Hostname: page-component-76fb5796d-2lccl Total loading time: 0 Render date: 2024-04-25T15:01:16.768Z Has data issue: false hasContentIssue false

Phyto-oestrogens and bone health

Symposium on ‘Diet and bone health’

Published online by Cambridge University Press:  15 April 2008

Véronique Coxam*
Affiliation:
Unité de Nutrition Humaine (UMR1019), INRA Theix, 63122 Saint Genès Champanelle, France
*
Corresponding author: Dr Véronique Coxam, fax +33 473 62 46 38, email coxam@clermont.inra.fr
Rights & Permissions [Opens in a new window]

Abstract

As oestrogen deficiency is the main cause in the pathogenesis of osteoporosis hormone-replacement therapy remains the mainstay for prevention. However, prophylaxis by hormone-replacement therapy is limited. Phyto-oestrogens, which are weakly-oestrogenic compounds present in plants, deserve particular mention because emerging data support the suggestion that they may prevent bone loss associated with the menopause. In the past few years extensive research using animal models has provided convincing data to indicate a significant improvement in bone mass or other end points following feeding with soyabean. Moreover, observational studies relate the lower incidence of osteoporosis among women in the Eastern world to a diet rich in phyto-oestrogens. However, it is not valid to extrapolate to the Western situation. The varied clinical trials that have been published suggest that isoflavones reduce bone loss in women in the early period post menopause, but a definitive result requires more investigations of the effect of phyto-oestrogens on bone health that have substantial sample size and are of long duration. In addition, the clinical efficacy of soya foods in preventing osteopenia depends on their intestinal metabolism. Thus, phyto-oestrogens are a source for putative innovative dietary health intervention for post-menopausal women. However, more data are necessary, particularly in relation to their effect on the risk of fracture.

Type
Research Article
Copyright
Copyright © The Author 2008

Abbreviation:
ER

oestrogen receptor

Osteoporosis, a systemic skeletal disease characterized by a low bone mass and microarchitectural deterioration, is one of society's most debilitating and costly diseases, particularly in elderly women and increasingly in men. It has become a worldwide problem that is expected to worsen in many countries with ageing populations; the lifetime risk for osteoporotic fracture is 40% for women and approximately 15% for men(Reference Cooper, Campion and Melton1). This condition is characterized by an imbalance between the deposition of matrix and mineralization and the resorption activity resulting from an impairment in the remodelling process as a result of changes in the functional lifespan of osteoclasts and osteoblasts(Reference Raisz2).

Hormonal changes during the menopausal transition, which ultimately result in a decline in oestrogen, play a pivotal role in the development of this chronic disease(Reference Albright, Reifenstein, Albright and Reifenstein3). Indeed, oestrogens exhibit a broad range of physiological activities that are important in the homeostasic regulation of many cells, including bone cells. In women the menopause initiates an accelerated phase of predominantly cancellous bone loss that declines rapidly over 5–10 years to become asymptotic, with a subsequent slow phase that continues indefinitely. Oestrogen deficiency affects remodelling in several ways: it increases the activation frequency of bone modelling units, which leads to higher bone turnover; it induces a remodelling imbalance by prolonging the resorption phase and shortening the formation phase. As a consequence, the volume of the resorption cavity increases beyond the capacity of the osteoblasts to refill it(Reference Manolagas4). Thus, hormone-replacement therapies have been the first line of treatment for osteoporosis(Reference Gallagher5). However, available evidence appears to suggest that the long-term use of hormone-replacement therapy has numerous side effects (e.g. risk for developing breast and uterine cancers and CVD)(Reference Hulley, Grady, Bush, Furberg, Herrington, Riggs and Vittinghoff6Reference Beral, Bull, Green and Reeves8) that have led to a drastic reduction in the percentage of post-menopausal women on hormone-replacement therapy in the Western world.

Currently, natural alternatives with oestrogen-like activities are being investigated as possible new strategies for the treatment of osteoporosis. As it is evident that the effects of oestrogen on bone to a large extent are mediated via activation of its oestrogen receptors (ER; oestrogen signalling in mammalian cells is primarily mediated at the molecular level by two members of the nuclear receptor superfamily, ERα and ERβ)(Reference Moggs and Orphanides9), there is a great need for new activators of ER that selectively reproduce only the beneficial effects of oestrogen. Moreover, in addition to existing drug therapies, certain lifestyle and nutritional factors are known to reduce the risk of osteoporosis. Indeed, research into human nutrition has led to an awareness of the health benefits that diet can offer by providing several naturally-occurring bioactive molecules such as the phytochemicals, which include the phyto-oestrogens (whose common biological roles are to protect plants from stress or to act as part of a plant's defence mechanism) that can act as oestrogen mimics. While these natural secondary metabolites are widely occurring, being ubiquitous in higher plants, the major classes of current interest from a nutritional perspective are the isoflavones and the lignans, which are mainly found in soyabean and flaxseed respectively(Reference Axelson, Sjovall, Gustafsson and Setchell10) (although the lignans are much more widespread in plant foods, their investigation has been limited because of the complexity of measurement techniques). These compounds are capable of interacting with ER, showing both agonist and antagonist properties. A conspicuous feature of their non-steroidal chemical structure is the phenolic ring that is theoretically a prerequisite for an oestrogenic activity (binding to the ER). They also share a pair of hydroxyl groups, one being a substituent of an aromatic A ring, while the second lies at the opposite end(Reference Miksicek11, Reference Setchell12). These phyto-oestrogens thus have potential as a putative and innovative dietary health intervention for post-menopausal women, and are currently being studied for the prevention of sex hormone-related diseases such as breast cancer and prostate cancer(Reference Adlercreutz and Mazur13), as well as post-menopausal osteoporosis(14Reference Brynin16).

The rationale for phyto-oestrogens

Foods based on soyabean have generated much interest recently as a result of evidence that populations consuming large amounts of soyabeans have a lower risk of some chronic diseases, notably osteoporosis. Indeed, the marked differences in the incidence of clinically-diagnosed osteoporotic fractures within Europe and even worldwide (the lowest rate occurring in countries in the East and to the South) could be related to specific nutritional practices. However, identifying the individual or multiple components of the diet involved in the pathogenesis of osteoporosis is extremely difficult because of the complex interplay between lifestyle factors, genetics and many confounding variables. Soyabean, a legume that has been grown for thousands of years, is traditionally used to prepare both fermented and non-fermented foods and is a staple among Asian populations. Soya foods contain an array of biologically-active phytochemicals that may confer important health benefits. These compounds include the isoflavones, which have received considerable attention because of their oestrogen-like properties in certain tissues, including bone. Thus, some investigators have referred to them as naturally-occurring selective ER modulators(Reference Branca and Lorenzetti17, Reference Ososki and Kennelly18).

