D. The healthy cell bias of estrogen action: Mitochondrial bioenergetics and neurological implications. Trends Neurosci. 2008, 31, 529–537. [Google Scholar] [CrossRef]
- Lai, F.; Mhatre, P.G.; Yang, Y.; Wang, M.C.; Schupf, N.; Rosas, H.D. Sex differences in risk of Alzheimer’s disease in adults with Down syndrome. Alzheimer’s Dement. Diagn. Assess. Dis. 2020, 12, e12084. [Google Scholar] [CrossRef]
- Papathanasiou, A.; MacDonal, A.; Amor, S. Multiple sclerosis and gender: The role of sex hormones in disease pathogenesis. Hormones 2019, 18, 219–229. [Google Scholar]
- Yao, J.; Hamilton, R.T.; Cadenas, E.; Brinton, R.D. Decline in mitochondrial bioenergetics and shift to ketogenic profile in brain during reproductive senescence. Biochim. Biophys. Acta 2010, 1800, 1121–1126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de la Rubia Ortí, J.E.; Fernández, D.; Platero, F.; García-Pardo, M.P. Can ketogenic diet improve Alzheimer’s disease? Association with anxiety, depression, and glutamate system. Front. Nutr. 2021, 8, 744398. [Google Scholar] [CrossRef]
- Ota, M.; Matsuo, J.; Ishida, I.; Takano, H.; Yoko, Y.; Hori, H.; Yoshida, S.; Ashida, K.; Nakamura, K.; Takahashi, T.; et al. Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in patients with mild-to-moderate Alzheimer’s disease. Neurosci. Lett. 2019, 690, 232–236. [Google Scholar] [CrossRef]
- Pike, C.J.; Carroll, J.C.; Rosario, E.R.; Barron, A.M. Protective actions of sex steroid hormones in Alzheimer’s disease. Front. Neuroendocrinol. 2009, 30, 239–258. [Google Scholar] [CrossRef] [Green Version]
- Fortier, M.; Castellano, C.A.; St-Pierre, V.; Myette-Côté, E.; Langlois, F.; Roy, M.; Morin, M.C.; Bocti, C.; Fulop, T.; Godin, J.P.; et al. A ketogenic drink improves cognition in mild cognitive impairment: Results of a 6-month RCT. Alzheimer’s Dement. 2021, 17, 543–552. [Google Scholar] [CrossRef] [PubMed]
- Loughrey, D.G.; Lavecchia, S.; Brennan, S.; Lawlor, B.A.; Kelly, M. The impact of the Mediterranean diet on the cognitive functioning of healthy older adults: A systematic review and meta-analysis. Adv. Nutr. 2017, 8, 571–586. [Google Scholar] [CrossRef]
- Katsiardanis, K.; Diamantaras, A.A.; Dessypris, N.; Michelakos, T.; Anastasiou, A.; Katsiardani, K.P.; Kanavidis, P.; Papadopoulos, F.C.; Stefanadis, C.; Panagiotakos, D.B.; et al. Cognitive impairment and dietary habits among elders: The Velestino study. J. Med. Food 2013, 16, 343–350. [Google Scholar] [CrossRef] [PubMed]
- Kesse-Guyot, E.; Andreeva, V.A.; Lassale, C.; Ferry, M.; Jeandel, C.; Hercberg, S.S. Mediterranean diet and cognitive function: A French study. Am. J. Clin. Nutr. 2013, 97, 369–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zbeida, M.; Goldsmith, R.; Shimony, T.; Vardi, H.; Naggan, L.; Shahar, D.R. Mediterranean diet and functional indicators among older adults in non-Mediterranean and Mediterranean countries. J. Nutr. Health Aging 2014, 18, 411–418. [Google Scholar] [CrossRef]
- Anastasiou, C.A.; Yannakoulia, M.; Kontogianni, M.D.; Kosmidis, M.H.; Mamalaki, E.; Dardiotis, E.; Hadjigeorgiou, G.; Sakka, P.; Tsapanou, A.; Lykou, A.; et al. Mediterranean Lifestyle in Relation to Cognitive Health: Results from the HELIAD Study. Nutrients 2018, 10, 1557. [Google Scholar] [CrossRef] [Green Version]
- Roberts, R.O.; Geda, Y.E.; Cerhan, J.R.; Knopman, D.S.; Cha, R.H.; Christianson, T.J.; Pankratz, V.S.; Ivnik, R.J.; Boeve, B.F.; O’Connor, H.M.; et al. Vegetables, unsaturated fats, moderate alcohol intake, and mild cognitive impairment. Dement. Geriatr. Cogn. Disord. 2010, 29, 413–423. [Google Scholar] [CrossRef] [Green Version]
