Hostname: page-component-76fb5796d-25wd4 Total loading time: 0 Render date: 2024-04-27T12:39:55.167Z Has data issue: false hasContentIssue false

Effect of probiotics on postmenopausal bone health: a preclinical meta-analysis

Published online by Cambridge University Press:  23 October 2023

Shibani Bose
Affiliation:
Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru 570020, India
Kunal Sharan*
Affiliation:
Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru 570020, India Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
*
*Corresponding author: Dr Kunal Sharan, email kunalsharan@cftri.res.in; sharan.kunal@gmail.com

Abstract

Postmenopausal osteoporosis is a major concern for women worldwide due to increased risk of fractures and diminished bone quality. Recent research on gut microbiota has suggested that probiotics can combat various diseases, including postmenopausal bone loss. Although several preclinical studies have explored the potential of probiotics in improving postmenopausal bone loss, the results have been inconsistent and the mechanism of action remains unclear. To address this, a meta-analysis was conducted to determine the effect of probiotics on animal models of postmenopausal osteoporosis. The bone parameters studied were bone mineral density (BMD), bone volume fractions (BV/TV), and hallmarks of bone formation and resorption. Pooled analysis showed that probiotic treatment significantly improves BMD and BV/TV of the ovariectomised animals. Probiotics, while not statistically significant, exhibited a tendency towards enhancing bone formation and reducing bone resorption. Next, we compared the effects of Lactobacillus sp. and Bifidobacterium sp. on osteoporotic bone. Both probiotics improved BMD and BV/TV compared with control, but Lactobacillus sp. had a larger effect size. In conclusion, our findings suggest that probiotics have the potential to improve bone health and prevent postmenopausal osteoporosis. However, further studies are required to investigate the effect of probiotics on postmenopausal bone health in humans.

Type
Systematic Review and Meta-Analysis
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Brandi, ML (2009) Microarchitecture, the key to bone quality. Rheumatology 48, iv38.CrossRefGoogle Scholar
Kalpakcioglu, BB, Morshed, S, Engelke, K, et al. (2008) Advanced imaging of bone macrostructure and microstructure in bone fragility and fracture repair. J Bone Joint Surg Am 90, 6878.CrossRefGoogle ScholarPubMed
Neer, RM, Arnaud, CD, Zanchetta, JR, et al. (2001) Effect of parathyroid hormone (1–34) on fractures and bone mineral density in postmenopausal women with osteoporosis. N Engl J Med 344, 14341441.CrossRefGoogle ScholarPubMed
Miller, PD, Hattersley, G, Riis, BJ, et al. (2016) Effect of abaloparatide v. placebo on new vertebral fractures in postmenopausal women with osteoporosis: a randomized clinical trial. JAMA 316, 722733.CrossRefGoogle Scholar
Behera, J, Ison, J, Tyagi, SC, et al. (2020) The role of gut microbiota in bone homeostasis. Bone 135, 115317.CrossRefGoogle ScholarPubMed
Ding, K, Hua, F & Ding, W (2020) Gut microbiome and osteoporosis. Aging Dis 11, 438447.CrossRefGoogle ScholarPubMed
Rettedal, EA, Ilesanmi-Oyelere, BL, Roy, NC, et al. (2021) The gut microbiome is altered in postmenopausal women with osteoporosis and osteopenia. JBMR Plus 5, e10452.CrossRefGoogle ScholarPubMed
