POSTBIOTICS AS FACTORS AGAINST DISEASES ALONG METABOLIC AXES

Authors

  • V. Lakhtin G.N. Gabrichevsky Research Institute for Epidemiology and Microbiology
  • M. Lakhtin G.N. Gabrichevsky Research Institute for Epidemiology and Microbiology
  • A. Melikhova G.N. Gabrichevsky Research Institute for Epidemiology and Microbiology
  • I. Davydkin G.N. Gabrichevsky Research Institute for Epidemiology and Microbiology
  • V. Davydkin G.N. Gabrichevsky Research Institute for Epidemiology and Microbiology
  • E. Klimova Moscow State University of Fine Chemical Technologies named after M.V. Lomonosov

DOI:

https://doi.org/10.31618/asj.2707-9864.2021.1.57.158

Keywords:

postbiotics, prebiotics, probiotic lectins, pathologies, diseases, prophylaxis, therapy.

Abstract

The review represents an analysis of recent year publications in connection with preventive and therapeutic use of postbiotics (PB). Postbiotics are widely used as immunomodulators, anti-inflammatory agents, protectors and the normalizers of metabolism of the open cavity mucosal biotopes, liver, brain and other organs, tissues and innate immunity cell populations that co-function to intestinal microbial metabolites as a network (cofunction within a number of metabolic axes “intestine-other locations”). They act in combination with other effectors as auxiliary agents prolonging the effect of drugs and supporting them. Prophylactic and therapeutic uses of PB are directed against groups of intestinal infection diseases, hepatitis, tumors, neurodegenerative brain disorders, other metabolic disorders and pathologies. New aspects of research of PB include the study and application of recognizing and binding therapeutic PB according to and in connection with a network of “Probiotic lectins—Glycoconjugates” interactions. The data indicate the prospects of a search and application of the new groups and combinations of PB directed at glycoconjugate exposed targets in accompanying and supportive therapy. Probiotic bifidobacteria, lactobacilli, baker’s yeast and probiotic lectins are perspective resources of synergistic sets of metabolite-cellular PB against diseases, pathologies and groups of infections.

References

Patel R.M., Denning P.W. Therapeutic use of prebiotics, probiotics, and postbiotics to prevent necrotizing enterocolitis: What is the current evidence? // Clin. Perinatol. 2013; 40 (1): 11-25. doi:10.1016/j.clp.2012.12.002.

Mosca F., Gianni M.L., Rescigno M. Can Postbiotics Represent a New Strategy for NEC? Adv. Exp. Med. Biol. 2019; 1125: 37-45. doi:10.1007/5584_2018_314.

Panebianco C., Andriulli A., Pazienza V. Pharmacomicrobiomics: exploiting the drugmicrobiota interactions in anticancer therapies // Microbiome. 2018 May22; 6(1): 92. doi:10.1186/s40168-018-0483-7.

Homayouni Rad A., Aghebati Maleki L., Samadi Kafil H., Abbasi A. Postbiotics: A novel strategy in food allergy treatment // Crit. Rev. Food Sci. Nutr. 2020:1-8. doi:10.1080/10408398.2020.1738333.

Albillos A., de Gottardi A., Rescigno M. The Gut-Liver Axis in Liver Disease: Pathophysiological Basis for Therapy // J. Hepatol. 2020;72(3):558-577. doi:10.1016/j.jhep.2019.10.003.

Serra D., Almeida L.M., Dinis T.C.P. The Impact of Chronic Intestinal Inflammation on Brain Disorders: The Microbiota-Gut-Brain Axis // Mol. Neurobiol. 2019;56(10):6941-6951. doi:10.1007/s12035-019-1572-8.

Oleskin A.V., Shenderov B.A. Probiotics and Psychobiotics: The Role of Microbial Neurochemicals // Probiotics Antimicrobial Proteins. 2019; 11 (4): 1071-85. doi:10.1007/s12602-019-09583-0.

Clua P., Kanmani P., Zelaya H., Tada A., Kober A.K.M.H., Salva S., Alvarez S., Kitazawa H., Villena J. Peptidoglycan from Immunobiotic Lactobacillus rhamnosus Improves Resistance of Infant Mice to Respiratory Syncytial Viral Infection and Secondary Pneumococcal Pneumonia // Front. Immunol. 2017 Aug 10; 8: 948. doi:10.3389/fimmu.2017.00948.