- Hoscheidt, S.; Sanderlin, A.H.; Baker, L.D.; Jung, Y.; Lockhart, S.; Kellar, D.; Whitlow, C.T.; Hanson, A.J.; Friedman, S.; Register, T.; et al. Mediterranean and Western diet effects on Alzheimer’s disease biomarkers, cerebral perfusion, and cognition in mid-life: A randomized trial. Alzheimer’s Dement. 2022, 18, 457–468. [Google Scholar] [CrossRef]
- Galbete, C.; Toledo, E.; Toledo, J.B.; Bes-Rastrollo, M.; Buil-Cosiales, P.; Marti, A.; Guillén-Grima, F.; Martínez-González, M.A. Mediterranean diet and cognitive function: The SUN project. J. Nutr. Health Aging 2015, 19, 305–312. [Google Scholar] [CrossRef] [PubMed]
- Shinto, L.; Quinn, J.; Montine, T.; Dodge, H.H.; Woodward, W.; Baldauf-Wagner, S.; Waichunas, D.; Bumgarner, L.; Bourdette, D.; Silbert, L.; et al. A randomized placebo-controlled pilot trial of omega-3 fatty acids and alpha lipoic acid in Alzheimer’s disease. J. Alzheimer’s Dis. 2014, 38, 111–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, J.; Wen, S.; Zhou, J.; Ding, S. Association between malnutrition and hyperhomocysteine in Alzheimer’s disease patients and diet intervention of betaine. J. Clin. Lab. Anal. 2016, 31, e22090. [Google Scholar] [CrossRef] [PubMed]
- Sala-Vila, A.; Valls-Pedret, C.; Rajaram, S.; Coll-Padrós, N.; Cofán, M.; Serra-Mir, M.; Pérez-Heras, A.M.; Roth, I.; Freitas-Simoes, T.M.; Doménech, M.; et al. Effect of a 2-year diet intervention with walnuts on cognitive decline. The Walnuts And Healthy Aging (WAHA) study: A randomized controlled trial. Am. J. Clin. Nutr. 2020, 111, 590–600. [Google Scholar] [CrossRef]
- Halder, S.; Anand, U.; Nandy, S.; Oleksak, P.; Qusti, S.; Alshammari, E.M.; Batiha, G.E.-S.; Koshy, E.P.; Dey, A. Herbal drugs and natural bioactive products as potential therapeutics: A review on pro-cognitives and brain boosters perspectives. Saudi Pharm. J. 2021, 29, 879–907. [Google Scholar] [CrossRef]
- Gurley, B.J.; Swain, A.; Hubbard, M.A.; Williams, D.K.; Barone, G.; Hartsfield, F.; Tong, Y.; Carrier, D.J.; Cheboyina, S.; Battu, S.K. Clinical assessment of CYP2D6-mediated herb-drug interactions in humans: Effects of milk thistle, black cohosh, goldenseal, kava kava, St. John’s wort, and Echinacea. Mol. Nutr. Food Res. 2005, 49, 443–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dorsey, E.R.; Constantinescu, R.; Thompson, J.P.; Biglan, K.M.; Holloway, R.G.; Kieburtz, K.; Marshall, F.J.; Ravina, B.M.; Schifitto, G.; Siderowf, A.; et al. Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology 2007, 68, 384–386. [Google Scholar] [CrossRef]
- Nussbaum, R.L.; Ellis, C.E. Alzheimer’s disease and Parkinson’s disease. N. Engl. J. Med. 2003, 348, 1356–1364. [Google Scholar] [CrossRef] [Green Version]
- Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Prim. 2017, 3, 17013. [Google Scholar] [CrossRef]
- Koch, S.; Laabs, B.H.; Kasten, M.; Vollstedt, E.J.; Becktepe, J.; Brüggemann, N.; Caliebe, A. Validity and Prognostic Value of a Polygenic Risk Score for Parkinson’s Disease. Genes 2021, 12, 185. [Google Scholar] [CrossRef]
- Park, H.-A.; Ellis, A.C. Dietary Antioxidants and Parkinson’s disease: A systematic review and meta-analysis. Antioxidants. 2020, 50, 13–20. [Google Scholar]
- Chen, H.; Zhang, S.M.; Hernán, M.A.; Willett, W.C.; Ascherio, A. Diet and Parkinson’s disease: A potential role of dairy products in men. Ann. Neurol. 2002, 52, 793–801. [Google Scholar] [CrossRef]