The biological effects of phyto-oestrogens have been characterized using DNA microarrays (Reference Ise, Han, Takahashi, Terasaka, Inoue, Tanji and Kiyama19). Gene expression profiling has demonstrated that 17β-oestradiol, genistein (one of the two major isoflavones) and the synthetic oestrogen diethylstilbestrol alter the expression of the same 179 genes in the intact immature mouse uterus (under conditions in which each compound produces an equivalent gravimetric and histological uterotrophic effect), including lactotransferrin, complement component 3 and c-fos(Reference Moggs, Ashby, Tinwell, Lim, Moore, Kimber and Orphanides20).

Phyto-oestrogens may therefore have potential for maintaining or modestly improving the bone mass of human subjects. A substantial amount of research carried out in recent years using animal models has provided convincing data to indicate that feeding soyabeans results in a significant improvement in bone mass or other end points(Reference Coxam21). These studies strongly indicate that this bone-sparing effect of soyabean is attributable to its isoflavone component. However, because of the constraints associated with the systems used to study the ageing skeleton, the information gained is necessarily limited and the focus must be on clinical trials.

Phyto-oestrogens and bone health trials

Observational studies

Several studies have indicated a relationship between the lower incidence of oestrogen-deficiency-related diseases (i.e. osteoporosis) among women in Eastern countries and a diet rich in phyto-oestrogens. Moreover, a clear relationship has been demonstrated between the level of soyabean consumption and bone mineral density (see Table 1). However, it has been shown that extrapolation to the Western situation is not valid; a single component of a total lifestyle of such communities, in which several other significant lifestyle factors are operative, cannot be expected to show a definitive correlation with disease. Moreover, consumption of soyabean is very low within the Western population and phyto-oestrogen exposure varies substantially across human populations and individuals. Furthermore, Asian diets, which are particularly high in soyabean, result in isoflavone consumption as high as 1 mg/g body weight per d, with plasma isoflavone concentrations reaching 1 μm in Japanese subjects. In Europe circulating levels are usually <0·07 μm for omnivores, and 0·4 μm-isoflavones and 0·8 μm-lignans among vegetarians(Reference Adlercreutz, Yamada, Wähälä and Watanabe22, Reference Whitten and Patisaul23). Another limitation is the window of exposure(Reference Coxam and Gerber24).

Table 1. Epidemiological studies in women

BMD, bone mineral density; NTX, N-terminal cross-linked telopeptides; bALP, bone-specific alkaline phosphatase.

* Fermented soyabean.

Intervention trials

In adolescents isoflavone supplementation (Novasoy; Archer Daniels Midland Company, Decatur, IL, USA; 50 mg isoflavone equivalents/d) for 6 weeks has been shown to have no measurable effect on bone turnover in adolescent boys(Reference Jones, Dwyer, Hynes, Dalais, Parameswaran and Greenaway25), even though results from a study of 104 Chinese adolescent girls aged 14–16 years indicate that supplementation with 375 ml Ca-fortified soya milk, or an equivalent of about two glasses, can be considered among the effective strategies for bone acquisition and the optimization of peak bone mass in adolescent girls (because the Chinese diet is low in Ca; although this study did not establish which component(s) of the Ca-fortified soya milk (or their combined effect) resulted in the beneficial effect on bone health)(Reference Ho, Guldan, Woo, Yu, Tse, Sham and Cheng26). In young adult females with normal menses (21–25 years of age) an isoflavone-rich soya preparation (approximately 90 mg total isoflavones/d) has been shown to have no effects on bone mineral content and bone mineral density over a 12-month period(Reference Anderson, Chen, Boass, Symons, Kohlmeier, Renner and Garner27).

Intervention trials carried out in post-menopausal women are reported in Tables 2 and 3. These studies used either biochemical indices of bone turnover (Table 2) or direct measurements of bone mineral density (Table 3) to examine the effect of soyabean isoflavones ranging from 54 mg to 300 mg/d (most studies used 80–110 mg/d). Evidence from several studies suggests that soyabean proteins and/or their isoflavones may have beneficial effects on bone in post-menopausal women, whereas other trials do not show a benefit over the same or a longer period. Nevertheless, both studies that also targeted Ca have failed to show any significant effect on Ca retention(Reference Spence, Lipscomb, Cadogan, Martin, Wastney, Peacock and Weaver28, Reference Roughead, Hunt, Johnson, Badger and Lykken29), despite a 15–20% lower renal acid excretion with the soyabean diet compared with meat(29). Moreover, there is also inconsistency among the studies that show favourable effects, with some finding a benefit in the spine but not the hip and others a benefit in the hip but not the spine.

Table 2. Intervention trials: effect of soyabean and isoflavones (IF) consumption on bone turnover in women

NTX, N-terminal cross-linked telopeptides; DPD, deoxypyridinoline; CTX, C-terminal cross-linked telopeptides; ALP, alkaline phosphatase; IGFBP, insulin-like growth factor-binding protein.

Table 3. Intervention trials: effect of soyabean and isoflavones (IF) consumption on bone mineral density in post-menopausal women

HRT, hormone-replacement therapy; BMC, bone mineral content; BMD, bone mineral density; DPD, deoxypyridinoline.

One study has identified fifteen clinical trials that have examined the effects of isoflavones or isoflavone-rich soyabean protein on bone mineral density(Reference Messina, Ho and Alekel30). Most trials were conducted for ⩽1 year and involved relatively few (less than thirty) participants per group. Although the findings from these studies are inconsistent, in general they suggest that isoflavones reduce bone loss in post-menopausal women in the early years following the menopause (<5 years post menopause). Furthermore, two consensus views of soyabean phyto-oestrogens in clinical interventions in post-menopausal women have been published recently; one by the French Food Safety Agency(Reference Coxam and Gerber24) and one from the EU-funded project Phytohealth(Reference Cassidy, Albertazzi and Lise Nielsen31). The comprehensive literature search with well-defined inclusion or exclusion criteria suggests, although there is no conclusive evidence, that isoflavones from the sources studied so far have a beneficial effect on bone health. The health benefits of soyabean phyto-oestrogens in healthy post-menopausal women are subtle and even some well-designed studies do not show protective effects. Both papers conclude that until more definite data are available, soya foods and isoflavones cannot be viewed as substitutes for established anti-osteoporotic medication.

