The Potential Effects of Probiotics and ω-3 Fatty Acids on Chronic Low-Grade Inflammation
Abstract
:1. Introduction
2. Modulating the Gastrointestinal Microbiota: The Effect of Environmental Exposure and Dietary Supplementation
3. Human Health and the Gut–Brain Axis
4. Microbiota and Immune Modulation by Probiotics, Prebiotics, and Ω-3 Fatty Acids
4.1. Lactobacillus, Bifidobacterium, and Chronic Low-Grade Inflammation
4.2. Lactobacillus
4.3. Bifidobacterium
4.4. Lactobacillus and Bifidobacterium in Combination
5. Fatty Acids and Chronic Low-Grade Inflammation
6. Synergism between ω-3 Fatty Acids and Probiotics
7. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Immunity in Brief. Available online: https://lpi.oregonstate.edu/mic/health-disease/immunity-in-brief (accessed on 16 April 2020).
- Calder, P.C.; Bosco, N.; Bourdet-Sicard, R.; Capuron, L.; Delzenne, N.; Dore, J.; Franceschi, C.; Lehtinen, M.J.; Recker, T.; Salvioli, S.; et al. Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res. Rev. 2017, 40, 95–119. [Google Scholar] [CrossRef] [PubMed]
- Pahwa, R.; Goyal, A.; Bansal, P.; Jialal, I. Chronic Inflammation; StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
- Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [PubMed] [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] [PubMed] [Green Version]
- Liang, D.; Leung, R.K.; Guan, W.; Au, W.W. Involvement of gut microbiome in human health and disease: Brief overview, knowledge gaps and research opportunities. Gut Pathog. 2018, 10, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dai, D.; Wang, T.; Wu, S.; Gao, N.L.; Chen, W.H. Metabolic dependencies underlie interaction patterns of gut microbiota during enteropathogenesis. Front. Microbiol. 2019, 10, 1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sommer, F.; Anderson, J.M.; Bharti, R.; Raes, J.; Rosenstiel, P. The resilience of the intestinal microbiota influences health and disease. Nat. Rev. Microbiol. 2017, 15, 630–638. [Google Scholar] [CrossRef] [PubMed]
- Telle-Hansen, V.H.; Holven, K.B.; Ulven, S.M. Impact of a healthy dietary pattern on gut microbiota and systemic inflammation in humans. Nutrients 2018, 10, 1783. [Google Scholar] [CrossRef] [Green Version]
- Torres-Fuentes, C.; Schellekens, H.; Dinan, T.G.; Cryan, J.F. The microbiota-gut-brain axis in obesity. Lancet Gastroenterol. Hepatol. 2017, 2, 747–756. [Google Scholar] [CrossRef]
- Bellenger, J.; Bellenger, S.; Escoula, Q.; Bidu, C.; Narce, M. N-3 polyunsaturated fatty acids: An innovative strategy against obesity and related metabolic disorders, intestinal alteration and gut microbiota dysbiosis. Biochimie 2019, 159, 66–71. [Google Scholar] [CrossRef]
- Konig, J.; Wells, J.; Cani, P.D.; Garcia-Rodenas, C.L.; MacDonald, T.; Mercenier, A.; Whyte, J.; Troost, F.; Brummer, R.J. Human intestinal barrier function in health and disease. Clin. Transl. Gastroenterol. 2016, 7, e196. [Google Scholar] [CrossRef]
- 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] [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] [Green Version]
- Agusti, A.; Garcia-Pardo, M.P.; Lopez-Almela, I.; Campillo, I.; Maes, M.; Romani-Perez, M.; Sanz, Y. Interplay between the gut-brain axis, obesity and cognitive function. Front. Neurosci. 2018, 12, 155. [Google Scholar] [CrossRef] [PubMed]
- Dumitrescu, L.; Popescu-Olaru, I.; Cozma, L.; Tulba, 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]
- Hart, A.; Kamm, M.A. Review article: Mechanisms of initiation and perpetuation of gut inflammation by stress. Aliment. Pharm. 2002, 16, 2017–2028. [Google Scholar] [CrossRef]
- Moro-Garcia, M.A.; Alonso-Arias, R.; Baltadjieva, M.; Fernandez Benitez, C.; Fernandez Barrial, M.A.; Diaz Ruisanchez, E.; Alonso Santos, R.; Alvarez Sanchez, M.; Saavedra Mijan, J.; Lopez-Larrea, C. Oral supplementation with Lactobacillus delbrueckii subsp. bulgaricus 8481 enhances systemic immunity in elderly subjects. Age 2013, 35, 1311–1326. [Google Scholar] [CrossRef]