- Tamtaji, O.R.; Taghizadeh, M.; Kakhaki, R.D.; Kouchaki, E.; Bahmani, F.; Borzabadi, S.; 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]
- Prasanna, P.H.P.; Grandison, A.S.; Charalampopoulos, D. Bifidobacteria in milk products: An overview of physiological and biochemical properties, exopolysaccharide production, selection criteria of milk products and health benefits. Food Res. Int. 2014, 55, 247–262. [Google Scholar] [CrossRef]
- Leite, A.M.D.O.; Miguel, M.A.L.; Peixoto, R.S.; Rosado, A.S.; Silva, J.T.; Paschoalin, V.M.F. Microbiological, technological and therapeutic properties of kefir: A natural probiotic beverage. Braz. J. Microbiol. 2013, 44, 341–349. [Google Scholar] [CrossRef] [PubMed]
- Yoon, J.H.; Kang, S.S.; Mheen, T.I.; Ahn, J.S.; Lee, H.J.; Kim, T.K.; Park, Y.H. Lactobacillus kimchii sp. nov.; a new species from kimchi. Int. J. Syst. Evol. Microbiol. 2000, 50, 1789–1795. [Google Scholar] [CrossRef] [Green Version]
- Markowiak, P.; Śliżewska, K. Effects of probiotics, prebiotics, and synbiotics on human health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef] [PubMed]
- Paknahad, Z.; Sheklabadi, E.; Derakhshan, Y.; Bagherniya, M.; Chitsaz, A. The effect of the Mediterranean diet on cognitive function in patients with Parkinson’s disease: A randomized clinical controlled trial. Complement. Ther. Med. 2020, 50, 102366. [Google Scholar] [CrossRef]
- VanItallie, T.B.; Nonas, C.; Di Rocco, A.; Boyar, K.; Hyams, K.; Heymsfield, S.B. Treatment of Parkinson disease with diet-induced hyperketonemia: A feasibility study. Neurology 2005, 64, 728–730. [Google Scholar] [CrossRef]
- Olanow, C.W.; Obeso, J.A.; Stocchi, F. Continuous dopamine-receptor treatment of Parkinson’s disease: Scientific rationale and clinical implications. Lancet Neurol. 2006, 13, 1175–1187. [Google Scholar] [CrossRef]
- Caccia, C. Pharmaco-EEG and brain mapping: Theory, applications and validation. J. Psychopharmacol. 2014, 12, 8–16. [Google Scholar]
- Kraeuter, A.K.; Phillips, R.; Sarnyai, Z. Ketogenic therapy in neurodegenerative and psychiatric disorders: From mice to men. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2020, 101, 109913. [Google Scholar] [CrossRef] [PubMed]
- Moghaddam, M.H.; Bayat, A.H.; Eskandari, N.; Abdollahifar, M.A.; Fotouhi, F.; Forouzannia, A.; Rafiei, R.; Hatari, S.; Seraj, A.; Shahidi, A.; et al. Elderberry diet ameliorates motor function and prevents oxidative stress-induced cell death in rat models of Huntington’s disease. Brain Res. 2021, 1762, 147444. [Google Scholar] [CrossRef] [PubMed]
- Piccini, P.; Lahiri, N.; Niccolini, F.; Su, P.; Wu, K.; Giannetti, P.; Scahill, R.I.; Turkheimer, F.E.; Tabrizi, S.J.; Piccini, P. Increased central microglial activation associated with peripheral cytokine levels in premanifest Huntington’s disease gene carriers. Increased central microglial activation associated with peripheral cytokine levels in premanifest Huntington’s disease gene carriers. Neurobiol. Dis. 2015, 83, 115–121. [Google Scholar] [CrossRef]
- Marder, K.; Zhao, H.; Eberly, S.; Tanner, C.M.; Oakes, D.; Shoulson, I. Dietary intake in adults at risk for Huntington disease: Analysis of PHAROS research participants. Neurology 2009, 73, 385–392. [Google Scholar] [CrossRef] [Green Version]
- Marder, K.; Gu, Y.; Eberly, S.; Tanner, C.M.; Scarmeas, N.; Oakes, D.; Shoulson, I. Huntington Study Group PHAROS Investigators. Relationship of Mediterranean diet and caloric intake to phenoconversion in Huntington disease. JAMA Neurol. 2013, 70, 1382–1388. [Google Scholar] [CrossRef] [Green Version]