Wang, H, Liu, J, Wu, Z, et al. (2023) Gut microbiota signatures and fecal metabolites in postmenopausal women with osteoporosis. Gut Pathog 15, 33.CrossRefGoogle ScholarPubMed
Ling, CW, Miao, Z, Xiao, ML, et al. (2021) The association of gut microbiota with osteoporosis is mediated by amino acid metabolism: multiomics in a large cohort. J Clin Endocrinol Metab 106, e3852e3864.CrossRefGoogle ScholarPubMed
Ilesanmi-Oyelere, BL, Roy, NC & Kruger, MC (2021) Modulation of bone and joint biomarkers, gut microbiota, and inflammation status by synbiotic supplementation and weight-bearing exercise: human study protocol for a randomized controlled trial. JMIR Res Protoc 10, e30131.CrossRefGoogle ScholarPubMed
Rodrigues, FC, Castro, AS, Rodrigues, VC, et al. (2012) Yacon flour and bifidobacterium longum modulate bone health in rats. J Med Food 15, 664670.CrossRefGoogle ScholarPubMed
Howarth, GS & Wang, H (2013) Role of endogenous microbiota, probiotics and their biological products in human health. Nutrients 5, 5881.CrossRefGoogle ScholarPubMed
Clarke, G, Stilling, RM, Kennedy, PJ, et al. (2014) Minireview: gut microbiota: the neglected endocrine organ. Mol Endocrinol 28, 12211238.CrossRefGoogle ScholarPubMed
Villa, JKD, Diaz, MAN, Pizziolo, VR, et al. (2017) Effect of vitamin K in bone metabolism and vascular calcification: a review of mechanisms of action and evidences. Crit Rev Food Sci Nutr 57, 39593970.CrossRefGoogle ScholarPubMed
van Wijngaarden, JP, Doets, EL, Szczecińska, A, et al. (2013) Vitamin B12, folate, homocysteine, and bone health in adults and elderly people: a systematic review with meta-analyses. J Nutr Metab 2013, 486186.CrossRefGoogle ScholarPubMed
Ohlsson, C, Engdahl, C, Fåk, F, et al. (2014) Probiotics protect mice from ovariectomy-induced cortical bone loss. PLoS One 9, e92368.CrossRefGoogle ScholarPubMed
McCabe, LR, Irwin, R, Schaefer, L, et al. (2013) Probiotic use decreases intestinal inflammation and increases bone density in healthy male but not female mice. J Cell Physiol 228, 17931798.CrossRefGoogle Scholar
Collins, FL, Rios-Arce, ND, Schepper, JD, et al. (2019) Beneficial effects of Lactobacillus reuteri 6475 on bone density in male mice is dependent on lymphocytes. Sci Rep 9, 14708.CrossRefGoogle Scholar
Behera, J, Ison, J, Voor, MJ, et al. (2021) Probiotics stimulate bone formation in obese mice via histone methylations. Theranostics 11, 86058623.CrossRefGoogle ScholarPubMed
Higgins, JPT, Chandler, J, Cumpston, M, et al. (2019) Cochrane Handbook for Systematic Reviews of Interventions. Chichester, England: John Wiley & Sons.CrossRefGoogle Scholar
Cochran, WG (1954) The combination of estimates from different experiments. Biom 10, 101129.CrossRefGoogle Scholar
Vesterinen, HM, Sena, ES, Egan, KJ, et al. (2014) Meta-analysis of data from animal studies: a practical guide. J Neurosci Methods 221, 92102.CrossRefGoogle ScholarPubMed
Hooijmans, CR, Rovers, MM, de Vries, RB, et al. (2014) SYRCLE’s risk of bias tool for animal studies. BMC Med Res Methodol 14, 43.CrossRefGoogle ScholarPubMed
Narva, M, Rissanen, J, Halleen, J, et al. (2007) Effects of bioactive peptide, valyl-prolyl-proline (VPP), and lactobacillus helveticus fermented milk containing VPP on bone loss in ovariectomized rats. Ann Nutr Metab 51, 6574.CrossRefGoogle Scholar