- Wang, Y.; Huang, F. N-3 polyunsaturated fatty acids and inflammation in obesity: Local effect and systemic benefit. Biomed. Res. Int. 2015, 2015, 581469. [Google Scholar] [CrossRef] [Green Version]
- Turnbaugh, P.J.; Gordon, J.I. The core gut microbiome, energy balance and obesity. J. Physiol. 2009, 587, 4153–4158. [Google Scholar] [CrossRef]
- Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T.; et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef] [Green Version]
- Bessac, A.; Cani, P.D.; Meunier, E.; Dietrich, G.; Knauf, C. Inflammation and gut-brain axis during type 2 diabetes: Focus on the crosstalk between intestinal immune cells and enteric nervous system. Front. Neurosci. 2018, 12, 725. [Google Scholar] [CrossRef] [PubMed]
- Koloski, N.A.; Jones, M.; Talley, N.J. Evidence that independent gut-to-brain and brain-to-gut pathways operate in the irritable bowel syndrome and functional dyspepsia: A 1-year population-based prospective study. Aliment. Pharm. 2016, 44, 592–600. [Google Scholar] [CrossRef] [PubMed]
- Costantini, L.; Molinari, R.; Farinon, B.; Merendino, N. Impact of omega-3 fatty acids on the gut microbiota. Int. J. Mol. Sci. 2017, 18, 2645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ferreira, C.M.; Vieira, A.T.; Vinolo, M.A.; Oliveira, F.A.; Curi, R.; Martins Fdos, S. The central role of the gut microbiota in chronic inflammatory diseases. J. Immunol. Res. 2014, 2014, 689492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [Green Version]
- Jakobsson, H.E.; Abrahamsson, T.R.; Jenmalm, M.C.; Harris, K.; Quince, C.; Jernberg, C.; Bjorksten, B.; Engstrand, L.; Andersson, A.F. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut 2014, 63, 559–566. [Google Scholar] [CrossRef] [Green Version]
- Rodriguez, 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]
- Toscano, M.; De Grandi, R.; Grossi, E.; Drago, L. Role of the human breast milk-associated microbiota on the newborns’ immune system: A mini review. Front. Microbiol. 2017, 8, 2100. [Google Scholar] [CrossRef]
- Abrahamsson, T.R.; Jakobsson, H.E.; Andersson, A.F.; Bjorksten, B.; Engstrand, L.; Jenmalm, M.C. Low diversity of the gut microbiota in infants with atopic eczema. J. Allergy Clin. Immunol. 2012, 129, 434–440. [Google Scholar] [CrossRef] [Green Version]
- Jenmalm, M.C. The mother-offspring dyad: Microbial transmission, immune interactions and allergy development. J. Intern. Med. 2017, 282, 484–495. [Google Scholar] [CrossRef] [Green Version]
- West, C.E.; Renz, H.; Jenmalm, M.C.; Kozyrskyj, A.L.; Allen, K.J.; Vuillermin, P.; Prescott, S.L. The gut microbiota and inflammatory noncommunicable diseases: Associations and potentials for gut microbiota therapies. J. Allergy Clin. Immunol. 2015, 135, 3–13. [Google Scholar] [CrossRef] [Green Version]
- Salminen, S.; Gibson, G.R.; McCartney, A.L.; Isolauri, E. Influence of mode of delivery on gut microbiota composition in seven year old children. Gut 2004, 53, 1388–1389. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Zhang, M.; Wang, S.; Han, R.; Cao, Y.; Hua, W.; Mao, Y.; Zhang, X.; Pang, X.; Wei, C.; et al. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J. 2010, 4, 232–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bron, P.A.; Kleerebezem, M.; Brummer, R.J.; Cani, P.D.; Mercenier, A.; MacDonald, T.T.; Garcia-Rodenas, C.L.; Wells, J.M. Can probiotics modulate human disease by impacting intestinal barrier function? Br. J. Nutr. 2017, 117, 93–107. [Google Scholar] [CrossRef] [PubMed]