- Rivadeneyra, J.; Cubo, E.; Gil, C.; Calvo, S.; Mariscal, N.; Martínez, A. Factors associated with Mediterranean diet adherence in Huntington’s disease. Clin. Nutr. ESPEN 2016, 12, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Sanberg, P.R.; Fibiger, H.C.; Mark, R.F. Body weight and dietary factors in Huntington’s disease patients compared with matched controls. Med. J. Aust. 1981, 1, 407–409. [Google Scholar] [CrossRef]
- Cubo, E.; Rivadeneyra, J.; Armesto, D.; Mariscal, N.; Martinez, A.; Camara, R.J. Relationship between nutritional status and the severity of Huntington’s disease. A Spanish multicenter dietary intake study. J. Huntingt. Dis. 2015, 4, 75–85. [Google Scholar] [CrossRef]
- Phillips, M.C.; Murtagh, D.K. Therapeutic potential of ketogenic diets for neurological disorders. Neurochem. Int. 2019, 129, 1–25. [Google Scholar]
- Ruskin, D.N.; Ross, J.L., Jr.; Kawamura, M.; Ruiz, T.L.; Geiger, J.D.; Masino, S.A. A ketogenic diet delays weight loss and does not impair working memory or motor function in the R6/2 1J mouse model of Huntington’s disease. Physiol. Behav. 2011, 103, 501–507. [Google Scholar] [CrossRef] [Green Version]
- Dobson, R.; Giovannoni, G. Multiple sclerosis—A review. Eur. J. Neurol. 2019, 26, 27–40. [Google Scholar] [CrossRef] [Green Version]
- McGinley, M.P.; Goldschmidt, C.H.; Rae-Grant, A.D. Diagnosis and treatment of multiple sclerosis: A review. JAMA 2021, 325, 765–779. [Google Scholar] [CrossRef]
- Esposito, S.; Bonavita, S.; Sparaco, M.; Gallo, A.; Tedeschi, G. The role of diet in multiple sclerosis: A review. Nutr. Neurosci. 2018, 21, 377–390. [Google Scholar] [CrossRef] [PubMed]
- De Angelis, F.; Plantone, D.; Chataway, J. Pharmacotherapy in secondary progressive multiple sclerosis: An overview. CNS Drugs 2018, 32, 499–526. [Google Scholar] [CrossRef] [PubMed]
- Bock, M.; Steffen, F.; Zipp, F.; Bittner, S. Impact of Dietary Intervention on Serum Neurofilament Light Chain in Multiple Sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2021, 9, e1102. [Google Scholar] [CrossRef] [PubMed]
- Katz Sand, I.; Benn, E.K.T.; Fabian, M.; Fitzgerald, K.C.; Digga, E.; Deshpande, R.; Miller, A.; Gallo, S.; Arab, L. Randomized-controlled trial of a modified Mediterranean dietary program for multiple sclerosis: A pilot study. Mult. Scler. Relat. Disord. 2019, 36, 101403. [Google Scholar] [CrossRef]
- Platero, J.L.; Cuerda-Ballester, M.; Ibáñez, V.; Sancho, D.; Lopez-Rodríguez, M.M.; Drehmer, E.; Ortí, J.E.R. The Impact of Coconut Oil and Epigallocatechin Gallate on the Levels of IL-6, Anxiety and Disability in Multiple Sclerosis Patients. Nutrients 2020, 12, 305. [Google Scholar] [CrossRef] [Green Version]
- Ascherio, A.; Munger, K.L. Epidemiology of multiple sclerosis: From risk factors to prevention—An update. Semin. Neurol. 2017, 37, 347–361. [Google Scholar] [CrossRef] [PubMed]
- Mauriz, E.; Laliena, A.; Vallejo, D.; Tuñón, M.J.; Rodríguez-López, J.M.; Rodríguez-Pérez, R.; García-Fernández, M.C. Effects of a low-fat diet with antioxidant supplementation on biochemical markers of multiple sclerosis long-term care residents. Nutr. Hosp. 2013, 28, 2229–2235. [Google Scholar] [PubMed]
- Institute for Health Metrics and Evaluation. New Global Burden of Disease Analyses Show Depression and Anxiety among the Top Causes of Health Loss Worldwide, and a Significant Increase Due to the COVID-19 Pandemic; Institute for Health Metrics and Evaluation: Seattle, WA, USA, 2020. [Google Scholar]