Parvaneh, M, Karimi, G, Jamaluddin, R, et al. (2018) Lactobacillus helveticus (ATCC 27558) upregulates Runx2 and Bmp2 and modulates bone mineral density in ovariectomy-induced bone loss rats. Clin Interv Aging 13, 15551564.CrossRefGoogle ScholarPubMed
Shim, KS, Kim, T, Ha, H, et al. (2012) Hwangryun-haedok-tang fermented with lactobacillus casei suppresses ovariectomy-induced bone loss. Evid Based Complement Alternat Med 2012, 325791.CrossRefGoogle ScholarPubMed
Lee, YM, Kim, IS & Lim, BO (2019) Black rice (Oryza sativa L.) fermented with lactobacillus casei attenuates osteoclastogenesis and ovariectomy-induced osteoporosis. Biomed Res Int 2019, 5073085.CrossRefGoogle ScholarPubMed
Shim, KS, Kim, T, Ha, H, et al. (2013) Lactobacillus fermentation enhances the inhibitory effect of Hwangryun-haedok-tang in an ovariectomy-induced bone loss. BMC Complement Altern Med 13, 106.CrossRefGoogle Scholar
Lim, EY, Song, EJ, Kim, JG, et al. (2021) Lactobacillus intestinalis YT2 restores the gut microbiota and improves menopausal symptoms in ovariectomized rats. Benef Microbes 12, 503516.CrossRefGoogle ScholarPubMed
Montazeri-Najafabady, N, Ghasemi, Y, Dabbaghmanesh, MH, et al. (2019) Supportive role of probiotic strains in protecting rats from ovariectomy-induced cortical bone loss. Probiotics Antimicrob Proteins 11, 11451154.CrossRefGoogle ScholarPubMed
Parvaneh, K, Ebrahimi, M, Sabran, MR, et al. (2015) Probiotics (Bifidobacterium longum) increase bone mass density and upregulate Sparc and Bmp-2 genes in rats with bone loss resulting from ovariectomy. Biomed Res Int 2015, 897639.CrossRefGoogle ScholarPubMed
Dar, HY, Pal, S, Shukla, P, et al. (2018) Bacillus clausii inhibits bone loss by skewing Treg-Th17 cell equilibrium in postmenopausal osteoporotic mice model. Nutrition 54, 118128.CrossRefGoogle ScholarPubMed
Lee, S, Jung, DH, Park, M, et al. (2021) The effect of lactobacillus gasseri BNR17 on postmenopausal symptoms in ovariectomized rats. J Microbiol Biotechnol 31, 12811287.CrossRefGoogle ScholarPubMed
Lee, CS, Kim, JY, Kim, BK, et al. (2021) Lactobacillus-fermented milk products attenuate bone loss in an experimental rat model of ovariectomy-induced post-menopausal primary osteoporosis. J Appl Microbiol 130, 20412062.CrossRefGoogle Scholar
Britton, RA, Irwin, R, Quach, D, et al. (2014) Probiotic L. reuteri treatment prevents bone loss in a menopausal ovariectomized mouse model. J Cell Physiol 229, 18221830.CrossRefGoogle Scholar
Sapra, L, Dar, HY, Bhardwaj, A, et al. (2021) Lactobacillus rhamnosus attenuates bone loss and maintains bone health by skewing Treg-Th17 cell balance in Ovx mice. Sci Rep 11, 1807.CrossRefGoogle ScholarPubMed
Dar, HY, Shukla, P, Mishra, PK, et al. (2018) Lactobacillus acidophilus inhibits bone loss and increases bone heterogeneity in osteoporotic mice via modulating Treg-Th17 cell balance. Bone Rep 8, 4656.CrossRefGoogle ScholarPubMed
Wallimann, A, Hildebrand, M, Groeger, D, et al. (2021) An exopolysaccharide produced by bifidobacterium longum 35624® inhibits osteoclast formation via a TLR2-dependent mechanism. Calcif Tissue Int 108, 654666.CrossRefGoogle Scholar
Yang, LC, Lin, SW, Li, IC, et al. (2020) Lactobacillus plantarum GKM3 and Lactobacillus paracasei GKS6 supplementation ameliorates bone loss in ovariectomized mice by promoting osteoblast differentiation and inhibiting osteoclast formation. Nutrients 12, 1914.CrossRefGoogle ScholarPubMed