- Flach, J.; van der Waal, M.B.; Kardinaal, A.F.M.; Schloesser, J.; Ruijschop, R.M.A.J.; Claassen, E. Probiotic research priorities for the healthy adult population: A review on the health benefits of Lactobacillus rhamnosus GG and Bifidobacterium animalis subspecies lactic BB-12. Cogent Food Agric. 2018, 4, 1452839. [Google Scholar] [CrossRef]
- Lyte, M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: Microbial endocrinology in the design and use of probiotics. Bioessays 2011, 33, 574–581. [Google Scholar] [CrossRef]
- Rosas-Ballina, M.; Tracey, K.J. Cholinergic control of inflammation. J. Intern. Med. 2009, 265, 663–679. [Google Scholar] [CrossRef]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Backhed, F. From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [Green Version]
- Buhner, S.; Schemann, M. Mast cell-nerve axis with a focus on the human gut. Biochim. Biophys. Acta 2012, 1822, 85–92. [Google Scholar] [CrossRef] [Green Version]
- Farhadi, A.; Fields, J.Z.; Keshavarzian, A. Mucosal mast cells are pivotal elements in inflammatory bowel disease that connect the dots: Stress, intestinal hyperpermeability and inflammation. World J. Gastroenterol. 2007, 13, 3027–3030. [Google Scholar] [CrossRef]
- Giau, V.V.; Wu, S.Y.; Jamerlan, A.; An, S.S.A.; Kim, S.Y.; Hulme, J. Gut microbiota and their neuroinflammatory implications in Alzheimer’s disease. Nutrients 2018, 10, 1765. [Google Scholar] [CrossRef] [Green Version]
- Adamczyk-Sowa, M.; Medrek, A.; Madej, P.; Michlicka, W.; Dobrakowski, P. Does the gut microbiota influence immunity and inflammation in multiple sclerosis pathophysiology? J. Immunol. Res. 2017, 2017, 7904821. [Google Scholar] [CrossRef] [PubMed]
- Boirivant, M.; Strober, W. The mechanism of action of probiotics. Curr. Opin. Gastroenterol. 2007, 23, 679–692. [Google Scholar] [CrossRef] [PubMed]
- Shin, W.; Kim, H.J. Intestinal barrier dysfunction orchestrates the onset of inflammatory host-microbiome cross-talk in a human gut inflammation-on-a-chip. Proc. Natl. Acad. Sci. USA 2018, 115, E10539–E10547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Slyepchenko, A.; Maes, M.; Jacka, F.N.; Kohler, C.A.; Barichello, T.; McIntyre, R.S.; Berk, M.; Grande, I.; Foster, J.A.; Vieta, E.; et al. Gut microbiota, bacterial translocation, and interactions with diet: Pathophysiological links between major depressive disorder and non-communicable medical comorbidities. Psychother. Psychosom. 2017, 86, 31–46. [Google Scholar] [CrossRef] [Green Version]
- Taleb, S. Tryptophan dietary impacts gut barrier and metabolic diseases. Front. Immunol. 2019, 10, 2113. [Google Scholar] [CrossRef] [PubMed]
- O’Mahony, S.M.; Clarke, G.; Borre, Y.E.; Dinan, T.G.; Cryan, J.F. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef] [PubMed]
- Waclawikova, B.; El Aidy, S. Role of microbiota and tryptophan metabolites in the remote effect of intestinal inflammation on brain and depression. Pharmaceuticals 2018, 11, 63. [Google Scholar] [CrossRef] [Green Version]
- Wallace, C.J.K.; Milev, R. The effects of probiotics on depressive symptoms in humans: A systematic review. Ann. Gen. Psychiatry 2017, 16, 14. [Google Scholar] [CrossRef] [Green Version]
- Tillisch, K.; Labus, J.; Kilpatrick, L.; Jiang, Z.; Stains, J.; Ebrat, B.; Guyonnet, D.; Legrain-Raspaud, S.; Trotin, B.; Naliboff, B.; et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013, 144, 1394–1401. [Google Scholar] [CrossRef] [Green Version]
- Akkasheh, G.; Kashani-Poor, Z.; Tajabadi-Ebrahimi, M.; Jafari, P.; Akbari, H.; Taghizadeh, M.; Memarzadeh, M.R.; Asemi, Z.; Esmaillzadeh, A. Clinical and metabolic response to probiotic administration in patients with major depressive disorder: A randomized, double-blind, placebo-controlled trial. Nutrition 2016, 32, 315–320. [Google Scholar] [CrossRef]
- Micronutrient Information Center: Fiber. Available online: https://lpi.oregonstate.edu/mic/other-nutrients/fiber (accessed on 16 April 2020).