- Santomauro, D.F.; Herrera, A.M.M.; Shadid, J.; Zheng, P.; Ashbaugh, C.; Pigott, D.M.; Ferrari, A.J. Global prevalence and burden of depressive and anxiety disorders in 204 countries and territories in 2020 due to the COVID-19 pandemic. Lancet 2021, 398, 1700–1712. [Google Scholar] [CrossRef]
- Hölzel, L.; Härter, M.; Reese, C.; Kriston, L. Risk factors for chronic depression—A systematic review. J. Affect. Disord. 2011, 129, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Kheirouri, S.; Alizadeh, M. Dietary inflammatory potential and the risk of incident depression in adults: A systematic review. Adv. Nutr. 2019, 10, 9–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Zhou, Y.; Chen, K.; Jing, Y.; He, J.; Sun, H.; Hu, X. Dietary inflammatory index and depression: A meta-analysis. Public Health Nutr. 2019, 22, 654–660. [Google Scholar] [CrossRef]
- Shivappa, N.; Hebert, J.R.; Tehrani, A.N.; Bayzai, B.; Naja, F.; Rashidkhani, B. A pro-inflammatory diet is associated with an increased odds of depression symptoms among iranian female adolescents: A cross-sectional study. Front. Psychiatry 2018, 9, 400. [Google Scholar] [CrossRef] [Green Version]
- Shivappa, N.; Hébert, J.R.; Veronese, N.; Caruso, M.G.; Notarnicola, M.; Maggi, S.; Stubbs, B.; Firth, J.; Fornaro, M.; Solmi, M. The relationship between the dietary inflammatory index (DII®) and incident depressive symptoms: A longitudinal cohort study. J. Affect. Disord. 2018, 235, 39–44. [Google Scholar] [CrossRef] [Green Version]
- Marx, W.; Lane, M.; Hockey, M.; Aslam, H.; Berk, M.; Walder, K.; Borsini, A.; Firth, J.; Pariante, C.M.; Berding, K.; et al. Diet and depression: Exploring the biological mechanisms of action. Mol. Psychiatry 2021, 26, 134–150. [Google Scholar] [CrossRef]
- Godos, J.; Currenti, W.; Angelino, D.; Mena, P.; Castellano, S.; Caraci, F.; Galvano, F.; Del Rio, D.; Ferri, R.; Grosso, G. Diet and mental health: Review of the recent updates on molecular mechanisms. Antioxidants 2020, 9, 346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ljungberg, T.; Bondza, E.; Lethin, C. Evidence of the importance of dietary habits regarding depressive symptoms and depression. Int. J. Environ. Res. Public Health 2020, 17, 1616. [Google Scholar] [CrossRef] [Green Version]
- Park, C.; Brietzke, E.; Rosenblat, J.D.; Musial, N.; Zuckerman, H.; Ragguett, R.M.; Pan, Z.; Rong, C.; Fus, D.; McIntyre, R.S. Probiotics for the treatment of depressive symptoms: An anti-inflammatory mechanism? Brain Behav. Immun. 2018, 73, 115–124. [Google Scholar] [CrossRef]
- Grosso, G.; Pajak, A.; Marventano, S.; Castellano, S.; Galvano, F.; Bucolo, C.; Drago, F.; Caraci, F. Role of omega-3 fatty acids in the treatment of depressive disorders: A comprehensive meta-analysis of randomized clinical trials. PLoS ONE 2014, 9, e96905. [Google Scholar] [CrossRef] [Green Version]
- Lippi, G. Schizophrenia in a member of the family: Burden, expressed emotion and addressing the needs of the whole family. S. Afr. J. Psychiatr. 2016, 22, 922. [Google Scholar] [CrossRef] [Green Version]
- Jacka, F.N.; Kremer, P.J.; Berk, M.; de Silva-Sanigorski, A.M.; Moodie, M.; Leslie, E.R.; Pasco, J.A.; Swinburn, B.A. A prospective study of diet quality and mental health in adolescents. PLoS ONE 2017, 12, e0186090. [Google Scholar] [CrossRef]
- Lassale, C.; Batty, G.; Baghdadli, A.; Jacka, F.; Sanchez-Villegas, A.; Kivimaki, M.; Akbaraly, T. Healthy dietary indices and risk of depressive outcomes: A systematic review and meta-analysis of observational studies. Mol. Psychiatry 2019, 24, 965–986. [Google Scholar] [CrossRef] [Green Version]