Lew L.-C., Liong M.-T. Bioactives From Probiotics for Dermal Health: Functions and Benefits // J. Appl. Microbiol. 2013;114(5):1241-1253. doi:10.1111/jam.12137.

Rinaldi F., Trink A., Pinto D. Efficacy of Postbiotics in a PRPLike Cosmetic Product for the Treatment of Alopecia Area Celsi: A Randomized Double-Blinded Parallel-Group Study // Dermatol. Ther. (Heidelb). 2020; 10 (3): 483-93. doi:10.1007/s13555-020-00369-9.

Pérez-Sánchez T., Mora-Sánchez B., Vargas A., Balcázar J.L. Changes in intestinal microbiota and disease resistance following dietary postbiotic supplementation in rainbow trout (Oncorhynchus mykiss) // Microbial Pathogenesis. 2020 Feb 11; 142: 104060. doi:10.1016/j.micpath.2020.104060.

Aguilar-Toalá J.E., Hall F.G., Urbizo-Reyes U.C., Garcia H.S., Vallejo-Cordoba B., GonzálezCórdova A.F., Hernández-Mendoza A., Liceaga A.M. In Silico Prediction and In Vitro Assessment of Multifunctional Properties of Postbiotics Obtained From Two Probiotic Bacteria // Probiotics Antimicrobial Proteins. 2019 Jul 6. doi:10.1007/s12602-019-09568-z.

Chuah L.O., Foo H.L., Loh T.C., Mohammed Alitheen N.B., Yeap S.K., Abdul Mutalib N.E., Abdul Rahim R., Yusoff K. Postbiotic Metabolites Produced by Lactobacillus plantarum Strains Exert Selective Cytotoxicity Effects on Cancer Cells // BMC Complement Altern Med. 2019 Jun 3; 19 (1): 114. doi: 10.1186/s12906-019-2528-2.

Petrova M.I. , Lievens E. , Verhoeven T.L.A. , Macklaim J.M. , Gloor G. , Schols D. , Vanderleyden J., Reid G. , Lebeer S. The Lectin-Like Protein 1 in Lactobacillus rhamnosus GR-1 Mediates TissueSpecific Adherence to Vaginal Epithelium and Inhibits Urogenital Pathogens. Sci. Rep. 2016 Nov 21;6:37437. doi:10.1038/srep37437.

Gao J., Li Y., Wan Y., Hu T., Liu L., Yang S., Gong Z., Zeng Q., Wei Y., Yang W., Zeng Z., He X., Huang S.H., Cao H. A Novel Postbiotic From Lactobacillus rhamnosus GG With a Beneficial Effect on Intestinal Barrier Function // Front. Microbiol. 2019 Mar 14;10:477. doi:10.3389/fmicb.2019.00477.

Popović N, Djokić J, Brdarić E, Dinić M, Terzić-Vidojević A, Golić N, Veljović K. The Influence of Heat-Killed Enterococcus faecium BGPAS1-3 on the Tight Junction Protein Expression and Immune Function in Differentiated Caco-2 Cells Infected With Listeria monocytogenes ATCC 19111 // Front. Microbiol. 2019 Mar 5; 10: 412. doi:10.3389/fmicb.2019.00412.

Pyclik M, Srutkova D, Schwarzer M, Górska S. Bifidobacteria cell wallderived exo-polysaccharides, lipoteichoic acids, peptidoglycans, polar lipids and proteins - their chemical structure and biological attributes // Int. J. Biol. Macromol. 2020; 147: 333-49. doi:10.1016/j.ijbiomac.2019.12.227.

Ren C, Faas MM, de Vos P. Disease managing capacities and mechanisms of host effects of lactic acid bacteria. Crit. Rev. Food Sci. Nutr. 2020:1-29. doi:10.1080/10408398.2020.1758625.

García-Carrizo F., Cannon B,, Nedergaard J,, Picó C,, Dols A,, Rodríguez A,M,, Palou A. Regulation of thermogenic capacity in brown and white adipocytes by the prebiotic high-esterified pectin and its postbiotic acetate // Int. J. Obes. (Lond). 2020; 44 (3): 715-26. doi:10.1038/s41366-019-0445-6.