The varied results of the clinical studies that have been published suggest the need for investigations of the effect of phyto-oestrogens on bone health that have substantial sample size and long duration to provide a definitive result. It has been suggested that the differences may be a result of synergies or antagonisms among the flavones, threshold or biphasic dose effects, life-stage oestrogen status or environmental interactions, including the ability to produce metabolites on ingestion of isoflavones(Reference Weaver and Cheong32). Peri-menopausal women and women in early menopause may therefore be more receptive to the therapeutic effects of isoflavones before the decrease in ER that occurs in the post-menopausal years(Reference Reinwald and Weaver33). Moreover, the major weakness of previous clinical trials is that the risk of fracture, which is actually the ultimate hallmark of bone quality, was not targeted. However, the relationship between usual soyabean food consumption and fracture incidence has been examined in 24 403 post-menopausal Chinese women (recruited in the Shanghai Women's Health Study, a cohort study of approximately 75 000 women aged 40–70 years)(Reference Zhang, Shu, Li, Yang, Li, Gao and Zheng34). Evidence was found that soya food consumption may reduce the risk of fracture in these women, particularly among those in the early years following menopause, (after adjustment for confounding factors, the relative risks of fracture were 1·00, 0·72 (95% CI 0·62, 0·83), 0·69 (95% CI 0·59,0·80), 0·64 (95% CI 0·55, 0·76) and 0·63 (95% CI 0·53, 0·76) across quintiles of soyabean protein intake (P<0·001 for trend). However, it is questionable whether these data can be extrapolated to the Caucasian population.

Phyto-oestrogen biovailability and bone health

It has been found that the intestinal metabolism of isoflavones could be the more important clue to the clinical efficacy of soya foods in preventing osteopenia(Reference Setchell, Brown and Lydeking-Olsen35). Indeed, the bioavailability of isoflavones requires an initial hydrolysis of the sugar moiety by intestinal bacterial (Lactobacilli, Bacteroides and Bifidobacteria) β-glucosidases to allow subsequent uptake by enterocytes. Moreover, the final exposure depends on the microbial potential to activate all different groups of phyto-oestrogens within each individual subject. Indeed, phyto-oestrogens (and more especially daidzein) can undergo a further metabolism and be converted into a more potent molecule, equol(Reference Setchell, Brown and Lydeking-Olsen35). However, this bacterial metabolite has been found, on average, in only 45% of the post-menopausal women studied(Reference Axelson, Kirk, Farrant, Cooley, Lawson and Setchell36). It has been suggested that individuals can be separated into high, moderate and low O-desmethylangolensin, equol, enterodiol, enterolactone or 8-prenylnaringenin producers, even though the metabolism of isoflavones, lignans and prenylflavonoids follows separate independent pathways(Reference Possemiers, Bolca, Eeckhaut, Depypere and Verstraete37). Moreover, enterodiol production correlates negatively with Clostridium coccoidesEubacterium rectale counts, while O-desmethylangolensin production is associated with the abundance of methanogens, whereas equol production is correlated with sulfate-reducing bacteria.

Equol has a longer half-life and a much higher affinity for the ER than its precursor (daidzein) and has the highest antioxidant capacity among isoflavones(Reference Setchell, Brown and Lydeking-Olsen35). It has been reported that isoflavone treatment in subjects who have the capacity to produce equol differentially affects gene expression as compared with non-producers(Reference Niculescu, Pop, Fischer and Zeisel38), supporting the plausibility of the importance of equol production. In general, isoflavones have a stronger effect on some putative oestrogen-responsive genes in equol producers than in non-producers. This finding could explain the results from a 2-year study carried out in post-menopausal women randomized to consume 500 ml soya milk either with or without isoflavones(Reference Lydeking-Olsen, Jensen, Setchell, Damhus and Jensen39). In this study greater effects on bone health were elicited when volunteers were able to produce equol (a 2·4% increase in lumbar-spine bone mineral density compared with the control group), while no significant difference was shown in non-producers. Similarly, it has been reported that in 128 volunteers classified by equol status (producers or non-producers) the percentage changes in bone mineral density for equol producers are –0·53 and +0·13 in the sub-whole body and total hip respectively, which are significantly different from the corresponding changes of –1·35 and –1·77 in non-producers in the isoflavone group (P=0·049 and P=0·040 respectively)(Reference Wu, Oka and Higuchi40). However, in another study it was found that total leg and head bone mineral density are greater (6%) in O-desmethylangolensin producers (n 76) compared with non-producers (n 16), while total and site-specific bone mineral density are not different in equol producers (n 24) and non-producers (n 68)(Reference Frankenfeld, McTiernan, Thomas, LaCroix, McVarish, Holt, Schwartz and Lampe41). Furthermore, equol has been shown to inhibit bone loss in ovariectomized mice, apparently without oestrogenic activity in the reproductive organs(Reference Fujioka, Uehara, Wu, Adlercreutz, Suzuki, Kanazawa, Takeda, Yamada and Ishimi42). Hence, substances that can modulate the intestinal microflora could affect equol production. The prebiotic fructo-oligosaccharides have been reported to enhance the effects of soyabean isoflavones on bone mineral density(Reference Mathey, Mardon and Fokialakis43) and to reverse the loss of certain microarchitectural variables such as tibial trabecular number, separation and thickness(Reference Devareddy, Khalil, Korlagunta, Hooshmand, Bellmer and Arjmandi44) in ovariectomized osteopenic female rats.

Mechanisms of action

Phyto-oestrogens could elicit their bone sparing effect by modulating Ca metabolism, as has been reported for oestrogens(Reference Ten Bolscher, Netelenbos, Barto, Van Buuren and Van der Vijgh45). There is evidence that intestinal Ca absorption is higher in ovariectomized rats given soya milk-containing diets than in those receiving a control diet(Reference Omi, Aoi, Murata and Ezawa46). Nevertheless, it has been shown that ipriflavone, a synthetic phyto-oestrogen-like compound that is similar in structure to the soyabean isoflavone daidzein, increases total Ca transport under low oestrogen conditions, but not under oestrogen-free conditions (via an increased transcellular transport)(Reference Cotter and Cashman47). Moreover, coumestrol or apigenin have no effect on Ca transport.

Phyto-oestrogens can directly modulate bone metabolism. Using a radioactive cDNA microarray to investigate gene expression profiles (1152 genes), it has been shown that genistein treatment in ovariectomized mice modulates bone metabolism-related gene expression, including calciotropic receptor, cytokines, growth factors and bone matrix protein; thirty-eight genes being up regulated (e.g. mitogen-activated protein kinase 10), while eighteen are down regulated (e.g. matrix metalloproteinase 13)(Reference Pie, Park, Park, Ryu, Kim, Suh, Becker, Cho-Chung and Kim48). Similarly, it has been demonstrated that in hFOB/ERα9 cells (normal fetal osteoblasts) both genistein and oestradiol increase the endogenous gene expression of the progesterone receptor, the proteoglycan versican and alkaline phosphatase, but inhibit osteopontin gene expression and IL-6 protein levels(Reference Rickard, Monroe, Ruesink, Khosla, Riggs and Spelsberg49).