- Neis, E.P.; van Eijk, H.M.; Lenaerts, K.; Olde Damink, S.W.; Blaak, E.E.; Dejong, C.H.; Rensen, S.S. Distal versus proximal intestinal short-chain fatty acid release in man. Gut 2019, 68, 764–765. [Google Scholar] [CrossRef] [PubMed]
- Li, K.; Huang, T.; Zheng, J.; Wu, K.; Li, D. Effect of marine-derived n-3 polyunsaturated fatty acids on C-reactive protein, interleukin 6 and tumor necrosis factor alpha: A meta-analysis. PLoS ONE 2014, 9, e88103. [Google Scholar]
- Paerregaard, S.I.; Agerholm, M.; Serup, A.K.; Ma, T.; Kiens, B.; Madsen, L.; Kristiansen, K.; Jensen, B.A. FFAR4 (GPR120) signaling is not required for anti-inflammatory and insulin-sensitizing effects of omega-3 fatty acids. Mediat. Inflamm. 2016, 2016, 1536047. [Google Scholar] [CrossRef] [Green Version]
- Dullemeijer, C.; Zock, P.L.; Coronel, R.; Den Ruijter, H.M.; Katan, M.B.; Brummer, R.J.; Kok, F.J.; Beekman, J.; Brouwer, I.A. Differences in fatty acid composition between cerebral brain lobes in juvenile pigs after fish oil feeding. Br. J. Nutr. 2008, 100, 794–800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dullemeijer, C.; Durga, J.; Brouwer, I.A.; van de Rest, O.; Kok, F.J.; Brummer, R.J.; van Boxtel, M.P.; Verhoef, P. n-3 fatty acid proportions in plasma and cognitive performance in older adults. Am. J. Clin. Nutr. 2007, 86, 1479–1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Custodero, C.; Mankowski, R.T.; Lee, S.A.; Chen, Z.; Wu, S.; Manini, T.M.; Hincapie Echeverri, J.; Sabba, C.; Beavers, D.P.; Cauley, J.A.; et al. Evidence-based nutritional and pharmacological interventions targeting chronic low-grade inflammation in middle-age and older adults: A systematic review and meta-analysis. Ageing Res. Rev. 2018, 46, 42–59. [Google Scholar] [CrossRef] [PubMed]
- Presti, I.; D’Orazio, G.; Labra, M.; La Ferla, B.; Mezzasalma, V.; Bizzaro, G.; Giardina, S.; Michelotti, A.; Tursi, F.; Vassallo, M.; et al. Evaluation of the probiotic properties of new Lactobacillus and Bifidobacterium strains and their in vitro effect. Appl. Microbiol. Biotechnol. 2015, 99, 5613–5626. [Google Scholar] [CrossRef]
- George, F.; Daniel, C.; Thomas, M.; Singer, E.; Guilbaud, A.; Tessier, F.J.; Revol-Junelles, A.M.; Borges, F.; Foligne, B. Occurrence and dynamism of lactic acid bacteria in distinct ecological niches: A multifaceted functional health perspective. Front. Microbiol. 2018, 9, 2899. [Google Scholar] [CrossRef] [Green Version]
- Ouwehand, A.C.; Rasinkangas, P. Digestive health benefits of HOWARU restore probiotic combination and its individual strains. Agro Food Ind. Hi Tech 2017, 28, 20–23. [Google Scholar]
- Collado, M.C.; Meriluoto, J.; Salminen, S. Role of commercial probiotic strains against human pathogen adhesion to intestinal mucus. Lett. Appl. Microbiol. 2007, 45, 454–460. [Google Scholar] [CrossRef]
- Kekkonen, R.A.; Lummela, N.; Karjalainen, H.; Latvala, S.; Tynkkynen, S.; Järvenpää, S.; Kautiainen, H.; Julkunen, I.; Vapaatalo, H.; Korpela, R. Probiotic intervention has strain-specific anti-inflammatory effects in healthy adults. World J. Gastroenterol. 2008, 14, 2029–2036. [Google Scholar] [CrossRef] [PubMed]