Reynés B., Palou M., Rodríguez A.M., Palou A. Regulation of Adaptive Thermogenesis and Browning by Prebiotics and Postbiotics // Front. Physiol. 2019 Jan 10;9:1908. doi:10.3389/fphys.2018.01908.

Patil S., Sawant S., Hauff K., Hampp G. Validated Postbiotic Screening Confirms Presence of Physiologically-Active Metabolites, Such as ShortChain Fatty Acids, Amino Acids and Vitamins in Hylak® Forte // Probiotics Antimicrobial Proteins.2019; 11 (4): 1124-31. doi:10.1007/s12602018-9497-5.

Lakhtin M.V., Lakhtin V.M., Davydkin I.Yu., MelikhovaA.V., Davydkin V.Yu. Approaches to the study of therapeutic postbiotics // Ural Scientific Bulletin (Uralsk, Kazakhstan). – 2020. – Volume 3, No 5: 3-15. http://www.rusnauka.com/pdf/282581.pdf [ISSN: 1561-6908 in Russian]

Zeng W., Shen J., Bo T., Peng L., Xu H., Nasser M.I., Zhuang Q., Zhao M. Cutting Edge: Probiotics and Fecal Microbiota Transplantation in Immunomodulation // J. Immunol. Res. 2019 Apr 16; 2019: 1603758. doi:10.1155/2019/1603758.

Ahiwe E.U., Chang'a E.P., Abdallh M.E., AlQahtani M., Kheravii S.K. , Wu S., Graham H., Iji P.A.. Dietary Hydrolysed Yeast Cell Wall Extract Is Comparable to Antibiotics in the Control of Subclinical Necrotic Enteritis in Broiler Chickens // Br. Poult. Sci. 2019; 60 (6): 757-65. doi:10.1080/00071668.2019.1664727.

Khalique A., Zeng D., Shoaib M., Wang H., Qing X., Rajput D.S., Pan K., Ni X. Probiotics Mitigating Subclinical Necrotic Enteritis as Potential Alternatives to Antibiotics in Poultry // AMB Express. 2020 Mar 14; 10 (1): 50. doi:10.1186/s13568-02000989-6.

Ren H., Vahjen W., Dadi T., Saliu E.M., Boroojeni F.G., Zentek J. Synergistic Effects of Probiotics and Phytobiotics on the Intestinal Microbiota in Young Broiler Chicken // Microorganisms. 2019 Dec 11; 7 (12). pii: E684. doi:10.3390/microorganisms7120684.

Whitfield-Cargile C.M., Cohen N.D., Chapkin R.S., Weeks B.R., Davidson L.A., Goldsby J.S., Hunt C.L., Steinmeyer S.H., Menon R., Suchodolski J.S.,Jayaraman A., Alaniz R.C. The microbiota-derived metabolite indole decreases mucosal inflammation and injury in a murine model of NSAID enteropathy // Gut Microbes. 2016; 7 (3): 246-61. doi:10.1080/19490976.2016.1156827.

Beaumont M., Neyrinck A.M., Olivares M., Rodriguez J., de Rocca Serra A., Roumain M., Bindels L.B., Cani P.D., Evenepoel P., Muccioli G.G., Demoulin J.B., Delzenne N.M. The gut microbiota metabolite indole alleviates liver inflammation in mice // FASEB J. 2018 Jun 15: fj201800544. doi:10.1096/fj.201800544.

Zhao Z.H., Xin F.Z., Xue Y., Hu Z., Han Y., Ma F., Zhou D., Liu X.L., Cui A., Liu Z., Liu Y., Gao J., Pan Q., Li Y., Fan J.G. Indole-3-propionic acid inhibits gut dysbiosis and endotoxin leakage to attenuate steatohepatitis in rats // Exp. Mol. Med. 2019; 51 (9): 1-14. doi: 10.1038/s12276-019-0304-5.