- Cha, H.Y.; Yang, S. Anti-inflammatory diets and schizophrenia. Clin. Nutr. Res. 2020, 9, 241. [Google Scholar] [CrossRef] [PubMed]
- McCutcheon, R.A.; Marques, T.; Howes, O. Schizophrenia—An overview. JAMA Psychiatry 2020, 77, 201–210. [Google Scholar] [CrossRef]
- Stępnicki, P.; Kondej, M.; Kaczor, A. Current concepts and treatments of schizophrenia. Molecules 2018, 23, 2087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jablensky, A. The diagnostic concept of schizophrenia: Its history, evolution, and future prospects. Dialogues Clin. Neurosci. 2022, 12, 271–287. [Google Scholar] [CrossRef]
- Pandurangi, A.K.; Buckley, P.F. Inflammation, antipsychotic drugs, and evidence for effectiveness of anti-inflammatory agents in schizophrenia. Curr. Top. Behav. Neurosci. 2019, 44, 227–244. [Google Scholar] [CrossRef]
- Fond, G.; Lançon, C.; Korchia, T.; Auquier, P.; Boyer, L. The role of inflammation in the treatment of schizophrenia. Front. Psychiatry 2020, 11, 160. [Google Scholar] [CrossRef] [Green Version]
- Firth, J.; Veronese, N.; Cotter, J.; Shivappa, N.; Hebert, J.R.; Ee, C.; Smith, L.; Stubbs, B.; Jackson, S.E.; Sarris, J. What is the role of dietary inflammation in severe mental illness? A review of observational and experimental findings. Front. Psychiatry 2019, 10, 350. [Google Scholar] [CrossRef] [Green Version]
- Jahrami, H.; Faris, M.E.A.I.; Ghazzawi, H.A.; Saif, Z.; Habib, L.; Shivappa, N.; Hébert, J.R. Increased dietary inflammatory index is associated with schizophrenia: Results of a case–control study from Bahrain. Nutrients 2019, 11, 1867. [Google Scholar] [CrossRef] [Green Version]
- Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [Green Version]
- Guo, L.; Xiao, P.; Zhang, X.; Yang, Y.; Yang, M.; Wang, T.; Lu, H.; Tian, H.; Wang, H.; Liu, J. Inulin ameliorates schizophrenia via modulation of the gut microbiota and anti-inflammation in mice. Food Funct. 2021, 12, 1156–1175. [Google Scholar] [CrossRef] [PubMed]
- Marsh, W.K.; Penny, J.L.; Rothschild, A.J. Vitamin D supplementation in bipolar depression: A double blind placebo controlled trial. J. Psychiatr. Res. 2017, 95, 48–53. [Google Scholar] [CrossRef]
- Mitchell, E.S.; Conus, N.; Kaput, J. B vitamin polymorphisms and behavior: Evidence of associations with neurodevelopment, depression, schizophrenia, bipolar disorder and cognitive decline. Neurosci. Biobehav. Rev. 2014, 47, 307–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cereda, G.; Enrico, P.; Ciappolino, V.; Delvecchio, G.; Brambilla, P. The role of vitamin D in bipolar disorder: Epidemiology and influence on disease activity. J. Affect. Disord. 2021, 278, 209–217. [Google Scholar] [CrossRef] [PubMed]
- Arroll, M.A.; Wilder, L.; Neil, J. Nutritional interventions for the adjunctive treatment of schizophrenia: A brief review. Nutr. J. 2014, 13, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Joseph, J.; Depp, C.; Shih, P.-A.B.; Cadenhead, K.S.; Schmid-Schönbein, G. Modified Mediterranean Diet for Enrichment of Short Chain Fatty Acids: Potential Adjunctive Therapeutic to Target Immune and Metabolic Dysfunction in Schizophrenia? Front. Neurosci. 2017, 11, 155. [Google Scholar] [CrossRef] [Green Version]
- Vassilopoulou, E.; Efthymiou, D.; Tsironis, V.; Athanassis, P.; Chatzioannidis, S.; Kesoglou, T.; Severin, A.V.; Bozikas, V.P. The benefits of the Mediterranean diet in first episode psychosis patients taking antipsychotics. Toxicol. Rep. 2022, 9, 120–125. [Google Scholar] [CrossRef] [PubMed]