- Schultz, M.; Linde, H.J.; Lehn, N.; Zimmermann, K.; Grossmann, J.; Falk, W.; Schölmerich, J. Immunomodulatory consequences of oral administration of Lactobacillus rhamnosus strain GG in healthy volunteers. J. Dairy Res. 2003, 70, 165–173. [Google Scholar] [CrossRef] [PubMed]
- Lavasani, S.; Dzhambazov, B.; Nouri, M.; Fak, F.; Buske, S.; Molin, G.; Thorlacius, H.; Alenfall, J.; Jeppsson, B.; Westrom, B. A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS ONE 2010, 5, e9009. [Google Scholar] [CrossRef] [PubMed]
- Hwang, Y.H.; Park, S.; Paik, J.W.; Chae, S.W.; Kim, D.H.; Jeong, D.G.; Ha, E.; Kim, M.; Hong, G.; Park, S.H.; et al. Efficacy and safety of Lactobacillus plantarum C29-fermented soybean (DW2009) in individuals with mild cognitive impairment: A 12-week, multi-center, randomized, double-blind, placebo-controlled clinical trial. Nutrients 2019, 11, 305. [Google Scholar] [CrossRef] [Green Version]
- West, N.P.; Horn, P.L.; Pyne, D.B.; Gebski, V.J.; Lahtinen, S.J.; Fricker, P.A.; Cripps, A.W. Probiotic supplementation for respiratory and gastrointestinal illness symptoms in healthy physically active individuals. Clin. Nutr. 2014, 33, 581–587. [Google Scholar] [CrossRef] [Green Version]
- Rodes, L.; Khan, A.; Paul, A.; Coussa-Charley, M.; Marinescu, D.; Tomaro-Duchesneau, C.; Shao, W.; Kahouli, I.; Prakash, S. Effect of probiotics Lactobacillus and Bifidobacterium on gut-derived lipopolysaccharides and inflammatory cytokines: An in vitro study using a human colonic microbiota model. J. Microbiol. Biotechnol. 2013, 23, 518–526. [Google Scholar] [CrossRef] [Green Version]
- O’Mahony, C.; Scully, P.; O’Mahony, D.; Murphy, S.; O’Brien, F.; Lyons, A.; Sherlock, G.; MacSharry, J.; Kiely, B.; Shanahan, F.; et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog. 2008, 4, e1000112. [Google Scholar]
- Rajilic-Stojanovic, M.; Biagi, E.; Heilig, H.G.; Kajander, K.; Kekkonen, R.A.; Tims, S.; de Vos, W.M. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology 2011, 141, 1792–1801. [Google Scholar] [CrossRef]
- Roessler, A.; Friedrich, U.; Vogelsang, H.; Bauer, A.; Kaatz, M.; Hipler, U.C.; Schmidt, I.; Jahreis, G. The immune system in healthy adults and patients with atopic dermatitis seems to be affected differently by a probiotic intervention. Clin. Exp. Allergy 2008, 38, 93–102. [Google Scholar]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
- Ringel-Kulka, T.; Palsson, O.S.; Maier, D.; Carroll, I.; Galanko, J.A.; Leyer, G.; Ringel, Y. Probiotic bacteria Lactobacillus acidophilus NCFM and Bifidobacterium lactis Bi-07 versus placebo for the symptoms of bloating in patients with functional bowel disorders: A double-blind study. J. Clin. Gastroenterol. 2011, 45, 518–525. [Google Scholar] [CrossRef] [PubMed]
- Leyer, G.J.; Li, S.; Mubasher, M.E.; Reifer, C.; Ouwehand, A.C. Probiotic effects on cold and influenza-like symptom incidence and duration in children. Pediatrics 2009, 124, e172–e179. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Kuda, O.; Brezinova, M.; Rombaldova, M.; Slavikova, B.; Posta, M.; Beier, P.; Janovska, P.; Veleba, J.; Kopecky, J., Jr.; Kudova, E.; et al. Docosahexaenoic acid-derived fatty acid esters of hydroxy fatty acids (FAHFAs) with anti-inflammatory properties. Diabetes 2016, 65, 2580–2590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacobo-Cejudo, M.G.; Valdes-Ramos, R.; Guadarrama-Lopez, A.L.; Pardo-Morales, R.V.; Martinez-Carrillo, B.E.; Harbige, L.S. Effect of n-3 polyunsaturated fatty acid supplementation on metabolic and inflammatory biomarkers in type 2 diabetes mellitus patients. Nutrients 2017, 9, 573. [Google Scholar] [CrossRef] [Green Version]