Gaetani L. , Boscaro F. , Pieraccini G., Calabresi P., Romani L., Di Filippo M., Zelante T. Host and Microbial Tryptophan Metabolic Profiling in Multiple Sclerosis // Front. Immunol. 2020 Feb 18;11:157. doi:10.3389/fimmu.2020.00157.

Puccetti M., Giovagnoli S., Zelante T., Romani L., Ricci M. Development of Novel Indole-3Aldehyde-Loaded Gastro-Resistant Spray-Dried Microparticles for Postbiotic Small Intestine Local Delivery // J. Pharm. Sci. 2018; 107 (9): 2341-53. doi:10.1016/j.xphs.2018.04.023.

Lakhtin M.V., Lakhtin V.M., Afanasiev S.S., Aleshkin V.A. Selected prospects for application of symbiotic lectin systems of human microbiocenoses // Modern Scientific Bulletin (Belgorod, Russia). - 2017. – Volume 7. - No 2. – P. 48-72. https://elibrary.ru/item.asp?id=28381423 [ISSN: 1561-6886]

Cuellar-Guevara F.L., Barrón-González M.P., Menchaca-Arredondo J.L. Effect of Lactobacillus postbiotics on Entamoeba histolytica trophozoites // Rev. Invest. Clin // 2019; 71 (6): 402-7. doi:10.24875/RIC.19003134.

Rigo-Adrover M.D.M., Knipping K., Garssen J., van Limpt K., Knol J., Franch À., Castell M., Rodríguez-Lagunas M.J.,Pérez-Cano F.J. Prevention of Rotavirus Diarrhea in Suckling Rats by a Specific Fermented Milk Concentrate with Prebiotic Mixture // Nutrients. 2019 Jan 18; 11 (1). pii: E189. doi:10.3390/nu11010189.

Wang Y., Pan C.Q., Xing H. Advances in Gut Microbiota of Viral Hepatitis Cirrhosis // Biomed. Res Int. 2019 Nov 22; 2019: 9726786. doi:10.1155/2019/9726786.

Rad A.H., Aghebati-Maleki L., Kafil H.S., Abbasi A. Molecular mechanisms of postbiotics in colorectal cancer prevention and treatment // Crit. Rev. Food Sci. Nutr. 2020 May 15:1-17. doi:10.1080/10408398.2020.1765310.

Paul D., Manna S., Mandal S.M. Antibiotics Associated Disorders and Postbiotics Induced Rescue in Gut Health // Curr. Pharm. Des. 2018; 24 (7): 821-9. doi:10.2174/1381612824666171227221731.

Lakhtin M.V., Lakhtin V.M., Davydkin V.Yu. Recognizing glycoconjugate systems in medical biotechnology // Problems of scientific thought (Dnepropetrovsk, Ukraine). – 2020. – Volume 3, No 6. – P. 15-35. https://elibrary.ru/title_about.asp?id=56372 [ISSN: 1561-6916 in Russian]

Humam A.M., Loh T.C., Foo H.L., Samsudin A.A., Mustapha N.M., Zulkifli I., Izuddin W.I. Effects of Feeding Different Postbiotics Produced by Lactobacillus plantarum on Growth Performance, Carcass Yield, Intestinal Morphology, Gut Microbiota Composition, Immune Status, and Growth Gene Expression in Broilers under Heat Stress // Animals (Basel). 2019 Sep 2; 9 (9). pii: E644. doi:10.3390/ani9090644.

Lakhtin M.V., Afanasiev S.S., Lakhtin V.M., Aleshkin V.A. New glycoconjugates-recognition systems in culture fluids of promising probiotic strains of bifidobacteria and lactobacilli // Materiały IX Międzynarodowej naukowi-praktycznej konferencji

«Wykształcenie i nauka bez granic - 2013» Volume 37. Nauk biologicznych : Przemyśl. Nauka i studia. – 64-68. DOI link:

http://doi.org/10.17686/sced_rusnauka_2013-1314 http://www.rusnauka.com/Page_ru.htm [ISBN: 978-966-8736-05-6 in Russian]

Lakhtin M.V., Afanasiev S.S., Lakhtin V.M., Aleshkin V.A. Glycoconjugates-recognizing systems of bacterial cultures // Materiały X Międzynarodowej naukowi-praktycznej konferencji «Kluczowe aspekty naukowej działalności - 2014» Volume 16. - P. 17-21.