- Mitra, S.; Natarajan, R.; Ziedonis, D.; Fan, X. Antioxidant and anti-inflammatory nutrient status, supplementation, and mechanisms in patients with schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2017, 78, 1–11. [Google Scholar] [CrossRef]
- Sommer, I.E.; van Westrhenen, R.; Begemann, M.J.; de Witte, L.D.; Leucht, S.; Kahn, R.S. Efficacy of anti-inflammatory agents to improve symptoms in patients with schizophrenia: An update. Schizophr. Bull. 2014, 40, 181–191. [Google Scholar] [CrossRef] [Green Version]
- Anderson, I.M.; Haddad, P.M.; Scott, J. Bipolar disorder. Br. Med. J. 2012, 345, e8508. [Google Scholar] [CrossRef] [PubMed]
- Carvalho, A.F.; Firth, J.; Vieta, E. Bipolar disorder. N. Engl. J. Med. 2020, 383, 58–66. [Google Scholar] [CrossRef] [PubMed]
- Grande, I.; Berk, M.; Birmaher, B.; Vieta, E. Bipolar disorder. Lancet 2016, 387, 1561–1572. [Google Scholar] [CrossRef] [PubMed]
- Rosenblat, J.D. Targeting the immune system in the treatment of bipolar disorder. Psychopharmacology 2019, 236, 2909–2921. [Google Scholar] [CrossRef]
- Rosenblat, J.D.; Kakar, R.; Berk, M.; Kessing, L.V.; Vinberg, M.; Baune, B.T.; Mansur, R.B.; Brietzke, E.; McIntyre, B.I.G.R.S. Anti-inflammatory agents in the treatment of bipolar depression: A systematic review and meta-analysis. Bipolar Disord. 2016, 18, 89–101. [Google Scholar] [CrossRef]
- Sublette, M.E.; Cheung, S.; Lieberman, E.; Hu, S.; Mann, J.J.; Uhlemann, A.C.; Miller, J.M. Bipolar disorder and the gut microbiome: A systematic review. Bipolar Disord. 2021, 23, 544–564. [Google Scholar] [CrossRef] [PubMed]
- Grandjean, E.L.; van Zonneveld, S.M.; Sommer, I.E.; Haarman, B.C. Anti-inflammatory dietary patterns to treat bipolar disorder? J. Affect. Disord. 2022, 311, 254–255. [Google Scholar] [CrossRef] [PubMed]
- Madireddy, S.; Madireddy, S. Therapeutic Interventions to Mitigate Mitochondrial Dysfunction and Oxidative Stress–Induced Damage in Patients with Bipolar Disorder. Int. J. Mol. Sci. 2022, 23, 1844. [Google Scholar] [CrossRef]
- Parletta, N.; Milte, C.M.; Meyer, B.J. Nutritional modulation of cognitive function and mental health. J. Nutr. Biochem. 2013, 24, 725–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacka, F.N. Nutritional psychiatry: Where to next? eBioMedicine 2017, 17, 24–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirschfeld, R.M. Differential diagnosis of bipolar disorder and major depressive disorder. J. Affect. Disord. 2014, 169, S12–S16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Łojko, D.; Stelmach, M.; Suwalska, A. Is diet important in bipolar disorder. Psychiatry Pol. 2018, 52, 783–795. [Google Scholar] [CrossRef] [PubMed]
Study | Study Design | Participants | Intervention | Neurological Disease | Duration | Outcome Measures | Results |
---|---|---|---|---|---|---|---|
Mousavi-Shirazi-Fard, Z. et al., (2021) [40] | Randomized controlled trial | 100 patients with multiple sclerosis (MS) | Anti-inflammatory diet vs. usual diet | MS | 12 weeks | Serum levels of inflammatory markers (CRP, IL-4, IL-17) Assessment of fatigue and quality of life (MFIS and MSQoL-54) | The anti-inflammatory diet group had significantly increased IL-4 and improvement in MFIS as well as of MSQoL-54 compared to the usual diet group |
Akbari, M. et al., (2008) [41] | Randomized controlled trial | 80 patients with Alzheimer’s disease (AD) | Ketogenic diet vs. usual diet | AD | 12 weeks | Cognitive function (Mini-Mental State Examination), serum levels of inflammatory markers (IL-6, TNF-alpha) | The ketogenic diet group had significant improvements in cognitive function and reductions in inflammatory markers compared to the usual diet group |
