Obstet Gynecol Sci.  2022 Jan;65(1):14-28. 10.5468/ogs.21185.

A narrative review of the role of gastrointestinal dysbiosis in the pathogenesis of polycystic ovary syndrome

Affiliations
  • 1School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia
  • 2Faculty of Science and Technology, University of Canberra, Canberra, Australia
  • 3College of Health and Medicine, University of Tasmania, Tasmania, Australia

Abstract

Diet-induced gastrointestinal dysbiosis has been hypothesized to play a significant role in stimulating an increase in gastrointestinal permeability and activating systemic inflammation in women with polycystic ovary syndrome (PCOS). We reviewed the current proof-of-concept studies on the proposed mechanism of dysbiosis in the pathogenesis of PCOS. A literature search was performed to identify articles on changes in the intestinal microbiome (dysbiosis) and increased intestinal mucosal permeability involving lipopolysaccharide (LPS), LPS-binding protein (LPS-BP), and zonulin. We also searched for systematic reviews and meta-analyses that synthesized the results of studies on the therapeutic effects of prebiotics, probiotics, or synbiotics in women with PCOS. Our search was confined to human studies between 2012 and 2021 using the PubMed, Scopus, and Cochrane databases. Thirty-one studies met the inclusion criteria (14 microbiota, 1 LPS, 1 LPS-BP, 1 LPS and LPS-BP, 5 zonulin, 9 systematic reviews). Our analysis revealed that most studies reported reduced alpha diversity and dysbiosis in women with PCOS. Preliminary studies suggest that LPS, LPS-BP, and zonulin may be involved in the pathophysiology of increased intestinal permeability. Treatment of PCOS with prebiotics, probiotics, and synbiotics appears to have a range of beneficial effects on metabolic and biochemical profiles. This review highlights the need for continued research into the pathophysiological mechanisms of dysbiosis and the clinical efficacy of prebiotics, probiotics, and synbiotics in women with PCOS.

Keyword

Dysbiosis; Lipopolysaccharides; Polycystic ovary syndrome; Probiotics; Zonulin

Figure

  • Fig. 1 PRISMA (preferred reporting items for systematic reviews and meta-analyses) flow diagram of study selection.


Reference

References

1. Ding T, Hardiman PJ, Petersen I, Wang FF, Qu F, Baio G. The prevalence of polycystic ovary syndrome in reproductive-aged women of different ethnicity: a systematic review and meta-analysis. Oncotarget. 2017; 8:96351–8.
Article
2. Boyle JA, Cunningham J, O’Dea K, Dunbar T, Norman RJ. Prevalence of polycystic ovary syndrome in a sample of Indigenous women in Darwin, Australia. Med J Aust. 2012; 196:62–6.
Article
3. Pathak G, Nichter M. Polycystic ovary syndrome in globalizing India: an ecosocial perspective on an emerging lifestyle disease. Soc Sci Med. 2015; 146:21–8.
Article
4. Szmigiera M. Proportion of selected age groups of world population in 2021, by region [Internet]. Hamburg (DEU): Statista;c2021. [cited 2021 May 1]. Available form: https://www.statista.com/statistics/265759/world-population-by-age-and-region/ .
5. Parker J. Understanding the pathogenesis of polycystic ovary syndrome: a transgenerational evolutionary adaptation to lifestyle and the environment. J ACNEM. 2020; 39:18–26.
6. Charifson MA, Trumble BC. Evolutionary origins of polycystic ovary syndrome: an environmental mismatch disorder. Evol Med Public Health. 2019; 2019:50–63.
Article
7. Azziz R, Dumesic DA, Goodarzi MO. Polycystic ovary syndrome: an ancient disorder? Fertil Steril. 2011; 95:1544–8.
Article
8. Teede H, Misso M, Costello M, Dokras A, Laven J, Moran L, et al. International evidence-based guideline for the assessment and management of polycystic ovary syndrome 2018 [Internet]. NHMRC Canberra (ACT). c2018. [cited 2020 Out 15]. Available form: https://www.monash.edu/__data/assets/pdf_file/0004/1412644/PCOS_Evidence-Based-Guidelines_20181009.pdf .
9. Benton ML, Abraham A, LaBella AL, Abbot P, Rokas A, Capra JA. The influence of evolutionary history on human health and disease. Nat Rev Genet. 2021; 22:269–83.