- Oliver, E.; McGillicuddy, F.; Phillips, C.; Toomey, S.; Roche, H.M. The role of inflammation and macrophage accumulation in the development of obesity-induced type 2 diabetes mellitus and the possible therapeutic effects of long-chain n-3 PUFA. Proc. Nutr. Soc. 2010, 69, 232–243. [Google Scholar] [CrossRef] [Green Version]
- Tull, S.P.; Yates, C.M.; Maskrey, B.H.; O’Donnell, V.B.; Madden, J.; Grimble, R.F.; Calder, P.C.; Nash, G.B.; Rainger, G.E. Omega-3 Fatty acids and inflammation: Novel interactions reveal a new step in neutrophil recruitment. PLoS Biol. 2009, 7, e1000177. [Google Scholar] [CrossRef] [Green Version]
- Calder, P.C. Omega-3 polyunsaturated fatty acids and inflammatory processes: Nutrition or pharmacology? Br. J. Clin. Pharm. 2013, 75, 645–662. [Google Scholar] [CrossRef] [Green Version]
- Dullemeijer, C.; Verhoef, P.; Brouwer, I.A.; Kok, F.J.; Brummer, R.J.; Durga, J. Plasma very long-chain n-3 polyunsaturated fatty acids and age-related hearing loss in older adults. J. Nutr. Health Aging 2010, 14, 347–351. [Google Scholar] [CrossRef]
- de Mello, A.H.; Uberti, M.F.; de Farias, B.X.; de Souza, N.A.R.; Rezin, G.T. n-3 PUFA and obesity: From peripheral tissues to the central nervous system. Br. J. Nutr. 2018, 119, 1312–1323. [Google Scholar] [CrossRef] [Green Version]
- Allaire, J.; Couture, P.; Leclerc, M.; Charest, A.; Marin, J.; Lepine, M.C.; Talbot, D.; Tchernof, A.; Lamarche, B. A randomized, crossover, head-to-head comparison of eicosapentaenoic acid and docosahexaenoic acid supplementation to reduce inflammation markers in men and women: The Comparing EPA to DHA (ComparED) Study. Am. J. Clin. Nutr. 2016, 104, 280–287. [Google Scholar] [CrossRef] [PubMed]
- Khorsan, R.; Crawford, C.; Ives, J.A.; Walter, A.R.; Jonas, W.B. The effect of omega-3 fatty acids on biomarkers of inflammation: A rapid evidence assessment of the literature. Mil. Med. 2014, 179, 2–60. [Google Scholar] [CrossRef] [Green Version]
- Rajkumar, H.; Mahmood, N.; Kumar, M.; Varikuti, S.R.; Challa, H.R.; Myakala, S.P. Effect of probiotic (VSL#3) and omega-3 on lipid profile, insulin sensitivity, inflammatory markers, and gut colonization in overweight adults: A randomized, controlled trial. Mediat. Inflamm. 2014, 2014, 348959. [Google Scholar]
- Menni, C.; Zierer, J.; Pallister, T.; Jackson, M.A.; Long, T.; Mohney, R.P.; Steves, C.J.; Spector, T.D.; Valdes, A.M. Omega-3 fatty acids correlate with gut microbiome diversity and production of N-carbamylglutamate in middle aged and elderly women. Sci. Rep. 2017, 7, 11079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Candido, F.G.; Valente, F.X.; Grzeskowiak, L.M.; Moreira, A.P.B.; Rocha, D.; Alfenas, R.C.G. Impact of dietary fat on gut microbiota and low-grade systemic inflammation: Mechanisms and clinical implications on obesity. Int. J. Food Sci. Nutr. 2018, 69, 125–143. [Google Scholar] [CrossRef] [PubMed]