Nauk biologicznych. Fizyczna kultura i sport.: Przemyśl. Nauka i studia. DOI link: http://doi.org/10.17686/sced_rusnauka_2014-1316 , http://www.rusnauka.com/Page_ru.htm [ISBN: 978-966-8736-05-6 in Russian]

Lakhtin M.V., Lakhtin V.M., Afanasiev S.S., Aleshkin V.A. Diversity of lectin systems of probiotic bacteria // Bulletin of the VSSC SB RAMS (Acta biomedical scientifica). – 2015. - No 5: 79-82. https://elibrary.ru/title_about.asp?id= 8451

Haileselassie Y., Navis M., Vu N., Qazi K.R., Rethi B., Sverremark-Ekström E. Postbiotic Modulation of Retinoic Acid Imprinted Mucosal-like Dendritic Cells by Probiotic Lactobacillus reuteri 17938 in vitro // Front. Immunol. 2016 Mar 17; 7: 96. doi: 10.3389/fimmu.2016.00096.

Giorgetti G., Brandimarte G., Fabiocchi F., Ricci S., Flamini P., Sandri G., Trotta M.C., Elisei W., Penna A., Lecca P.G., Picchio M., Tursi A. Interactions between Innate Immunity, Microbiota, and Probiotics // J. Immunol. Res. 2015; 2015: 501361. doi:10.1155/2015/501361.

Lakhtin M.V., Lakhtin V.M., Afanasiev S.S., Aleshkin V.A. Mucosal immunity against pathogens and tumors with the participation of the system "Lectins of probiotics—Glycopolymers" // Bulletin of the VSNC SB RAMS (Acta Biomedica Scientifica). - 2015. - No 3. - P.62-9.

https://elibrary.ru/item.asp?id=23998391 [ISSN: 1811-0649 in Russian]

Lakhtin M.V., Lakhtin V.M., Afanasiev S.S., Aleshkin V.A. Functioning of protective systems: Mucosal immunity and the human complement system // Bulletin of the VSNC SB RAMS (Acta Biomedica Scientifica). – 2015. - No 5. – P. 113-22. https://elibrary.ru/item.asp?id=25307833 [ISSN: 18110649 in Russian]

Domínguez Rubio A.P., Martínez J.H., Martínez Casillas D.C., Coluccio Leskow F., Piuri M., Pérez O.E. Lactobacillus casei BL23 Produces Microvesicles Carrying Proteins That Have Been Associated with Its Probiotic Effect // Front. Microbiol. 2017 Sep20; Vol. 8: Article 1783.

doi:10.3389/fmicb.2017.01783. www.frontiersin.org

Cuccui J.. Wren B. Hijacking bacterial glycosylation for the production of glycoconjugates, from vaccines to humanised glycoproteins // J. Pharm. Pharmacol. 2015;67(3):338-350. doi:10.1111/jphp.12321.

Delgado S., Sánchez B., Margolles A., RuasMadiedo P., Ruiz L. Molecules Produced by Probiotics and Intestinal Microorganisms with Immunomodulatory Activity // Nutrients. 2020 Feb 1; 12 (2). pii: E391. doi:10.3390/nu12020391.

Wegh C.A.M., Geerlings S.Y., Knol J., Roeselers G., Belzer C. Postbiotics and Their Potential Applications in Early Life Nutrition and Beyond // Int. J. Mol. Sci. 2019 Sep 20; 20 (19). pii: E4673. doi:10.3390/ijms20194673.

Malagón-Rojas J.N., Mantziari A., Salminen S., Szajewska H. Postbiotics for Preventing and Treating Common Infectious Diseases in Children: A Systematic Review // Nutrients. 2020 Jan 31; 12 (2). pii: E389. doi:10.3390/nu12020389.

Klemashevich C., Wu C., Howsmon D., Alaniz R.C., Lee K., Jayaraman A. Rational identification of diet-derived postbiotics for improving intestinal microbiota function // Curr. Opin. Biotechnol. 2014; 26: 85-90. doi:10.1016/j.copbio.2013.10.006.