Article
10. Tremellen K, Pearce K. Dysbiosis of gut microbiota (DOGMA)--a novel theory for the development of polycystic ovarian syndrome. Med Hypotheses. 2012; 79:104–12.
11. Nuriel-Ohayon M, Neuman H, Koren O. Microbial changes during pregnancy, birth, and infancy. Front Microbiol. 2016; 7:1031.
Article
12. Yurtdaş G, Akdevelioğlu Y. A new approach to polycystic ovary syndrome: the gut microbiota. J Am Coll Nutr. 2020; 39:371–82.
Article
13. Zhao X, Jiang Y, Xi H, Chen L, Feng X. Exploration of the relationship between gut microbiota and polycystic ovary syndrome (PCOS): a review. Geburtshilfe Frauenheilkd. 2020; 80:161–71.
Article
14. He FF, Li YM. Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: a review. J Ovarian Res. 2020; 13:73.
Article
15. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012; 486:222–7.
Article
16. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012; 486:207–14.
17. Tuddenham S, Sears CL. The intestinal microbiome and health. Curr Opin Infect Dis. 2015; 28:464.
Article
18. García-Montero C, Fraile-Martínez O, Gómez-Lahoz AM, Pekarek L, Castellanos AJ, Noguerales-Fraguas F, et al. Nutritional components in western diet versus mediterranean diet at the gut microbiota-immune system interplay. Implications for health and disease. Nutrients. 2021; 13:699.
Article
19. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014; 505:559–63.
Article
20. Perez NB, Dorsen C, Squires A. Dysbiosis of the gut microbiome: a concept analysis. J Holist Nurs. 2020; 38:223–32.
Article
21. Moschen AR, Wieser V, Tilg H. Dietary factors: major regulators of the gut’s microbiota. Gut Liver. 2012; 6:411–6.
Article
22. Hawrelak JA, Myers SP. The causes of intestinal dysbiosis: a review. Altern Med Rev. 2004; 9:180–97.
23. Vujkovic-Cvijin I, Sklar J, Jiang L, Natarajan L, Knight R, Belkaid Y. Host variables confound gut microbiota studies of human disease. Nature. 2020; 587:448–54.
Article
24. Moya A, Ferrer M. Functional redundancy-induced stability of gut microbiota subjected to disturbance. Trends Microbiol. 2016; 24:402–13.
Article
25. Partula V, Mondot S, Torres MJ, Kesse-Guyot E, Deschasaux M, Assmann K, et al. Associations between usual diet and gut microbiota composition: results from the Milieu Intérieur cross-sectional study. Am J Clin Nutr. 2019; 109:1472–83.
Article
26. Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, Pelletier E, Frangeul L, et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006; 55:205–11.
Article
27. O’Brien CL, Pavli P, Gordon DM, Allison GE. Detection of bacterial DNA in lymph nodes of Crohn’s disease patients using high throughput sequencing. Gut. 2014; 63:1596–606.
Article
28. Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, et al. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012; 6:320–9.
Article
29. Mammadova G, Ozkul C, Yilmaz Isikhan S, Acikgoz A, Yildiz BO. Characterization of gut microbiota in polycystic ovary syndrome: findings from a lean population. Eur J Clin Invest. 2021; 51:e13417.
Article
30. Haudum C, Lindheim L, Ascani A, Trummer C, Horvath A, Münzker J, et al. Impact of short-term isoflavone intervention in polycystic ovary syndrome (PCOS) patients on microbiota composition and metagenomics. Nutrients. 2020; 12:1622.
Article
31. Zhang J, Sun Z, Jiang S, Bai X, Ma C, Peng Q, et al. Probiotic bifidobacterium lactis V9 regulates the secretion of sex hormones in polycystic ovary syndrome patients through the gut-brain axis. mSystems. 2019; 4:e00017–19.
Article
32. Insenser M, Murri M, Del Campo R, Martínez-García MÁ, Fernández-Durán E, Escobar-Morreale HF. Gut microbiota and the polycystic ovary syndrome: influence of sex, sex hormones, and obesity. J Clin Endocrinol Metab. 2018; 103:2552–62.