Yu, J, Hang, Y, Sun, W, et al. (2022) Anti-osteoporotic effect of lactobacillus brevis AR281 in an ovariectomized mouse model mediated by inhibition of osteoclast differentiation. Biol 11, 359.CrossRefGoogle Scholar
Chiang, SS & Pan, TM (2011) Antiosteoporotic effects of Lactobacillus -fermented soy skim milk on bone mineral density and the microstructure of femoral bone in ovariectomized mice. J Agric Food Chem 59, 77347742.CrossRefGoogle Scholar
Kim, DE, Kim, JK, Han, SK, et al. (2019) Lactobacillus plantarum NK3 and bifidobacterium longum NK49 alleviate bacterial vaginosis and osteoporosis in mice by suppressing NF-κB-linked TNF-α expression. J Med Food 22, 10221031.CrossRefGoogle ScholarPubMed
Sapra, L, Shokeen, N, Porwal, K, et al. (2022) Bifidobacterium longum ameliorates ovariectomy-induced bone loss via enhancing anti-osteoclastogenic and immunomodulatory potential of regulatory B cells (Bregs). Front Immunol 13, 875788.CrossRefGoogle ScholarPubMed
Yamada, T, Park, G, Node, J, et al. (2019) Daily intake of polyamine-rich Saccharomyces cerevisiae S631 prevents osteoclastic activation and bone loss in ovariectomized mice. Food Sci Biotechnol 28, 12411245.CrossRefGoogle ScholarPubMed
Ryan, R (2016) Cochrane Consumers and Communication Group: Meta-Analysis. Cochrane Consumers and Communication Review Group. http://cccrg.cochrane.org (accessed July 2023).Google Scholar
Li, C, Pi, G & Li, F (2021) The role of intestinal flora in the regulation of bone homeostasis. Front Cell Infect Microbiol 11, 579323.CrossRefGoogle ScholarPubMed
Davie, JR (2003) Inhibition of histone deacetylase activity by butyrate. J Nutr 133, 2485s2493s.CrossRefGoogle ScholarPubMed
Sanford, JA, Zhang, LJ, Williams, MR, et al. (2016) Inhibition of HDAC8 and HDAC9 by microbial short-chain fatty acids breaks immune tolerance of the epidermis to TLR ligands. Sci Immunol 1, eaah4609.CrossRefGoogle ScholarPubMed
Tyagi, AM, Yu, M, Darby, TM, et al. (2018) The microbial metabolite butyrate stimulates bone formation via T Regulatory cell-mediated regulation of WNT10B expression. Immunity 49, 11161131.e1117.CrossRefGoogle Scholar
Bodine, PV, Zhao, W, Kharode, YP, et al. (2004) The Wnt antagonist secreted frizzled-related protein-1 is a negative regulator of trabecular bone formation in adult mice. Mol Endocrinol 18, 12221237.CrossRefGoogle ScholarPubMed
Ohland, CL & Macnaughton, WK (2010) Probiotic bacteria and intestinal epithelial barrier function. Am J Physiol Gastrointest Liver Physiol 298, G807819.CrossRefGoogle ScholarPubMed
Guan, Z, Luo, L, Liu, S, et al. (2022) The role of depletion of gut microbiota in osteoporosis and osteoarthritis: a narrative review. Front Endocrinol (Lausanne) 13, 847401.CrossRefGoogle ScholarPubMed
Li, L, Rao, S, Cheng, Y, et al. (2019) Microbial osteoporosis: the interplay between the gut microbiota and bones via host metabolism and immunity. Microbiologyopen 8, e00810.CrossRefGoogle ScholarPubMed
Tanabe, S (2013) The effect of probiotics and gut microbiota on Th17 cells. Int Rev Immunol 32, 511525.CrossRefGoogle ScholarPubMed
Turner, AS (2001) Animal models of osteoporosis--necessity and limitations. Eur Cell Mater 1, 6681.CrossRefGoogle ScholarPubMed
Finkelstein, JS, Brockwell, SE, Mehta, V, et al. (2008) Bone mineral density changes during the menopause transition in a multiethnic cohort of women. J Clin Endocrinol Metab 93, 861868.CrossRefGoogle Scholar