Dunand E., Burns P., Binetti A., Bergamini C., Peralta G.H., Forzani L., Reinheimer J., Vinderola G. Postbiotics produced at laboratory and industrial level as potential functional food ingredients with the capacity to protect mice against Salmonella infection // J. Appl. Microbiol. 2019 Jul; 127 (1): 219-29. doi:10.1111/jam.14276.

Paparo L., Nocerino R., Di Scala C., Della Gatta G., Di Costanzo M., Buono A., Bruno C., Berni Canani R. Targeting Food Allergy with Probiotics // Adv. Exp. Med. Biol. 2019; 1125: 57-68. doi:10.1007/5584_2018_316.

Cavallari J.F., Fullerton M.D., Duggan B.M., Foley K.P., Denou E., Smith B.K., Desjardins E.M., Henriksbo B.D., Kim K.J., Tuinema B.R., Stearns J.C., Prescott D., Rosenstiel P., Coombes B.K., Steinberg G.R., Schertzer J.D. Muramyl Dipeptide-Based Postbiotics Mitigate Obesity-Induced Insulin Resistance via IRF4 // Cell Metab. 2017 May 2 ;25 (5): 1063-74.e3. doi:10.1016/j.cmet.2017.03.021.

Yao Ang C., Sano M., Dan S. Leelakriangsak M., Lal T. M. Postbiotics Applications as Infectious Disease Control Agent in Aquaculture // Biocontrol. Sci. 2020; 25 (1): 1-7. doi:10.4265/bio.25.1.

Compare D., Rocco A., Coccoli P., Angrisani D., Sgamato C., Iovine B., Salvatore U., Nardone G. Lactobacillus casei DG and its postbiotic reduce the inflammatory mucosal response: an ex-vivo organ culture model of post-infectious irritable bowel syndrome // BMC Gastroenterol. 2017 Apr 14; 17 (1): 53. doi:10.1186/s12876-017-0605-x.

Han X., Lee A., Huang S., Gao J., Spence J.R., Owyang C. Lactobacillus rhamnosus GG prevents epithelial barrier dysfunction induced by interferongamma and fecal supernatants from irritable bowel syndrome patients in human intestinal enteroids and colonoids // Gut Microbes. 2019; 10 (1): 59-76. doi:10.1080/19490976.2018.1479625.

Tsilingiri K., Rescigno M. Postbiotics: What else? // Benef. Microbes. 2013; 4 (1): 101-7. doi:10.3920/BM2012.0046.

Wong A.C., Levy M. New Approaches to Microbiome-Based Therapies. mSystems. 2019 Jun 4; 4 (3). pii: e00122-19. doi:10.1128/mSystems. 00122-19.

Johnson C.N., Kogut M.H., Genovese K., He H., Kazemi S., Arsenault R.J. Administration of a Postbiotic Causes Immunomodulatory Responses in Broiler Gut and Reduces Disease Pathogenesis Following Challenge // Microorganisms. 2019 Aug 17; 7 (8). pii: E268. doi:10.3390/microorganisms7080268.

Kareem K.Y., Loh T.C., Foo H.L., Asmara S.A., Akit H. Influence of postbiotic RG14 and inulin combination on cecal microbiota, organic acid concentration, and cytokine expression in broiler chickens // Poult. Sci. 2017; 96 (4): 966-75. doi:10.3382/ps/pew362.

Roager H.M., Licht T.R. Microbial tryptophan catabolites in health and Disease // Nat. Commun. 2018 Aug 17; 9 (1): 3294. doi:10.1038/s41467-018-05470-4.

Dinić M., Lukić J., Djokić J., Milenković M., Strahinić I., Golić N., Begović J. Lactobacillus fermentum Postbiotic-induced Autophagy as Potential Approach for Treatment of Acetaminophen Hepatotoxicity // Front. Microbiol. 2017 Apr 6; 8: 594. doi:10.3389/fmicb.2017.00594.

Ji Y., Yin Y., Li Z., Zhang W. Gut Microbiota-Derived Components and Metabolites in the Progression of Non-Alcoholic Fatty Liver Disease (NAFLD) // Nutrients. 2019 Jul 25; 11 (8). pii: E1712. doi:10.3390/nu11081712.