Article
33. Eyupoglu ND, Ergunay K, Acikgoz A, Akyon Y, Yilmaz E, Yildiz BO. Gut Microbiota and oral contraceptive use in overweight and obese patients with polycystic ovary syndrome. J Clin Endocrinol Metab. 2020; 105:dgaa600.
Article
34. Qi X, Yun C, Sun L, Xia J, Wu Q, Wang Y, et al. Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat Med. 2019; 25:1225–33.
Article
35. Jobira B, Frank DN, Pyle L, Silveira LJ, Kelsey MM, Garcia-Reyes Y, et al. Obese adolescents with PCOS have altered biodiversity and relative abundance in gastrointestinal microbiota. J Clin Endocrinol Metab. 2020; 105:e2134–44.
Article
36. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, et al. Alterations in gut microbiome composition and barrier function are associated with reproductive and metabolic defects in women with polycystic ovary syndrome (PCOS): a pilot study. PLoS One. 2017; 12:e0168390.
Article
37. Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X, et al. Dysbiosis of gut microbiota associated with clinical parameters in polycystic ovary syndrome. Front Microbiol. 2017; 8:324.
Article
38. Torres PJ, Siakowska M, Banaszewska B, Pawelczyk L, Duleba AJ, Kelley ST, et al. Gut microbial diversity in women with polycystic ovary syndrome correlates with hyperandrogenism. J Clin Endocrinol Metab. 2018; 103:1502–11.
Article
39. Chen F, LAI Z, XU Z. Analysis of the gut microbial composition in polycystic ovary syndrome with acne. Zigong Matern Child Heal Hosp. 2019; 35:2246–51.
40. Zhou L, Ni Z, Cheng W, Yu J, Sun S, Zhai D, et al. Characteristic gut microbiota and predicted metabolic functions in women with PCOS. Endocr Connect. 2020; 9:63–73.
Article
41. Garcia-Beltran C, Malpique R, Carbonetto B, González-Torres P, Henares D, Brotons P, et al. Gut microbiota in adolescent girls with polycystic ovary syndrome: effects of randomized treatments. Pediatr Obes. 2021; 16:e12734.
Article
42. Zeng B, Lai Z, Sun L, Zhang Z, Yang J, Li Z, et al. Structural and functional profiles of the gut microbial community in polycystic ovary syndrome with insulin resistance (IR-PCOS): a pilot study. Res Microbiol. 2019; 170:43–52.
Article
43. Rizk MG, Thackray VG. Intersection of polycystic ovary syndrome and the gut microbiome. J Endocr Soc. 2020; 5:bvaa177.
Article
44. Lim SS, Hutchison SK, Van Ryswyk E, Norman RJ, Teede HJ, Moran LJ. Lifestyle changes in women with polycystic ovary syndrome. Cochrane Database Syst Rev. 2019; 3:CD007506.
Article
45. González F, Considine RV, Abdelhadi OA, Acton AJ. Saturated fat ingestion promotes lipopolysaccharide-mediated inflammation and insulin resistance in polycystic ovary syndrome. J Clin Endocrinol Metab. 2019; 104:934–46.
Article
46. Zhu Q, Zhou H, Zhang A, Gao R, Yang S, Zhao C, et al. Serum LBP Is associated with insulin resistance in women with PCOS. PLoS One. 2016; 11:e0145337.
Article
47. Gutsmann T, Müller M, Carroll SF, MacKenzie RC, Wiese A, Seydel U. Dual role of lipopolysaccharide (LPS)-binding protein in neutralization of LPS and enhancement of LPS-induced activation of mononuclear cells. Infect Immun. 2001; 69:6942–50.
Article
48. Zhang D, Zhang L, Yue F, Zheng Y, Russell R. Serum zonulin is elevated in women with polycystic ovary syndrome and correlates with insulin resistance and severity of anovulation. Eur J Endocrinol. 2015; 172:29–36.
Article
49. Anik Ilhan G, Yildizhan B. Evaluation of zonulin, a marker of intestinal permeability as a novel biomarker in polycystic ovary syndrome. Fertil Steril. 2018; 110:e117.
Article
50. Banaszewska B, Siakowska M, Chudzicka-Strugala I, Chang RJ, Pawelczyk L, Zwozdziak , et al. Elevation of markers of endotoxemia in women with polycystic ovary syndrome. Hum Reprod. 2020; 35:2303–11.