Phillips, M. C. L. et al., (2018) [42] | Randomized controlled trial | 44 patients with Parkinson’s disease (PD) | Ketogenic diet vs. usual diet | PD | 8 weeks | Serum levels of inflammatory markers motor and nonmotor symptoms | The ketogenic group showed improvements in nonmotor symptoms |
Singh, B. et al., (2014) [43] | Randomized controlled trial | patients with mild cognitive impairment (MCI) or AD | Mediterranean diet vs. control diet | MCI or AD | 6 months | Serum levels of inflammatory markers cognitive function | The Mediterranean diet is associated with a reduced risk of developing MCI and AD, and a reduced risk of progressing from MCI to AD. |
Paknahad, Z. et al., (2020) [44] | Randomized controlled trial | 80 patients with PD | Mediterranean diet vs. control diet (Iranian traditional diet) | PD | 3 months | Serum levels of Total Antioxidant Capacity (TAC) and motor function (Unified Parkinson’s Disease Rating Scale) | The Mediterranean diet had a beneficial effect on TAC and on the severity of the disease |
Zhang, Y. et al., (2014) [45] | Prospective cohort study | participants | Dietary patterns and risk of PD | N/A | 10 years | Incident cases of PD | Higher adherence to a prudent dietary pattern was associated with a lower risk of PD |
Gao, X. et al., (2007) [46] | Prospective cohort study | participants | Dietary patterns and risk of PD | N/A | 16 years | Alternate Healthy Eating Index (AHEI) and the alternate Mediterranean Diet Score (aMed) | A higher AHEI or aMED score was associated with a reduced risk of PD the Western pattern increased the risk of PD |
Lai, J.S., et al., (2014) [47] | Meta-analysis | participants | Dietary patterns and risk of depression | moderate to severe depression | N/A | Depression severity (BDI-II), anxiety (HADS-A), inflammatory markers | A diet high in fruits, vegetables, fish, and whole grains may be associated with reductions in depression severity and anxiety scores, as well as lower levels of inflammatory markers. |
El-Mallakh, R. S. et al., (2001) [48] | Meta-analysis | patients with depression | Ketogenic diet vs. control diet | depression | N/A | Depression severity (HDRS), anxiety (HAMA), inflammatory markers (CRP, IL-6) | The ketogenic diet group had significant reductions in depression severity and anxiety scores, as well as lower levels of inflammatory markers. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Kurowska, A.; Ziemichód, W.; Herbet, M.; Piątkowska-Chmiel, I. The Role of Diet as a Modulator of the Inflammatory Process in the Neurological Diseases. Nutrients 2023, 15, 1436. https://doi.org/10.3390/nu15061436
Kurowska A, Ziemichód W, Herbet M, Piątkowska-Chmiel I. The Role of Diet as a Modulator of the Inflammatory Process in the Neurological Diseases. Nutrients. 2023; 15(6):1436. https://doi.org/10.3390/nu15061436
Chicago/Turabian StyleKurowska, Antonina, Wojciech Ziemichód, Mariola Herbet, and Iwona Piątkowska-Chmiel. 2023. "The Role of Diet as a Modulator of the Inflammatory Process in the Neurological Diseases" Nutrients 15, no. 6: 1436. https://doi.org/10.3390/nu15061436
APA StyleKurowska, A., Ziemichód, W., Herbet, M., & Piątkowska-Chmiel, I. (2023). The Role of Diet as a Modulator of the Inflammatory Process in the Neurological Diseases. Nutrients, 15(6), 1436. https://doi.org/10.3390/nu15061436