In fact, both genomic and non-genomic mechanisms have been proposed to explain their effect. They are able to interact with enzymes and receptors and, because of their stable structure and low molecular weight, they can pass through cell membranes(Reference Adlercreutz50). Mechanistically, phyto-oestrogens have been shown to bind to the two types of ER, their relative affinity for ERβ being greater than that for ERα(Reference Kuiper, Lemmen, Carlsson, Corton, Safe, van der Saag, van der Burg and Gustafsson51), and may thus produce distinct clinical effects from oestrogens by differentially triggering transcriptional activation or repression pathways via ERβ(Reference De Wilde, Lieberherr, Colin and Pointillart52, Reference de Wilde, Heberden, Chaumaz, Bordat and Lieberherr53). Moreover, they can act on both bone cell lineages. It has been reported that phyto-oestrogens such as coumestrol, genistein and daidzein increase alkaline phosphatase activity and enhance bone mineralization in MC3T3-E1 cells (osteoblast-like cell)(Reference Kanno, Hirano and Kayama54). It has been demonstrated that daidzein stimulates osteoblast differentiation at various stages (from osteoprogenitors to terminally-differentiated osteoblasts)(Reference Jia, Wang, Xie, Wang and Zhang55, Reference Ge, Chen, Xie and Zhang56). Daidzein induces changes in the actin cytoskeleton responsible for cell adhesion and motility and activates two transcription factors, cAMP-response element-binding protein and Elk1, which are linked to early genes controlling cell proliferation and differentiation via the mitogen-activated protein kinases extracellular signal-regulated kinases 1 and 2(Reference de Wilde, Heberden, Chaumaz, Bordat and Lieberherr53). It is also recognized that isoflavones inhibit in vitro bone resorption, via direct targeting of osteoclasts and/or osteoclast progenitors. Certainly, genistein can decrease osteoclast differentiation and increase osteoclast apoptosis or interfere with signalling pathways such as intracellular Ca2+, cAMP or protein kinase and protein tyrosine phosphatase or via inhibition of topoisomerase II activity(Reference Gao and Yamaguchi57, Reference Sliwinski, Folwarczna, Janiec, Grynkiewicz and Kuzyk58). Tyrosine kinase inhibition, in turn, has been reported to directly inhibit osteoclast membrane HCl transport(Reference Williams, Jordan, Barnes and Blair59). Furthermore, the maturation of osteoclast is dependent on two factors, macrophage colony-stimulating factor and receptor activator of NF-κB-ligand. It has been shown that coumestrol has an inhibitory effect on the differentiation of osteoclasts, at least partially via decreased receptor activator of NF-κB-ligand-induced phosphorylation of extracellular signal-regulated kinases/p44/42 (Reference Jia, Wang, Xie, Wang and Zhang55). These data are consistent with other published data(Reference García Palacios, Robinson, Borysenko, Lehmann, Kalla and Blair60). Furthermore, phyto-oestrogens such as genistein are able to enhance osteoblastic osteoprotegerin production through ERα-dependent mechanisms and concurrently suppress receptor activator of NF-κB-ligand gene expression, which is associated with an inhibition of osteoclastogenesis; bone resorption during the remodelling cycle being coupled to bone formation through the receptor activator of NF-κB/receptor activator of NF-κB-ligand/osteoprotegerin system(Reference Chen, Garner and Anderson61).

Finally, PPAR have recently been identified as additional targets of phyto-oestrogen, which can dose dependently activate PPAR and induce divergent effects on adipogenesis and osteogenesis. Dominant ER-mediated effects (increased osteogenesis and decreased adipogenesis) can only be seen at low concentrations of phyto-oestrogens, whereas dominant PPAR-mediated effects are only evident at high concentrations(Reference Dang and Lowik62, Reference Dang and Lowik63). Consequently, divergent actions can be produced in the same cell–tissue system.

Conclusion

Continued awareness and promotion of preventive health services relevant to issues of post-menopausal women will certainly contribute to achieving a healthier population. Thus, the use of phyto-oestrogens is receiving great scrutiny for the purpose of both enhancing the health of tissue and preventing several common diseases, including osteoporosis. However, the limits of the information obtained so far must be appreciated and more data is needed before health professionals can actively advocate the increased consumption of such compounds. Indeed, it will be important to further characterize their physiological effects and margins of safety. In targeting bone health the main issue that remains to be resolved is the effect of phyto-oestrogens in terms of bone strength (which is the ultimate hallmark of bone quality).

References

1.Cooper, C, Campion, G & Melton, LJ (1992) Hip fractures in the elderly: a world-wide projection. Osteoporos Int 2, 285289.CrossRefGoogle ScholarPubMed
2.Raisz, LG (2005) Pathogenesis of osteoporosis: concepts, conflicts, and prospects. J Clin Invest 115, 33183325.CrossRefGoogle ScholarPubMed
3.Albright, F & Reifenstein, EC (1948) Metabolic bone disease; osteoporosis. The Parathyroid Glands and Metabolic Bone Disease, pp. 145204 [Albright, F and Reifenstein, EC editors]. Baltimore, MD: Williams & Wilkins.Google Scholar
4.Manolagas, SC (2000) Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev 21, 115137.Google ScholarPubMed
5.Gallagher, JC (2001) Role of estrogens in the management of postmenopausal bone loss. Rheum Dis Clin North Am 27, 143162.CrossRefGoogle ScholarPubMed
6.Hulley, S, Grady, D, Bush, T, Furberg, C, Herrington, D, Riggs, B & Vittinghoff, E (1998) Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA 280, 605613.CrossRefGoogle ScholarPubMed
7.Rossouw, JE, Anderson, GL, Prentice, RL et al. (2002) Women's health initiative. Risks and benefits of estrogen plus progestin in healthy post menopausal women. JAMA 288, 321333.Google Scholar
8.Beral, V, Million Women Study Collaborators, Bull, D, Green, J & Reeves, G (2007) Ovarian cancer and hormone replacement therapy in the Million Women Study. Lancet 369, 17031710.CrossRefGoogle ScholarPubMed
9.Moggs, JG & Orphanides, G (2004) The role of chromatin in molecular mechanisms of toxicity. Toxicol Sci 80, 218224.CrossRefGoogle ScholarPubMed
10.Axelson, M, Sjovall, J, Gustafsson, BE & Setchell, KD (1982) Origin of lignans in mammals and identification of a precursor from plants. Nature 298, 659660.CrossRefGoogle ScholarPubMed
11.Miksicek, RJ (1993) Commonly occurring plant flavonoids have estrogenic activity. Mol Pharmacol 44, 3743.Google ScholarPubMed
12.Setchell, KD (1998) Phytoestrogens: the biochemistry, physiology, and implications for human health of soy isoflavones. Am J Clin Nutr 68, Suppl., 1333S1346S.Google ScholarPubMed
13.Adlercreutz, H & Mazur, W (1997) Phyto-oestrogens and Western diseases. Ann Med 29, 95120.CrossRefGoogle ScholarPubMed
14.l'Agence Française de la Sécurité Sanitaires des Aliments (2005) Sécurité et Bénéfices des Phyto-estrogènes Apportés par l'Alimentation – Recommandations (The Safety and Benefits of Dietary Phyto-oestrogens – Recommendations). Maisons-Alfort, France: AFSSA.Google Scholar
15.Balk, E, Chung, M, Chew, P et al. (2005) Effects of Soy on Health Outcomes. Evidence Report/Technology Assessment no. 126. AHRQ Publication 05-E024–2. Rockville, MD: AHRQ.CrossRefGoogle Scholar
16.Brynin, R (2002) Soy and its isoflavones: a review of their effects on bone density. Alt Med Rev 7, 317327.Google ScholarPubMed
17.Branca, F & Lorenzetti, S (2005) Health effects of phytoestrogens. Forum Nutr 57, 100111.CrossRefGoogle Scholar
18.Ososki, AL & Kennelly, EJ (2003) Phytoestrogens: a review of the present state of research. Phytother Res 17, 845869.CrossRefGoogle ScholarPubMed
19.Ise, R, Han, D, Takahashi, Y, Terasaka, S, Inoue, A, Tanji, M & Kiyama, R (2005) Expression profiling of the estrogen responsive genes in response to phytoestrogens using a customized DNA microarray. FEBS Lett 579, 17321740.CrossRefGoogle ScholarPubMed
20.Moggs, JG, Ashby, J, Tinwell, H, Lim, FL, Moore, DJ, Kimber, I & Orphanides, G (2004) The need to decide if all estrogens are intrinsically similar. Environ Health Perspect 112, 11371142.CrossRefGoogle ScholarPubMed
21.Coxam, V (2003) Prevention of osteopenia by phyto-oestrogens: animal studies. Br J Nutr 89, Suppl., S75S85.CrossRefGoogle ScholarPubMed
22.Adlercreutz, H, Yamada, T, Wähälä, K & Watanabe, S (1999) Maternal and neonatal phytoestrogens in Japanese women during birth. Am J Obstet Gynecol 180, 737743.CrossRefGoogle ScholarPubMed
23.Whitten, PL & Patisaul, HB (2001) Cross-species and interassay comparisons of phytoestrogen action. Environ Health Perspect 109, Suppl. 1, 520.Google ScholarPubMed
24.Coxam, V & Gerber, M (2005) Effet des phytoestrogènes sur l'ostéoporose. In Sécurité et Bénéfices des Phyto-estrogènes Apportés par l'Alimentation – Recommandations (The Safety and Benefits of Dietary Phyto-oestrogens – Recommendations), pp. 221249. Maisons-Alfort, France: AFSSA.Google Scholar
25.Jones, G, Dwyer, T, Hynes, K, Dalais, FS, Parameswaran, V & Greenaway, TM (2003) A randomized controlled trial of phytoestrogen supplementation, growth and bone turnover in adolescent males. Eur J Clin Nutr 57, 324327.CrossRefGoogle ScholarPubMed
26.Ho, SC, Guldan, GS, Woo, J, Yu, R, Tse, MM, Sham, A & Cheng, J (2005) A prospective study of the effects of 1-year calcium-fortified soy milk supplementation on dietary calcium intake and bone health in Chinese adolescent girls aged 14 to 16. Osteoporos Int 16, 19071916.CrossRefGoogle ScholarPubMed
27.Anderson, JJ, Chen, X, Boass, A, Symons, M, Kohlmeier, M, Renner, JB & Garner, SC (2002) Soy isoflavones: no effects on bone mineral content and bone mineral density in healthy, menstruating young adult women after one year. J Am Coll Nutr 21, 388393.CrossRefGoogle Scholar
28.Spence, LA, Lipscomb, ER, Cadogan, J, Martin, B, Wastney, ME, Peacock, M & Weaver, CM (2005) The effect of soy protein and soy isoflavones on calcium metabolism in postmenopausal women: a randomized crossover study. Am J Clin Nutr 81, 916922.CrossRefGoogle ScholarPubMed
29.Roughead, ZK, Hunt, JR, Johnson, LK, Badger, TM & Lykken, GI (2005) Controlled substitution of soy protein for meat protein: effects on calcium retention, bone, and cardiovascular health indices in postmenopausal women. J Clin Endocrinol Metab 90, 181189.CrossRefGoogle ScholarPubMed
30.Messina, M, Ho, S & Alekel, DL (2004) Skeletal benefits of soy isoflavones: a review of the clinical trial and epidemiologic data. Curr Opin Clin Nutr Metab Care 7, 649658.CrossRefGoogle ScholarPubMed
31.Cassidy, A, Albertazzi, P, Lise Nielsen, I et al. (2006) Critical review of health effects of soyabean phyto-oestrogens in post-menopausal women. Proc Nutr Soc 65, 7692.CrossRefGoogle ScholarPubMed
32.Weaver, CM & Cheong, JM (2005) Soy isoflavones and bone health: the relationship is still unclear. J Nutr 135, 12431247.CrossRefGoogle ScholarPubMed
33.Reinwald, S & Weaver, CM (2006) Soy isoflavones and bone health: a double-edged sword? J Nat Prod 69, 450459.CrossRefGoogle Scholar
34.Zhang, X, Shu, XO, Li, H, Yang, G, Li, Q, Gao, YT & Zheng, W (2005) Prospective cohort study of soy food consumption and risk of bone fracture among postmenopausal women. Arch Intern Med 165, 18901895.CrossRefGoogle ScholarPubMed
35.Setchell, KD, Brown, NM & Lydeking-Olsen, E (2002) The clinical importance of the metabolite equol – a clue to the effectiveness of soy and its isoflavones. J Nutr 132, 35773584.CrossRefGoogle Scholar
36.Axelson, M, Kirk, DN, Farrant, RD, Cooley, G, Lawson, AM & Setchell, KD (1982) The identification of the weak oestrogen equol [7-hydroxy-3-(4′-hydroxyphenyl)chroman] in human urine. Biochem J 201, 353357.CrossRefGoogle Scholar
37.Possemiers, S, Bolca, S, Eeckhaut, E, Depypere, H & Verstraete, W (2007) Metabolism of isoflavones, lignans and prenylflavonoids by intestinal bacteria: producer phenotyping and relation with intestinal community. FEMS Microbiol Ecol 61, 372383.CrossRefGoogle ScholarPubMed
38.Niculescu, MD, Pop, EA, Fischer, LM & Zeisel, SH (2006) Dietary isoflavones differentially induce gene expression changes in lymphocytes from postmenopausal women who form equol as compared with those who do not. J Nutr Biochem 18, 380390.CrossRefGoogle ScholarPubMed
39.Lydeking-Olsen, E, Jensen, JBE, Setchell, KDR, Damhus, M & Jensen, TH (2002) Isoflavone-rich soymilk prevents bone-loss in the lumbar spine of postmenopausal women. A 2 year study. J Nutr 132, 581SAbstr.Google Scholar
40.Wu, J, Oka, J, Higuchi, M et al. (2006) Cooperative effects of isoflavones and exercise on bone and lipid metabolism in postmenopausal Japanese women: a randomized placebo-controlled trial. Metabolism 55, 423433.CrossRefGoogle Scholar
41.Frankenfeld, CL, McTiernan, A, Thomas, WK, LaCroix, K, McVarish, L, Holt, VL, Schwartz, SM & Lampe, JW (2006) Postmenopausal bone mineral density in relation to soy isoflavone-metabolizing phenotypes. Maturitas 53, 315324.CrossRefGoogle ScholarPubMed
42.Fujioka, M, Uehara, M, Wu, J, Adlercreutz, H, Suzuki, K, Kanazawa, K, Takeda, K, Yamada, K & Ishimi, Y (2004) Equol, a metabolite of daidzein, inhibits bone loss in ovariectomized mice. J Nutr 134, 26232627.CrossRefGoogle ScholarPubMed
43.Mathey, J, Mardon, J, Fokialakis, N et al. (2007) Modulation of soy isoflavones bioavailability and subsequent effects on bone health in ovariectomized rats, the case for equol. Osteoporosis Int 18, 671679.CrossRefGoogle Scholar
44.Devareddy, L, Khalil, DA, Korlagunta, K, Hooshmand, S, Bellmer, DD & Arjmandi, BH (2006) The effects of fructo-oligosaccharides in combination with soy protein on bone in osteopenic ovariectomized rats. Menopause 13, 692699.CrossRefGoogle Scholar
45.Ten Bolscher, M, Netelenbos, JC, Barto, R, Van Buuren, LM & Van der Vijgh, WJ (1999) Estrogen regulation of intestinal calcium absorption in the intact and ovariectomized adult rat. J Bone Miner Res 14, 11971202.CrossRefGoogle ScholarPubMed
46.Omi, N, Aoi, S, Murata, K & Ezawa, I (1994) Evaluation of the effect of soybean milk and soybean milk peptide on bone metabolism in the rat model with ovariectomized osteoporosis. J Nutr Sci Vitaminol 40, 201211.CrossRefGoogle Scholar
47.Cotter, AA & Cashman, KD (2005) The effect of two dietary and a synthetic phytoestrogen on transepithelial calcium transport in human intestinal-like Caco-2 cells. Eur J Nutr 44, 7278.CrossRefGoogle Scholar
48.Pie, JE, Park, JH, Park, YH, Ryu, YM, Kim, KN, Suh, SW, Becker, KG, Cho-Chung, YS & Kim, MK (2006) Effect of genistein on the expression of bone metabolism genes in ovariectomized mice using a cDNA microarray. J Nutr Biochem 17, 157164.CrossRefGoogle ScholarPubMed
49.Rickard, DJ, Monroe, DG, Ruesink, TJ, Khosla, S, Riggs, BL & Spelsberg, TC (2003) Phytoestrogen genistein acts as an estrogen agonist on human osteoblastic cells through oestrogen receptors alpha and beta. J Cell Biochem 89, 633646.CrossRefGoogle ScholarPubMed
50.Adlercreutz, H (1998) Evolution, nutrition, intestinal microflora, and prevention of cancer: a hypothesis. Proc Soc Exp Biol Med 217, 241246.CrossRefGoogle ScholarPubMed
51.Kuiper, GG, Lemmen, JG, Carlsson, B, Corton, JC, Safe, SH, van der Saag, PT, van der Burg, B & Gustafsson, JA (1998) Interaction of estrogenic chemicals and phytoestrogens with oestrogen receptor beta. Endocrinology 139, 42524263.CrossRefGoogle Scholar
52.De Wilde, A, Lieberherr, M, Colin, C & Pointillart, A (2004) A low dose of daidzein acts as an ERbeta-selective agonist in trabecular osteoblasts of young female piglets. J Cell Physiol 200, 253262.CrossRefGoogle ScholarPubMed
53.de Wilde, A, Heberden, C, Chaumaz, G, Bordat, C & Lieberherr, M (2006) Signaling networks from Gbeta1 subunit to transcription factors and actin remodeling via a membrane-located ERbeta-related protein in the rapid action of daidzein in osteoblasts. J Cell Physiol 209, 786801.CrossRefGoogle Scholar
54.Kanno, S, Hirano, S & Kayama, F (2004) Effects of the phytoestrogen coumestrol on RANK-ligand-induced differentiation of osteoclasts. Toxicology 203, 211220.CrossRefGoogle ScholarPubMed
55.Jia, TL, Wang, HZ, Xie, LP, Wang, XY & Zhang, RQ (2003) Daidzein enhances osteoblast growth that may be mediated by increased bone morphogenetic protein (BMP) production. Biochem Pharmacol 65, 709715.CrossRefGoogle ScholarPubMed
56.Ge, Y, Chen, D, Xie, L & Zhang, R (2006) Enhancing effect of daidzein on the differentiation and mineralization in mouse osteoblast-like MC3T3-E1 cells. Yakugaku Zasshi 126, 651656.CrossRefGoogle ScholarPubMed
57.Gao, YH & Yamaguchi, M (2000) Suppressive effect of genistein on rat bone osteoclasts: involvement of protein kinase inhibition and protein tyrosine phosphatase activation. Int J Mol Med 5, 261267.Google ScholarPubMed
58.Sliwinski, L, Folwarczna, J, Janiec, W, Grynkiewicz, G & Kuzyk, K (2005) Differential effects of genistein, estradiol and raloxifene on rat osteoclasts in vitro. Pharmacol Rep 57, 352359.Google ScholarPubMed
59.Williams, JP, Jordan, SE, Barnes, S & Blair, HC (1998) Tyrosine kinase inhibitor effects on avian osteoclastic acid transport. Am J Clin Nutr 68, Suppl., 1369S1374S.CrossRefGoogle ScholarPubMed
60.García Palacios, V, Robinson, LJ, Borysenko, CW, Lehmann, T, Kalla, SE & Blair, HC (2005) Negative regulation of RANKL-induced osteoclastic differentiation in RAW264.7 cells by estrogen and phytoestrogens. J Biol Chem 280, 1372013727.CrossRefGoogle ScholarPubMed
61.Chen, XW, Garner, SC & Anderson, JJ (2002) Isoflavones regulate interleukin-6 and osteoprotegerin synthesis during osteoblast cell differentiation via an estrogen-receptor-dependent pathway. Biochem Biophys Res Commun 295, 417422.CrossRefGoogle ScholarPubMed
62.Dang, Z & Lowik, CW (2004) The balance between concurrent activation of ERs and PPARs determines daidzein-induced osteogenesis and adipogenesis. J Bone Miner Res 19, 853861.CrossRefGoogle ScholarPubMed
63.Dang, ZC & Lowik, C (2005) Dose-dependent effects of phytoestrogens on bone. Trends Endocrinol Metab 16, 207213.CrossRefGoogle ScholarPubMed
64.Kardinaal, AFM, Morton, MS, Brüggemann-Rotgans, IEM & Van Beresteijn, ECH (1998) Phyto-oestrogen excretion and rate of bone loss in postmenopausal women. Eur J Clin Nutr 52, 850855.CrossRefGoogle ScholarPubMed
65.Tsuchida, K, Mizushima, S, Toba, M & Soda, K (1999) Dietary soybeans intake and bone mineral density among 995 middle-aged women in Yokohama. J Epidemiol 9, 1419.CrossRefGoogle ScholarPubMed
66.Horiuchi, T, Onouchi, T, Takahashi, M, Ito, H & Orimo, H (2000) Effect of soy protein on bone metabolism in postmenopausal women. Osteoporos Int 11, 721724.CrossRefGoogle Scholar
67.Mei, J, Yeung, SSC & Kung, AWC (2001) High dietary phytoestrogen intake is associated with higher bone mineral density in postmenopausal but not premenopausal women. J Clin Endocrinol Metab 86, 52175221.CrossRefGoogle Scholar
68.Somekawa, Y, Chiguchi, M, Ishibashi, T & Aso, T (2001) Soy intake related to menopausal symptoms, serum lipids, and bone mineral density in postmenopausal Japanese women. Obstet Gynecol 97, 109115.Google ScholarPubMed
69.Kim, MK, Chung, BC, Yu, VY, Nam, JH, Lee, HC, Huh, KB & Lim, SK (2002) Relationship of urinary phyto-oestrogen excretion to BMD in postmenopausal women. Clin Endocrinol 56, 321328.CrossRefGoogle ScholarPubMed
70.Kritz-Silverstein, D & Goodman-Gruen, DL (2002) Usual dietary isoflavone intake, bone mineral density, and bone metabolism in postmenopausal women. J Womens Health Gend Based Med 11, 6978.CrossRefGoogle ScholarPubMed
71.Greendale, GA, FitzGerald, G, Huang, MH, Sternfeld, B, Gold, E, Seeman, T, Sherman, S & Sowers, MF (2002) Dietary soy isoflavones and bone mineral density: results from the study of women's health across the nation. Am J Epidemiol 155, 746754.CrossRefGoogle ScholarPubMed
72.Nagata, C, Shimizu, H, Takami, R, Hayashi, M, Takeda, N & Yasuda, K (2002) Soy product and serum isoflavonoid and estradiol concentrations in relation to bone mineral density in postmenopausal Japanese women. Osteoporos Int 13, 200204.CrossRefGoogle ScholarPubMed
73.Hanna, K, Wong, J, Patterson, C, O'Neill, S & Lyons-Wall, P (2004) Phytoestrogen intake, excretion and markers of bone health in Australian women. Asia Pac J Clin Nutr 13, Suppl., S74.Google Scholar
74.Ikeda, Y, Morita, A, Kajita, E, Kagamimori, S, Kagawa, Y & Yoneshima, H (2006) Intake of fermented soybeans, natto, is associated with reduced bone loss in postmenopausal women: Japanese population-based osteoporosis (JPOS) study. J Nutr 136, 13231328.CrossRefGoogle ScholarPubMed
75.Scambia, G, Mango, D, Signorile, PG, Angeli, RA, Palena, C, Bombardelli, E, Morazzoni, P, Riva, A & Mancuso, S (2000) Clinical effects of a standardised soy extract in postmenopausal women: a pilot study. Menopause 6, 233241.Google Scholar
76.Wangen, KE, Duncan, AM, Merz-Demlow, BE, Xu, X, Marcus, R, Phipps, WR & Kurzer, MS (2000) Effects of soy isoflavones on markers of bone turnover in premenopausal and postmenopausal women. J Clin Endocrinol Metab 85, 30433048.Google ScholarPubMed
77.Upmalis, DH, Lobo, R, Bradley, L, Warren, M, Cone, FL & Lamia, CA (2000) Vasomotor symptom relief by soy isoflavone extract tablets in postmenopausal women: a multicenter, double blind, randomized, placebo-controlled study. Menopause 7, 236242.CrossRefGoogle ScholarPubMed
78.Scheiber, MD, Liu, JH, Subbiah, MTR, Rebar, RW & Setchell, K (2001). Dietary inclusion of whole soy foods results in significant reductions in clinical risk factors for osteoporosis and cardiovascular disease in normal postmenopausal women. Menopause 8, 384392.CrossRefGoogle ScholarPubMed
79.Yamori, Y, Moriguchi, EH, Teramoto, T, Miura, A, Fukui, Y, Honda, K, Fukui, M, Nara, Y, Taira, K & Moriguchi, Y (2002) Soybean isofavones reduce postmenopausal bone resorption in female Japanese immigrants in Brazil: a ten-week study. J Am Coll Nutr 21, 560563.CrossRefGoogle Scholar
80.Uesugi, T, Fukui, Y & Yamori, Y (2002) Beneficial effects of soybean isoflavone supplementation on bone metabolism and serum lipids in postmenopausal Japanese women: a four-week study. J Am Coll Nutr 21, 97102.CrossRefGoogle Scholar
81.Lucas, EA, Wild, RD, Hammond, LJ, Khalil, DA, Jum, S, Daggy, BP, Stoecker, BJ & Arjmandi, BH (2002) Flaxseed improves lipid profile without altering biomarkers of bone metabolism in post-menopausal women. J Clin Endocrinol Metab 87, 15271532.CrossRefGoogle Scholar
82.Arjmandi, BH, Khalil, DA, Smith, BJ, Lucas, EA, Juma, S, Payton, ME & Wild, RA (2003) Soy protein has a greater effect on bone in postmenopausal women not on hormone replacement therapy, as evidenced by reducing bone resorption and urinary calcium excretion. J Clin Endocrinol Metab 88, 10481054.CrossRefGoogle Scholar
83.Dalais, FS, Ebeling, PR, Kotsopoulos, D, McGrath, BP & Teede, HJ (2003) The effects of soy protein containing isoflavones on lipids and indices of bone resorption in postmenopausal women. Clin Endocrinol 58, 704709.CrossRefGoogle ScholarPubMed
84.Harkness, LS, Fiedler, K, Sehgal, AR, Oravec, D & Lerner, E (2004) Decreased bone resorption with soy isoflavone supplementation in postmenopausal women. J Womens Health (Larchmt) 13, 100010007.CrossRefGoogle ScholarPubMed
85.Zittermann, A, Geppert, J, Baier, S, Zehn, N, Gouni-Berthold, I, Berthold, HK, Reinsberg, J & Stehle, P (2004) Short-term effects of high soy supplementation on sex hormones, bone markers, and lipid parameters in young female adults. Eur J Nutr 43, 100108.CrossRefGoogle ScholarPubMed
86.Roudsari, AH, Tahbaz, F, Hossein-Nezhad, A, Arjmandi, B, Larijani, B & Kimiagar, SM (2005) Assessment of soy phytoestrogens' effects on bone turnover indicators in menopausal women with osteopenia in Iran: a before and after clinical trial. Nutr J 4, 30.CrossRefGoogle Scholar
87.Potter, SM, Baun, JA, Teng, H, Stillman, RJ, Shay, NF & Erdman, JW (1998) Soy protein and isoflavones: their effects on blood lipids and bone density in postmenopausal women. Am J Clin Nutr 68, 13751379.CrossRefGoogle ScholarPubMed
88.Dalais, FS, Rice, GE, Wahlqvist, ML, Grehan, M, Murkies, AL, Medley, G, Ayton, R & Strauss, BJG (1998) Effects of dietary phytoestrogens in postmenopausal women. Climateric 1, 124129.CrossRefGoogle ScholarPubMed
89.Alekel, DL, St Germain, A, Peterson, CT, Hanson, KB, Stewart, JW & Toda, T (2000) Isoflavone-rich soy protein isolate attenuates bone loss in the lumbar spine of perimenopausal women. Am J Clin Nutr 72, 844852.CrossRefGoogle ScholarPubMed
90.Clifton-Bligh, PB, Baber, RJ, Fulcher, GR, Nery, ML & Moreton, T (2001) The effect of isoflavones extracted from red clover (Rimostil) on lipid and bone metabolism. Menopause 8, 259265.CrossRefGoogle ScholarPubMed
91.Hsu, CS, Shen, WW, Hsueh, YM & Yeh, SL (2001) Soy isoflavone supplementation in postmenopausal women. J Reprod Med 42, 221226.Google Scholar
92.Chiechi, LM, Secreto, G, D'Amore, M, Fanelli, M, Venturelli, E, Cantatore, F, Laselva, G & Loizzi, P (2002) Efficacy of a soy rich diet in preventing post-menopausal osteoporosis: the Menfis randomised trial. Maturitas 42, 295300.CrossRefGoogle Scholar
93.Vitolins, M, Anthony, M, Lenschik, L, Bland, DR & Burke, GL (2002) Does soy protein and its isoflavones prevent bone loss in peri- and postmenopausal women? Results of a two year randomized clinical trial. J Nutr 132, 582S.Google Scholar
94.Morabito, N, Crisafulli, A, Vergara, C et al. (2002) Effects of genistein and hormone-replacement therapy on bone loss in early postmenopausal women: a randomized double-blind placebo-controlled study. J Bone Min Res 17, 19041912.CrossRefGoogle Scholar
95.Chen, YM, Ho, SC, Lam, SSH, Ho, SSS & Woo, JLF (2003) Soy isoflavones have a favorable effect on bone loss in Chinese postmenopausal women with lower bone mass: a double-blind, randomized, controlled trial. J Clin Endocrinol Metab 88, 47404747.CrossRefGoogle Scholar
96.Gallagher, JC, Satpathy, R, Rafferty, K & Haynatzka, V (2004) The effect of soy protein isolate on bone metabolism. Menopause 11, 290298.CrossRefGoogle ScholarPubMed
97.Lydeking-Olsen, E, Jensen, JBE, Setchell, KDR & Jensen, TH (2004) Soymilk on progesterone for prevention of bone loss. A 2-year randomized, placebo-controlled trial. Eur J Nutr 43, 246257.CrossRefGoogle Scholar
98.Kreijkamp-Kaspers, S, Kok, L, Grobbee, DE, De Haan, EHF, Aleman, A, Lampe, JW & Van der Schouw, YT (2004). Effect of soy protein containing isoflavones on cognitive function, bone mineral density, and plasma lipids in postmenopausal women. A randomized controlled trial. JAMA 292, 6574.CrossRefGoogle ScholarPubMed
99.Atkinson, C, Compson, JE, Day, NE, Dowsett, M & Bingham, SA (2004) The effect of phytoestrogen isoflavones on bone density in women: a double-blind, randomized, placebo-controlled trial. Am J Clin Nutr 79, 326333.CrossRefGoogle Scholar
100.Arjmandi, BH, Lucas, EA, Khalil, DA, Devareddy, L, Smith, BJ, McDonald, J, Arquitt, AB, Payton, ME & Mason, C (2005) One year soy protein supplementation has positive effects on bone formation markers but not bone density in postmenopausal women. Nutr J 4, 8.CrossRefGoogle Scholar
101.Huang, HY, Yang, HP, Yang, HT, Yang, TC, Shieh, MJ & Huang, SY (2006) One-year soy isoflavone supplementation prevents early postmenopausal bone loss but without a dose-dependent effect. J Nutr Biochem 17, 509517.CrossRefGoogle ScholarPubMed
102.Newton, KM, LaCroix, AZ, Levy, L, Li, SS, Qu, P, Potter, JD & Lampe, JW (2006) Soy protein and bone mineral density in older men and women: a randomized trial. Maturitas 55, 270277.CrossRefGoogle ScholarPubMed
103.Wu, J, Oka, J, Tabata, I et al. (2006) Effects of isoflavone and exercise on BMD and fat mass in postmenopausal Japanese women: a 1-year randomized placebo-controlled trial. J Bone Miner Res 21, 780789.CrossRefGoogle ScholarPubMed
104.Ye, YB, Tang, XY, Verbruggen, MA & Su, YX (2006) Soy isoflavones attenuate bone loss in early postmenopausal Chinese women: a single-blind randomized, placebo-controlled trial. Eur J Nutr 45, 327334.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Epidemiological studies in women

Figure 1

Table 2. Intervention trials: effect of soyabean and isoflavones (IF) consumption on bone turnover in women

Figure 2

Table 3. Intervention trials: effect of soyabean and isoflavones (IF) consumption on bone mineral density in post-menopausal women