Article
51. Fasano A, Uzzau S, Fiore C, Margaretten K. The enterotoxic effect of zonula occludens toxin on rabbit small intestine involves the paracellular pathway. Gastroenterology. 1997; 112:839–46.
Article
52. Larkin M. Out with the jab, in with the painless pills. Lancet. 1997; 349:1676.
53. Fasano A. Zonulin and its regulation of intestinal barrier function: the biological door to inflammation, autoimmunity, and cancer. Physiol Rev. 2011; 91:151–75.
Article
54. Fasano A. Intestinal permeability and its regulation by zonulin: diagnostic and therapeutic implications. Clin Gastroenterol Hepatol. 2012; 10:1096–100.
Article
55. Sapone A, de Magistris L, Pietzak M, Clemente MG, Tripathi A, Cucca F, et al. Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives. Diabetes. 2006; 55:1443–9.
Article
56. Fasano A. Zonulin, regulation of tight junctions, and autoimmune diseases. Ann N Y Acad Sci. 2012; 1258:25–33.
Article
57. Kim AS, Ko HJ. Plasma concentrations of zonulin are elevated in obese men with fatty liver disease. Diabetes Metab Syndr Obes. 2018; 11:149–57.
Article
58. Aasbrenn M, Lydersen S, Farup PG. Changes in serum zonulin in individuals with morbid obesity after weight-loss interventions: a prospective cohort study. BMC Endocr Disord. 2020; 20:108.
Article
59. Yuan JH, Xie QS, Chen GC, Huang CL, Yu T, Chen QK, et al. Impaired intestinal barrier function in type 2 diabetic patients measured by serum LPS, zonulin, and IFABP. J Diabetes Complications. 2021; 35:107766.
Article
60. Lingaiah S, Arffman RK, Morin-Papunen L, Tapanainen JS, Piltonen T. Markers of gastrointestinal permeability and dysbiosis in premenopausal women with PCOS: a case-control study. BMJ Open. 2021; 11:e045324.
Article
61. Cetin Z, Kosem A, Can B, Baser O, Catak M, Turhan T, et al. Serum zonulin level is not elevated in patients with polycystic ovary syndrome without metabolic syndrome. Arch Gynecol Obstet. 2019; 300:1785–90.
Article
62. Ajamian M, Steer D, Rosella G, Gibson PR. Serum zonulin as a marker of intestinal mucosal barrier function: may not be what it seems. PLoS One. 2019; 14:e0210728.
Article
63. Scheffler L, Crane A, Heyne H, Tönjes A, Schleinitz D, Ihling CH, et al. Widely used commercial ELISA for human Zonulin reacts with Complement C3 rather than pre-Haptoglobin 2 [Internet]. NY (USA): CSH;c2017. [cited 2021 Apr 17]. Available from: http://www.biorxiv.org/content/biorxiv/early/2017/06/30/157578.full.pdf .
64. Scheffler L, Crane A, Heyne H, Tönjes A, Schleinitz D, Ihling CH, et al. Widely used commercial elisa does not detect precursor of haptoglobin2, but recognizes properdin as a potential second member of the zonulin family. Front Endocrinol (Lausanne). 2018; 9:22.
Article
65. Sina C, Kemper C, Derer S. The intestinal complement system in inflammatory bowel disease: shaping intestinal barrier function. Semin Immunol. 2018; 37:66–73.
Article
66. Sünderhauf A, Skibbe K, Preisker S, Ebbert K, Verschoor A, Karsten CM, et al. Regulation of epithelial cell expressed C3 in the intestine - relevance for the pathophysiology of inflammatory bowel disease? Mol Immunol. 2017; 90:227–38.
Article
67. Liao D, Zhong C, Li C, Mo L, Liu Y. Meta-analysis of the effects of probiotic supplementation on glycemia, lipidic profiles, weight loss and C-reactive protein in women with polycystic ovarian syndrome. Minerva Med. 2018; 109:479–87.
Article
68. Heshmati J, Farsi F, Yosaee S, Razavi M, Rezaeinejad M, Karimie E, et al. The effects of probiotics or synbiotics supplementation in women with polycystic ovarian syndrome: a systematic review and meta-analysis of randomized clinical trials. Probiotics Antimicrob Proteins. 2019; 11:1236–47.