- Wisniewski, P.J.; Dowden, R.A.; Campbell, S.C. Role of dietary lipids in modulating inflammation through the gut microbiota. Nutrients 2019, 11, 117. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
- Kaliannan, K.; Wang, B.; Li, X.Y.; Kim, K.J.; Kang, J.X. A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia. Sci. Rep. 2015, 5, 11276. [Google Scholar] [CrossRef]
- Mokkala, K.; Roytio, H.; Ekblad, U.; Laitinen, K. Opportunities for probiotics and polyunsaturated fatty acids to improve metabolic health of overweight pregnant women. Benef. Microbes 2017, 8, 3–15. [Google Scholar] [CrossRef]
- Claesson, M.J.; Jeffery, I.B.; Conde, S.; Power, S.E.; O’Connor, E.M.; Cusack, S.; Harris, H.M.; Coakley, M.; Lakshminarayanan, B.; O’Sullivan, O.; et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012, 488, 178–184. [Google Scholar] [CrossRef]
- Kobyliak, N.; Abenavoli, L.; Falalyeyeva, T.; Mykhalchyshyn, G.; Boccuto, L.; Kononenko, L.; Kyriienko, D.; Komisarenko, I.; Dynnyk, O. Beneficial effects of probiotic combination with omega-3 fatty acids in NAFLD: A randomized clinical study. Minerva Med. 2018, 109, 418–428. [Google Scholar] [CrossRef] [PubMed]
- Golkhalkhali, B.; Rajandram, R.; Paliany, A.S.; Ho, G.F.; Wan Ishak, W.Z.; Johari, C.S.; Chin, K.F. Strain-specific probiotic (microbial cell preparation) and omega-3 fatty acid in modulating quality of life and inflammatory markers in colorectal cancer patients: A randomized controlled trial. Asia Pac. J. Clin. Oncol. 2018, 14, 179–191. [Google Scholar] [CrossRef] [PubMed]
Probiotics | ω-3 Fatty Acids | Probiotics + ω-3 Fatty Acids |
---|---|---|
Positive effects on epithelial barrier function | Normalize secretion of cytokines | Associated with positive metabolic outcomes during pregnancy |
May play a role in forming neuroactive compounds | Adipose-specific blunting of inflammation and decreased storage of ingested fat | Improved insulin sensitivity in overweight adults |
Lower LPS-dependent chronic low-grade inflammation | Increase LPS-suppressing bacteria and decrease LPS-producing bacteria | Reductions in liver fat and systemic inflammation in adults with non-alcoholic fatty liver disease |
Reduce metabolic, inflammatory, and oxidative stress biomarkers | Promote SCFA production | Improved quality of life and inflammatory markers in colorectal cancer patients |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Hutchinson, A.N.; Tingö, L.; Brummer, R.J. The Potential Effects of Probiotics and ω-3 Fatty Acids on Chronic Low-Grade Inflammation. Nutrients 2020, 12, 2402. https://doi.org/10.3390/nu12082402
Hutchinson AN, Tingö L, Brummer RJ. The Potential Effects of Probiotics and ω-3 Fatty Acids on Chronic Low-Grade Inflammation. Nutrients. 2020; 12(8):2402. https://doi.org/10.3390/nu12082402
Chicago/Turabian StyleHutchinson, Ashley N., Lina Tingö, and Robert Jan Brummer. 2020. "The Potential Effects of Probiotics and ω-3 Fatty Acids on Chronic Low-Grade Inflammation" Nutrients 12, no. 8: 2402. https://doi.org/10.3390/nu12082402
APA StyleHutchinson, A. N., Tingö, L., & Brummer, R. J. (2020). The Potential Effects of Probiotics and ω-3 Fatty Acids on Chronic Low-Grade Inflammation. Nutrients, 12(8), 2402. https://doi.org/10.3390/nu12082402