Riggs, BL & Melton, LJ 3rd (1992) The prevention and treatment of osteoporosis. N Engl J Med 327, 620627.Google ScholarPubMed
Nguyen, TV, Sambrook, PN & Eisman, JA (1997) Sources of variability in bone mineral density measurements: implications for study design and analysis of bone loss. J Bone Miner Res 12, 124135.CrossRefGoogle ScholarPubMed
Tu, KN, Lie, JD, Wan, CKV, et al. (2018) Osteoporosis: a review of treatment options. Pharm Therapeut 43, 92104.Google ScholarPubMed
Singer, FR & Eyre, DR (2008) Using biochemical markers of bone turnover in clinical practice. Cleve Clin J Med 75, 739750.CrossRefGoogle ScholarPubMed
Ivaska, KK, Hentunen, TA, Vääräniemi, J, et al. (2004) Release of intact and fragmented osteocalcin molecules from bone matrix during bone resorption in vitro . J Biol Chem 279, 1836118369.CrossRefGoogle ScholarPubMed
Walker, HK, Hall, WD & Hurst, JW (1990) Clinical Methods: The History, Physical, and Laboratory Examinations. Boston: Butterworths. Copyright © 1990, Butterworth Publishers, a division of Reed Publishing.Google Scholar
Harris, H (1990) The human alkaline phosphatases: what we know and what we don’t know. Clin Chim Acta 186, 133150.CrossRefGoogle ScholarPubMed
Green, S, Anstiss, CL & Fishman, WH (1971) Automated differential isoenzyme analysis. II. The fractionation of serum alkaline phosphatases into ‘liver’, ‘intestinal’ and ‘other’ components. Enzymologia 41, 926.Google ScholarPubMed
Risteli, L & Risteli, J (1993) Biochemical markers of bone metabolism. Ann Med 25, 385393.CrossRefGoogle ScholarPubMed
Malluche, HH, Davenport, DL, Lima, F, et al. (2022) Prevalence of low bone formation in untreated patients with osteoporosis. PLoS One 17, e0271555.CrossRefGoogle ScholarPubMed
Whyte, MP, Bergfeld, MA, Murphy, WA, et al. (1982) Postmenopausal osteoporosis. A heterogeneous disorder as assessed by histomorphometric analysis of Iliac crest bone from untreated patients. Am J Med 72, 193202.CrossRefGoogle ScholarPubMed
Arlot, ME, Delmas, PD, Chappard, D, et al. (1990) Trabecular and endocortical bone remodeling in postmenopausal osteoporosis: comparison with normal postmenopausal women. Osteoporos Int 1, 4149.CrossRefGoogle ScholarPubMed
Udagawa, N, Takahashi, N, Jimi, E, et al. (1999) Osteoblasts/stromal cells stimulate osteoclast activation through expression of osteoclast differentiation factor/RANKL but not macrophage colony-stimulating factor: receptor activator of NF-kappa B ligand. Bone 25, 517523.CrossRefGoogle Scholar
Fujiwara, Y, Piemontese, M, Liu, Y, et al. (2016) RANKL (Receptor activator of NFκB ligand) produced by osteocytes is required for the increase in B cells and bone loss caused by estrogen deficiency in mice. J Biol Chem 291, 2483824850.CrossRefGoogle Scholar
Xiong, J, Piemontese, M, Onal, M, et al. (2015) Osteocytes, not osteoblasts or lining cells, are the main source of the RANKL required for osteoclast formation in remodeling bone. PLoS One 10, e0138189.CrossRefGoogle ScholarPubMed
Tap, J, Mondot, S, Levenez, F, et al. (2009) Towards the human intestinal microbiota phylogenetic core. Environ Microbiol 11, 25742584.CrossRefGoogle ScholarPubMed
Supplementary material: File

Bose and Sharan supplementary material

Bose and Sharan supplementary material
Download Bose and Sharan supplementary material(File)
File 329.2 KB