Article
69. Hadi A, Moradi S, Ghavami A, Khalesi S, Kafeshani M. Effect of probiotics and synbiotics on selected anthropometric and biochemical measures in women with polycystic ovary syndrome: a systematic review and meta-analysis. Eur J Clin Nutr. 2020; 74:543–7.
Article
70. Shamasbi SG, Ghanbari-Homayi S, Mirghafourvand M. The effect of probiotics, prebiotics, and synbiotics on hormonal and inflammatory indices in women with polycystic ovary syndrome: a systematic review and meta-analysis. Eur J Nutr. 2020; 59:433–50.
Article
71. Tabrizi R, Ostadmohammadi V, Akbari M, Lankarani KB, Vakili S, Peymani P, et al. The effects of probiotic supplementation on clinical symptom, weight loss, glycemic control, lipid and hormonal profiles, biomarkers of inflammation, and oxidative stress in women with polycystic ovary syndrome: a systematic review and meta-analysis of randomized controlled trials. Probiotics Antimicrob Proteins. 2019; Jun. 4. [Epub]. https://doi.ory/10.1007/s12602-019-09559-0 .
Article
72. Kazemi A, Soltani S, Ghorabi S, Keshtkar A, Daneshzad E, Nasri F, et al. Effect of probiotic and synbiotic supplementation on inflammatory markers in health and disease status: a systematic review and meta-analysis of clinical trials. Clin Nutr. 2020; 39:789–819.
Article
73. Cozzolino M, Vitagliano A, Pellegrini L, Chiurazzi M, Andriasani A, Ambrosini G, et al. Therapy with probiotics and synbiotics for polycystic ovarian syndrome: a systematic review and meta-analysis. Eur J Nutr. 2020; 59:2841–56.
Article
74. Miao C, Guo Q, Fang X, Chen Y, Zhao Y, Zhang Q. Effects of probiotic and synbiotic supplementation on insulin resistance in women with polycystic ovary syndrome: a meta-analysis. J Int Med Res. 2021; 49:3000605211031758.
Article
75. Li Y, Tan Y, Xia G, Shuai J. Effects of probiotics, prebiotics, and synbiotics on polycystic ovary syndrome: a systematic review and meta-analysis. Crit Rev Food Sci Nutr. 2021. Jul. 21. [Epub]. https://doi.ory/10.1080/10408398.2021.1951155 .
Article
76. Gibson GR, Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, et al. Expert consensus document: the international scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol. 2017; 14:491–502.
Article
77. Hawrelak J. Prebiotics, synbiotics and colonic foods. Murray M, Pizzorno J, editors. Textbook of natural medicine. 5th ed. Amsterdam: Elsevier;2020. p. 797–808.
Article
78. Gibson GR, Beatty ER, Wang X, Cummings JH. Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin. Gastroenterology. 1995; 108:975–82.
Article
79. Ballongue J, Schumann C, Quignon P. Effects of lactulose and lactitol on colonic microflora and enzymatic activity. Scand J Gastroenterol Suppl. 1997; 222:41–4.
Article
80. Vulevic J, Drakoularakou A, Yaqoob P, Tzortzis G, Gibson GR. Modulation of the fecal microflora profile and immune function by a novel trans-galactooligosaccharide mixture (B-GOS) in healthy elderly volunteers. Am J Clin Nutr. 2008; 88:1438–46.
Article
81. Dehghan P, Gargari BP, Jafar-Abadi MA, Aliasgharzadeh A. Inulin controls inflammation and metabolic endotoxemia in women with type 2 diabetes mellitus: a randomized-controlled clinical trial. Int J Food Sci Nutr. 2014; 65:117–23.
Article
82. Wang L, Yang H, Huang H, Zhang C, Zuo HX, Xu P, et al. Inulin-type fructans supplementation improves glycemic control for the prediabetes and type 2 diabetes populations: results from a GRADE-assessed systematic review and dose-response meta-analysis of 33 randomized controlled trials. J Transl Med. 2019; 17:410.
Article
83. Vulevic J, Juric A, Tzortzis G, Gibson GR. A mixture of trans-galactooligosaccharides reduces markers of metabolic syndrome and modulates the fecal microbiota and immune function of overweight adults. J Nutr. 2013; 143:324–31.