He X., Zeng Q., Puthiyakunnon S., Zeng Z., Yang W., Qiu J., Du L., Boddu S., Wu T., Cai D., Huang S.-H., Ca H. Lactobacillus rhamnosus GG supernatant enhance neonatal resistance to systemic Escherichia coli K1 infection by accelerating development of intestinal defense // Scientific Reports. 2017 Mar 6; 7: 43305. doi: 10.1038/srep43305

Danilenko V., Devyatkin A., Marsova M., Shibilova M., Ilyasov R., Shmyrev V. Common Inflammatory Mechanisms in COVID-19 and

Parkinson’s Diseases: The Role of Microbiome, Pharmabiotics and Postbiotics in Their Prevention // J. Inflammation Research. 2021; 14: 6349–81. doi: 10.2147/JIR.S333887

Chudzik A., Orzyłowska A., Rola R., Stanisz G.J. Probiotics, Prebiotics and Postbiotics on Mitigation of Depression Symptoms: Modulation of the Brain–Gut–Microbiome Axis // Biomolecules. 2021 Jul; 11(7): 1000. doi: 10.3390/biom11071000

Salva S., Kolling Y., Ivir M., Gutiérrez F., Alvarez S. The Role of Immunobiotics and Postbiotics in the Recovery of Immune Cell Populations From Respiratory Mucosa of Malnourished Hosts: Effect on the Resistance Against Respiratory Infections // Frontiers in Nutrition. 2021; 8: 704868. doi: 10.3389/fnut.2021.704868

Liu Y., Wang J., Wu C. Microbiota and Tuberculosis: A Potential Role of Probiotics, and Postbiotics // Frontiers in Nutrition. 2021; 8: 626254. doi: 10.3389/fnut.2021.626254

Lakhtin V.M., Lakhtin M.V., Davydkin V.Yu., Melikhova A.V., Davydkin I.Yu., Zhilenkova O.G. Specific communicative relationships between proteins and glycoconjugates in organism in connection with COVID-19 infections // Ural Scientific Bulletin (Uralsk, Kazakhstan). 2020; Volume 2, No 12: 15-28. https://www.elibrary.ru/title_about.asp?id=53818

Pandey M., Bhati A., Priya K., Sharma K.K., Singhal B. Precision Postbiotics and Mental Health: the Management of Post-COVID-19 Complications // Probiotics Antimicrobial Proteins. 2021 Nov 22 : 1–23. doi: 10.1007/s12602-021-09875-4

Golkar N., Ashoori Y., Heidari R., Omidifar N., Abootalebi S.N., Mohkam M., Gholami A. A Novel Effective Formulation of Bioactive Compounds for Wound Healing: Preparation, In Vivo Characterization, and Comparison of Various Postbiotics Cold Creams in a Rat Model // EvidenceBased Complementary and Alternative Medicine. 2021; 2021: 8577116. doi: 10.1155/2021/8577116

Anhê F.F., Jensen B.A.H, Perazza L.R., Tchernof A., Schertzer J.D., Marette A. Bacterial Postbiotics as Promising Tools to Mitigate Cardiometabolic Diseases // J. Lipid Atherosclerosis. 2021; 10 (2): 123–9. doi: 10.12997/jla.2021.10.2.123

Cuevas-Sierra A., Ramos-Lopez O., Riezu-Boj J.I., Milagro F.I., Martinez J.A. Diet, Gut Microbiota, and Obesity: Links with Host Genetics and Epigenetics and Potential Applications // Adv. Nutr. 2019; 10 (suppl. 1): S17S30. doi:10.1093/advances/nmy078.

Nichols R.G., Peters J.M., Patterson A.D. Interplay Between the Host, the Human Microbiome, and Drug Metabolism // Hum. Genomics. 2019 Jun 11; 13 (1): 27. doi:10.1186/s40246-019-0211-9.

Cabello-Olmo M., Araña M., Urtasun R., Encio I.J., Barajas M. Role of Postbiotics in Diabetes Mellitus: Current Knowledge and Future Perspectives // Foods. 2021 Jul; 10 (7): 1590. doi: 10.3390/foods10071590

Downloads

Published

2022-01-31

Issue

Section

Статьи