Article
84. Pain JA, Bailey ME. Experimental and clinical study of lactulose in obstructive jaundice. Br J Surg. 1986; 73:775–8.
Article
85. Jain L, Sharma BC, Srivastava S, Puri SK, Sharma P, Sarin S. Serum endotoxin, inflammatory mediators, and magnetic resonance spectroscopy before and after treatment in patients with minimal hepatic encephalopathy. J Gastroenterol Hepatol. 2013; 28:1187–93.
Article
86. Bianchi G, Ronchi M, Marchesini G. Effect of lactulose on carbohydrate metabolism and diabetes mellitus. Scand J Gastroenterol Suppl. 1997; 222:62–4.
Article
87. Sofi SA, Anjum A, Awsi J. Resistant starch as functional ingredient: a review. Int J Food Sci Nutr. 2017; 2:195–9.
88. Venkataraman A, Sieber JR, Schmidt AW, Waldron C, Theis KR, Schmidt TM. Variable responses of human microbiomes to dietary supplementation with resistant starch. Microbiome. 2016; 4:33.
Article
89. Bendiks ZA, Knudsen KEB, Keenan MJ, Marco ML. Conserved and variable responses of the gut microbiome to resistant starch type 2. Nutr Res. 2020; 77:12–28.
Article
90. Halajzadeh J, Milajerdi A, Reiner Ž, Amirani E, Kolahdooz F, Barekat M, et al. Effects of resistant starch on glycemic control, serum lipoproteins and systemic inflammation in patients with metabolic syndrome and related disorders: a systematic review and meta-analysis of randomized controlled clinical trials. Crit Rev Food Sci Nutr. 2020; 60:3172–84.
Article
91. Gholizadeh Shamasbi S, Dehgan P, Mohammad-Alizadeh Charandabi S, Aliasgarzadeh A, Mirghafourvand M. The effect of resistant dextrin as a prebiotic on metabolic parameters and androgen level in women with polycystic ovarian syndrome: a randomized, tripleblind, controlled, clinical trial. Eur J Nutr. 2019; 58:629–40.
Article
92. Shamasbi SG, Dehghan P, Charandabi SMA, Aliasgarzadeh A, Mirghafourvand M. Effect of prebiotic on anthropometric indices in women with polycystic ovarian syndrome: a triple-blind, randomized, controlled clinical trial. Iran Red Crescent Med J. 2018; 20:e67270.
Article
93. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014; 11:506–14.
94. Perraudeau F, McMurdie P, Bullard J, Cheng A, Cutcliffe C, Deo A, et al. Improvements to postprandial glucose control in subjects with type 2 diabetes: a multicenter, double blind, randomized placebo-controlled trial of a novel probiotic formulation. BMJ Open Diabetes Res Care. 2020; 8:e001319.
Article
95. Mishra V, Shah C, Mokashe N, Chavan R, Yadav H, Prajapati J. Probiotics as potential antioxidants: a systematic review. J Agric Food Chem. 2015; 63:3615–26.
Article
96. Imaoka A, Shima T, Kato K, Mizuno S, Uehara T, Matsumoto S, et al. Anti-inflammatory activity of probiotic Bifidobacterium: enhancement of IL-10 production in peripheral blood mononuclear cells from ulcerative colitis patients and inhibition of IL-8 secretion in HT-29 cells. World J Gastroenterol. 2008; 14:2511–6.
Article
97. Guillot CC, Bacallao EG, Dominguez MSC, Garcia MF, Gutierrez PM. Effects of Saccharomyces boulardii in children with chronic diarrhoea, especially cases due to giardiasis. Rev Mex De Puericultura Y Pediatria. 1995; 2:15.
98. In Kim H, Kim JK, Kim JY, Jang SE, Han MJ, Kim DH. Lactobacillus plantarum LC27 and Bifidobacterium longum LC67 simultaneously alleviate high-fat diet-induced colitis, endotoxemia, liver steatosis, and obesity in mice. Nutr Res. 2019; 67:78–89.
Article
99. Kocsis T, Molnár B, Németh D, Hegyi P, Szakács Z, Bálint A, et al. Probiotics have beneficial metabolic effects in patients with type 2 diabetes mellitus: a meta-analysis of randomized clinical trials. Sci Rep. 2020; 10:11787.
Article
100. del Campo R, Garriga M, Pérez-Aragón A, Guallarte P, Lamas A, Máiz L, et al. Improvement of digestive health and reduction in proteobacterial populations in the gut microbiota of cystic fibrosis patients using a Lactobacillus reuteri probiotic preparation: a double blind prospective study. J Cyst Fibros. 2014; 13:716–22.
Article
101. Odamaki T, Sugahara H, Yonezawa S, Yaeshima T, Iwatsuki K, Tanabe S, et al. Effect of the oral intake of yogurt containing Bifidobacterium longum BB536 on the cell numbers of enterotoxigenic Bacteroides fragilis in microbiota. Anaerobe. 2012; 18:14–8.
Article
102. Marteau P. Evidence of probiotic strain specificity makes extrapolation of results impossible from a strain to another, even from the same species [Internet]. Ann Gastroentol Hepatol. c2011. [cited 2021 Apr 12]. Available from: http://gastromasterclass2014.s3.amazonaws.com/Session1/Marteau+-+Evidence+of+probiotic+strain+specificity.pdf .
103. Swanson KS, Gibson GR, Hutkins R, Reimer RA, Reid G, Verbeke K, et al. The International scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat Rev Gastroenterol Hepatol. 2020; 17:687–701.
Article
104. Hadi A, Alizadeh K, Hajianfar H, Mohammadi H, Miraghajani M. Efficacy of synbiotic supplementation in obesity treatment: a systematic review and meta-analysis of clinical trials. Crit Rev Food Sci Nutr. 2020; 60:584–96.
Article
105. Ahmadi S, Jamilian M, Tajabadi-Ebrahimi M, Jafari P, Asemi Z. The effects of synbiotic supplementation on markers of insulin metabolism and lipid profiles in gestational diabetes: a randomised, double-blind, placebo-controlled trial. Br J Nutr. 2016; 116:1394–401.
Article
106. Zare Javid A, Aminzadeh M, Haghighi-Zadeh MH, Jamalvandi M. The effects of synbiotic supplementation on glycemic status, lipid profile, and biomarkers of oxidative stress in type 1 diabetic patients. A placebo-controlled, double-blind, randomized clinical trial. Diabetes Metab Syndr Obes. 2020; 13:607–17.
107. Tajabadi-Ebrahimi M, Sharifi N, Farrokhian A, Raygan F, Karamali F, Razzaghi R, et al. A randomized controlled clinical trial investigating the effect of synbiotic administration on markers of insulin metabolism and lipid profiles in overweight type 2 diabetic patients with coronary heart disease. Exp Clin Endocrinol Diabetes. 2017; 125:21–7.
Article
108. Rodgers RJ, Avery JC, Moore VM, Davies MJ, Azziz R, Stener-Victorin E, et al. Complex diseases and co-morbidities: polycystic ovary syndrome and type 2 diabetes mellitus. Endocr Connect. 2019; 8:R71–5.
Article
109. Zore T, Joshi NV, Lizneva D, Azziz R. Polycystic ovarian syndrome: long-term health consequences. Semin Reprod Med. 2017; 35:271–81.
Article
110. Wu J, Yao XY, Shi RX, Liu SF, Wang XY. A potential link between polycystic ovary syndrome and non-alcoholic fatty liver disease: an update meta-analysis. Reprod Health. 2018; 15:77.
Article
111. Esmaeilinezhad Z, Babajafari S, Sohrabi Z, Eskandari MH, Amooee S, Barati-Boldaji R. Effect of synbiotic pomegranate juice on glycemic, sex hormone profile and anthropometric indices in PCOS: a randomized, triple blind, controlled trial. Nutr Metab Cardiovasc Dis. 2019; 29:201–8.
Article
112. Darvishi S, Rafraf M, Asghari-Jafarabadi M, Farzadi L. Synbiotic supplementation improves metabolic factors and obesity values in women with polycystic ovary syndrome independent of affecting apelin levels: a randomized double-blind placebo - controlled clinical trial. Int J Fertil Steril. 2021; 15:51–9.
113. Karimi E, Moini A, Yaseri M, Shirzad N, Sepidarkish M, Hossein-Boroujerdi M, et al. Effects of synbiotic supplementation on metabolic parameters and apelin in women with polycystic ovary syndrome: a randomised double-blind placebo-controlled trial. Br J Nutr. 2018; 119:398–406.
Article
Full Text Links
  • OGS
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr