Skip to main content
World Journal of Diabetes logoLink to World Journal of Diabetes
. 2015 Mar 15;6(2):271–283. doi: 10.4239/wjd.v6.i2.271

Role of phytoestrogens in prevention and management of type 2 diabetes

Mohammad Talaei 1,2, An Pan 1,2
PMCID: PMC4360420  PMID: 25789108

Abstract

Type 2 diabetes (T2D) has become a major public health threat across the globe. It has been widely acknowledged that diet plays an important role in the development and management of T2D. Phytoestrogens are polyphenols that are structurally similar to endogenous estrogen and have weak estrogenic properties. Emerging evidence from pre-clinical models has suggested that phytoestrogens may have anti-diabetic function via both estrogen-dependent and estrogen-independent pathways. In the current review, we have summarized the evidence linking two major types of phytoestrogens, isoflavones and lignans, and T2D from epidemiological studies and clinical trials. The cross-sectional and prospective cohort studies have reported inconsistent results, which may due to the large variations in different populations and measurement errors in dietary intakes. Long-term intervention studies

using isoflavone supplements have reported potential beneficial effects on glycemic parameters in postmenopausal women, while results from short-term small-size clinical trials are conflicting. Taken together, the current evidence from different study designs is complex and inconsistent. Although the widespread use of phytoestrogens could not be recommended yet, habitual consumption of phytoestrogens, particularly their intact food sources like soy and whole flaxseed, could be considered as a component of overall healthy dietary pattern for prevention and management of T2D.

Keywords: Type 2 diabetes, Phytoestrogen, Isoflavone, Lignan, Epidemiological study, Clinical trial


Core tip: Phytoestrogens are a group of polyphenols that are structurally similar to endogenous estrogen. Animal experiments and pre-clinical models have provided strong evidence that phytoestrogens may have anti-diabetic function via both estrogen-dependent and estrogen-independent pathways. A number of epidemiological studies and clinical trials have thus been conducted in different populations linking two major types of phytoestrogens, isoflavones and lignans, to the prevention and management of diabetes. Although the current evidence is complex and inconsistent, habitual consumption of phytoestrogens, particularly their intact food sources, could be considered as a component of overall healthy dietary pattern for prevention and management of diabetes.

INTRODUCTION

Diabetes has become a global public health crisis, and the International Diabetes Federation estimated that 382 million adults were affected by diabetes in 2013, and 5.1 million deaths due to diabetes occurred annually. More than 90% of the diabetes cases are type 2 diabetes (T2D). The global prevalence of T2D has doubled in the last 30 years and is predicted to continue to rise at an alarming rate, and the number is projected to reach 592 million by 2035. The health and economic burden from diabetes is enormous[1]. T2D is a constellation of disorders precipitated by complex and poorly understood interactions between environmental and genetic factors, leading to diminished insulin sensitivity and pancreatic β cell failure. However, diabetes is largely preventable by the adoption of a healthier lifestyle, including normal body weight, not smoking, regular exercise, and a balanced and healthy diet.

It has been widely acknowledged that diet plays an essential role in the development of T2D. Historically, the prevalence of T2D was very low in the traditional Asian society. One hypothesis speculates that the traditional Asian diet, characterized with high intakes of whole grains, large amount of vegetables and fruits, but small portions of meat products, contains many protective components against the development of T2D. Among the many food groups, soybean and soy products as the unique element of traditional Asian diet have aroused much interest because of considerable difference in its intake levels comparing with Western diet[2]. Although there are several potential beneficial compounds (soy protein, dietary fiber, monounsaturated and polyunsaturated fat, vitamins and minerals) in soybean and soy products, one group of polyphenols concentrated in soy products, isoflavones, have been suggested to be beneficial for diabetes prevention and management[2,3].

Isoflavones belong to a group of phytochemicals called phytoestrogens[4,5]. Phytoestrogens are plant-derived compounds that are structurally similar to endogenous estrogen and also have weak estrogenic properties[4,5]. There are two major types of phytoestrogens: isoflavones and lignans[4,5]. The former is concentrated in beans and soy products, and the latter is concentrated in flaxseed, sesames, whole grain and other plant-based foods[4,5]. The other types of phytoestrogens, like prenylated flavonoids and coumestans, are not commonly consumed in daily diet and are not discussed in this article.

In this review, we aimed to examine the current evidence linking phytoestrogens and T2D from epidemiological studies and clinical trials, to explore the potential underlying mechanisms of phytoestrogens’ effect on glucose metabolism from animal and experimental studies, and to propose research priorities for future investigations in this field.

PHYTOESTROGENS

Isoflavones are primarily found in members of leguminosae family and occur in varying amounts in legumes consumed by humans, but soy exceptionally contains the highest isoflavone content[6]. Isoflavone contents of soy food ranges from approximately 0.1 to 5 mg/g of soy protein[7,8]. Asians generally consume very high amount of soy products, and studies have reported that the daily mean intake level of soy protein ranged from 2.0 g in Thailand to 9.6 g in North Korea[9]. Other studies have reported similar results: 5-9 g in Japanese[10] and Chinese[11-13]. The mean isoflavone intake was reported to be from 6 to 75 mg/d in these countries[9], while it was approximately 0.4 mg/d in Spain[14] and Dutch[15] populations, and approximately 0.3 mg/d in the United States population[16].

There are three main soy isoflavones, genistin, daidzin, and glycitin, in which the first two are the major ones available as sugars conjugated form (glycosides) in soybeans and most soy foods in Asian cuisines[4,5]. These biologically inactive forms are hydrolyzed in intestinal wall by the bacterial β-glucosidases and converted into the corresponding bioactive aglycones, daidzein and genistein, which then could be absorbed by intestine[7]. After initial hydrolysis of the glucoside moiety in colon, daidzein can be further metabolized to equol by colonic bacteria. In addition to the conversion by intestinal microflora, genistin and daidzin can also be converted into bioactive forms by in vitro fermentation that is common in traditional Asian methods of preparing soy foods[8]. The blood isoflavone concentration would be in the nanomolar range (< 40 nmol/L) in people who do not eat soy food, and can be increased to micromolar range by acute ingestion of dietary soy. Isoflavones and their metabolites are rapidly excreted in urine with a half-life of about 9 h for daidzein and 7 h for genistein[7].

The other type of phytoestrogens, plant lignans, are more ubiquitous than isoflavones, and the common food sources include oilseeds (flaxseed, sesame, soy, rapeseed), whole-grain cereals (wheat, oats, rye), and various vegetables[17,18]. Cereal fiber and wholegrain foods are among few food groups with established preventive effect for T2D[19], and lignans may be partially responsible for protective effects of dietary fiber complex[20]. Studies have suggested the use of urinary lignan excretion as a marker for fiber and whole grain intake[21,22]. Plant lignans (secoisolariciresinol and matairesinol) are converted to mammalian lignans, enterolactone and enterodiol, by mammalian gut microflora, and enterodiol can also be further oxidized to enterolactone[4,5]. Like isoflavones, the main factors influencing circulating concentration of enterolactone are the food contents of lignans and microflora function[20].

Because of the lack of complete databases of dietary phytoestrogens, large variations of phytoestrogen contents of foods, and comprehensive metabolism pathways influencing circulating concentrations of phytoestrogens, studies have started to use objectively measured blood or urinary phytoestrogen concentrations as a good indicator of dietary intake[22-24]. In Asians with high variations of soy intakes, studies have reported a reasonably well correlation between urinary concentrations of isoflavone metabolites and dietary soy intakes (mostly assessed by food-frequency questionnaires), using morning spot urine[25], or overnight urine samples[26]. This was consistently observed in Japanese[27] and other populations as well[24]. Some studies in United States populations also confirmed that urinary concentrations of isoflavone metabolites are reasonable options for assessing isoflavone intake in epidemiologic studies[28-30]. Studies also suggested that urine samples performed better than serum samples for correlating with dietary intakes[31].

For lignans, studies in Western populations indicated that enterolactone concentrations in overnight urine samples moderately correlated with fruit and vegetables intake[32], concentrations of enterodiol and enterolactone in spot urines were significantly correlated with dietary intakes of fiber[32,33], vegetables and rye products[34]. Few studies have been performed to estimate the urinary lignan metabolites in Asian populations. A small study in 19 Japanese adults found that concentration of urinary lignan metabolites was about one third of isoflavone metabolites, and was correlated with intakes of green and yellow vegetables, pulses and beans[27]. A study in 75 Korean postmenopausal women found that the concentration of lignan metabolites in 24-h urine samples was about half of isoflavone metabolites[35], similar results were found in a study among 68 Chinese T2D patients using first morning urine samples[36]. In another large cross-sectional study of 2165 middle-aged and elderly Chinese women, despite that the concentrations of lignan metabolites were substantially lower compared to isoflavone metabolites in spot urines, the urinary enterodiol was higher than and enterolactone was similar to that among United States women of comparable age[12]. This was observed in another study that collected urine samples from several Asian countries (Japan, Vietnam, India, and Cambodia) and United States[37]: high concentrations of isoflavone metabolites were detected in urine samples from Japan and Vietnam, while the concentrations in urine samples from Cambodia and India were much lower and comparable to that found in United States samples; the differences between urinary lignan metabolites were relatively small among samples from the five countries.

RECENT HUMAN STUDIES LINKING PHYTOESTROGENS TO DIABETES AND GLUCOSE HOMEOSTASIS

The epidemiological studies on the relation between phytoestrogens and risk of T2D or diabetes biomarkers are shown in Table 1. We have described the findings by study designs as below.

Table 1.

Epidemiological studies on the relation between phytoestrogens (lignans or isoflavones) and risk of diabetes or diabetes biomarkers

Ref. Ethnicity Population Sample size, total (outcome) Mean follow-up years Main exposures Outcome Exposure level (mean or median) Maximum effect (highest vs ref.)
Cross-sectional study
Goodman-Gruen et al[39] Mix1 Postmenopausal women, aged 45-74 yr 208 - Dietary isoflavones Diabetes biomarkers 4.4 mg/d genistein (mean) Inverse with 2-h insulin (β = -0.2); not significant for FG and insulin
Yang et al[38] Chinese Women aged 40-70 yr 39385 (323) - Tofu and other soy products Glycosuria 9 g/d soy protein Inverse association in postmenopausal women
van der Schouw et al[42] Caucasian Men aged 47-83 yr 468 - Dietary lignans and isoflavones Diabetes biomarkers Approximately 1 mg/d total phytoestrogens Inverse association of lignans with fasting insulin and C-peptide; no significant association with isoflavones
Pan et al[11] Chinese Men and women aged 50-70 yr 2811 - Dietary soy protein Hyperglycemia (FG ≥ 5.6 mmol/L) 7.8 g/d soy protein Increased odds in men, but not in women
Shi et al[41] Mix1 Pregnant women aged 28 yr 299 - Urinary isoflavones Diabetes biomarkers 502 mg/g creatinine Inverse association with FG, insulin, and HOMA-IR
Longitudinal study
Villegas et al[44] Chinese Women aged 40-70 yr 64191 (896) 4.6 Soy protein, soybeans, soy products T2D 7.7 g/d soy protein Inverse association with soybeans; inverse but not significant relation with soy protein or other products
Nanri et al[46] Japanese Men and women aged 45-75 yr 59791 (1114) 5 Soy products, daidzein, genistein T2D Approximately 73 g/d soy products, approximately 23 mg/d genistein, and 14.5 mg/d daidzein No significant association
Morimoto et al[47] Mix2 Men and women aged 45-75 yr 75344 (8564) 14 Soy products T2D Approximately 14.5 g/d in Japanese, approximately 8 g/d in Hawaiians, and 0 g/d in Caucasians A modest increased risk in men and women
Mueller et al[45] Chinese Men and women aged 45-74 yr 43176 (2252) 5.7 Isoflavones, unsweetened and sweetened soy products T2D Approximately 5.2 g/d for soy protein, 15.8 mg/d for soy isoflavones Inverse association for soy isoflavones and unsweetened soy products, while increased risk for sweetened soybean drinks
Zamora-Ros et al[48] European whites Men and women with mean age 52.4 yr 11559 cases and 15258 subcohort, case-cohort design Approximately 12 Dietary isoflavones and lignans T2D 0.9 mg/d isoflavones, 1.4 mg/d lignans No significant association for isoflavones and lignans
Sun et al[49] Caucasian Women aged 65.6 yr from NHS and 45.4 from NHS II 1107 cases and 1107 controls, nested case-control design Approximately 6 Urinary lignin metabolites (enterodiol and enterolactone) T2D 2.2 μmol/g creatinine for NHS women, and 1.9 μmol/g creatinine for NHS II women Inverse association and odds ratio 0.64 (95%CI: 0.45-0.91) comparing extreme quartiles
1

Mostly non-Hispanic whites;

2

Caucasian, Japanese American, and Native Hawaiian. FG: Fasting glucose level; NHS: Nurses’ Health Study; T2D: Type 2 diabetes; HOMA-IR: Homeostatic model assessment of insulin resistance.

Cross-sectional study evidence

Several cross-sectional studies have assessed the association between soy protein and isoflavone intakes and diabetes related markers. In the Shanghai Women’s Health study of 39385 women aged 40-70 years, it was observed that soy protein intake was inversely associated with glycosuria, an important indicator of diabetes, but only in normal weight postmenopausal women[38]. However, in another study among 2811 Chinese adults, soy protein intake was significantly associated with increased odds of hyperglycemia in men, but null association in postmenopausal women[11]. The median soy protein intake was around 8 g/d in both studies[11,38]. The increased odds of hyperglycemia in men could be a chance finding, and residual confounding and reverse causation are possible in the cross-sectional studies. The sex-specific effects may also linked to the estrogen-like activity of isoflavones[4,5], but the underlying mechanisms are complex and unclear[11]. In a study of 208 American postmenopausal women who ate much lower levels of soy foods, genistein intake was significantly associated with 2-h post challenge insulin concentrations, but not fasting or 2-h glucose concentrations[39]. This suggested that isoflavones may have direct effect on β-cell function and insulin secretion, which is supported by experimental studies[40]. Among 299 pregnant women who participated in the United States NHANES 2001-2008 surveys, Shi et al[41] found that urinary concentrations of total isoflavone metabolites were inversely associated with fasting glucose, insulin and homeostatic model assessment of insulin resistance (HOMA-IR).

Consumption of soy products is generally low in Western diet leading to modest effect of isoflavones on metabolic markers, while lignans may be the major form of phytoestrogens and exert a stronger effect. Dietary lignan intake was inversely associated with fasting insulin and C-peptide in 468 United States men, but the association was not found for isoflavones[42]. In the Framingham Offspring Study with 939 postmenopausal women in United States, high intake of phytoestrogens was associated with a favorable metabolic cardiovascular risk profile (waist-to-hip ratio, triglyceride and overall metabolic score), with stronger association for lignans compared to isoflavones[43]. No study has been conducted so far to investigate the cross-sectional relation between lignans and diabetes risk markers in Asian populations.

Prospective study evidence

A few larger prospective cohort studies have been conducted to investigate the relation between soy food consumption and risk of incident T2D in different populations. In a study with an average 4.6 years of follow-up among Chinese women from the Shanghai Women’s Health Study, Villegas et al[44] reported that soybean and soymilk intakes were significantly associated with a lower risk of incident T2D, while soy protein and other soy products were related to a trend of reduced risk, although not statistically significant. In another large prospective study in Chinese population, the Singapore Chinese Health Study, Mueller et al[45] pointed out that consumption of unsweetened soy products was inversely associated with T2D risk in a graded fashion (P for trend = 0.02), while consuming sweetened soybean drink was positively associated with T2D risk. The findings underline the importance of food context and preparation method. Furthermore, after full adjustment including sweetened soy items, the authors observed a marginally significant inverse association between intake of isoflavones and T2D (relative risk comparing extreme quintile: 0.76; 95%CI: 0.58-1.00; P for trend = 0.08). In a large-scale study in middle-aged and elderly Japanese from the Japan Public Health Center-Based Prospective Study, Nanri et al[46] found no significant association between soy products and isoflavones with incident T2D in either men or women. The suggestive protective association in overweight women disappeared when energy-adjusted intake was considered[46]. In the Multiethnic Cohort study in Hawaii with three ethnicities (Caucasian, Japanese American, and Native Hawaiian), Morimoto et al[47] reported a moderately elevated risk of T2D with soy food consumption and risk of T2D during 14 years of follow-up in men and women, particularly in overweight adults. However, the consumption level of soy products was substantially lower compared to that in the Asian populations. In the European populations, the recent EPIC-InterAct case-cohort study in 12403 incident T2D cases and a subcohort of 16154 participants found no significant association between isoflavones and risk of T2D, while a suggestive trend with lignans (the hazard ratio comparing extreme quintiles 0.88; 95%CI: 0.72-1.07; P for trend = 0.12)[48].

Therefore, the current evidence from large longitudinal studies regarding the relation between phytoestrogen and related food sources and incident T2D is still inconsistent. One methodology challenge could be the measurement error of dietary assessment by questionnaire data. This may be due to the incomplete inclusion of phytoestrogen-enriched food items in the questionnaire and lack of comprehensive food composition databases of phytoestrogens. Furthermore, phytoestrogen metabolism and circulating concentrations in human body can be influenced by many other factors in addition to dietary intake. Thus, studies have started to measure blood or urinary phytoestrogens and evaluate the relation with disease outcomes. Recently, a nested cases-control was conducted among 1107 T2D cases and 1107 control subjects from the Nurses’ Health Study (NHS) and NHS II[49]. Urinary concentrations of the lignan metabolites were assayed by liquid chromatography-mass spectrometry. After multivariate adjustment for lifestyle and dietary risk factors of T2D, the odds ratio for T2D was 0.70 (95%CI: 0.53-0.92) for each SD increment of urinary concentrations of total lignan metabolites. The association was seen in both enterolactone [odds ratio comparing the extreme quartiles 0.62 (95%CI: 0.44-0.88), P for trend = 0.003] and enterodiol [odds ratio comparing the extreme quartiles 0.67 (95%CI: 0.48-0.96), P for trend = 0.08]. Thus far, this is the only prospective study using objectively measured phytoestrogen biomarkers to link with diabetes risk. More studies are needed to examine the relation of urinary phytoestrogen excretion and risk of developing T2D in different studies and populations with varying intake levels.

Clinical trial evidence in participants without T2D

A meta-analysis of 24 intervention studies (n = 1518 in total) on soy intake and glycemic control was done including trials published before March 2010[50]. While no significant effect on fasting glucose and insulin was generally observed for soy intake, the authors found 3.85 mg/dL (95%CI: 2.41-5.28) reduction in fasting glucose concentrations in a subgroup analysis of 9 studies that used whole soy foods or soy diets as the intervention regime. No statistically significant association was identified in 8 studies with isoflavone extract (ranged 40 to 132 mg/d isoflavones) or 6 trials with isolated soy protein containing isoflavones as the main intervention. This suggests that other components of soy like soy protein and fiber, polysaccharides, phytosterol, and unsaturated fatty acid or their interactions may play roles in glycemic control in addition to isoflavones. However, the majority of the studies in this meta-analysis had small sample size (ranged from 14 to 203) and short intervention period (ranged 4 to 52 wk). One of the largest studies so far was a 1-year double-blind, randomized, placebo-controlled trial in 203 Chinese postmenopausal women aged 48 to 62 years[51]. They were randomly assigned to receive daily doses of 0 mg (placebo, n = 67), 40 mg (n = 68), and 80 mg (n = 68) isoflavone supplements along with 500 mg calcium in all groups. The mean differences in the changes of fasting glucose between the intervention and placebo groups were -5.2 mg/dL (95%CI: -9.4 to -1.0) and -3.3 mg/dL (95%CI: -7.5 to 0.9), respectively, for the mid-dose and high-dose groups, and the effect was much more significant in women with higher baseline glucose levels[51].

Another meta-analysis of 12 clinical trials conducted before October 2010 focused on the effects of isoflavone supplementation on blood glucose and insulin in non-Asian postmenopausal women[52]. Zhang et al[52] found that isoflavone supplementation significantly reduced fasting glucose by 0.19 mmol/L (95%CI: 0.03-0.34), and this effect was limited to the studies with more than 6-mo period of intervention. The meta-analysis also reported a significant reduction in fasting insulin by 0.94 μU/mL (95%CI: 0.16-1.72). One of the largest and longest studies so far was done in Italian postmenopausal women with osteopenia[53]. Participants were randomly assigned to receive genistein (54 mg/d; n = 198) or placebo (n = 191) for 2 years. Both groups received 500 mg/d calcium carbonate and 400 IU/d vitamin D. Compared with placebo, genistein significantly reduced fasting glucose and insulin as well as HOMA-IR after both 12 and 24 mo of treatment[53].

Since 2010, a few more trials have been published on the effects of isoflavone supplementation on glucose homeostasis. Two long-term (24 mo) clinical trials by the same research group found that daily intake of 40 mg of soy isoflavones together with lifestyle modification (Mediterranean diet and exercise) reduced HOMA-IR compared to lifestyle modification alone among 116 Spanish postmenopausal women with insulin resistance[54], this was confirmed using same study design (except for 80 mg/d of soy isoflavones) among 80 Spanish postmenopausal women[55]. Improvement of fasting glucose and insulin was also reported[55]. Another 1-year clinical trial among 120 postmenopausal women with metabolic syndrome revealed that 54 mg/d genistein supplements (n = 60) significantly reduced HOMA-IR, fasting glucose and insulin compared to placebo (n = 60)[56]. However, the beneficial effects of isoflavones on glucose metabolism were not found in some short-term trials[57-60]. Since S-equol is considered the most biologically active metabolite of isoflavones, a study was specifically designed to evaluate the effects of S-equol on metabolic profiles among 54 Japanese overweight/obese men and women using a cross-over study design[61]. Significant improvement in HbA1c was observed using 10 mg/d S-equol for 12-wk compared to placebo[61].

As for flaxseed and lignans, a meta-analysis found that flaxseed and/or flaxseed lignan intervention significantly improved lipid profiles[62]. Two small cross-over clinical trials in overweight/obese glucose intolerant participants found that flaxseed reduced insulin resistance after 12-wk interventions[63,64]. A large intervention study in 293 Chinese adults with metabolic syndrome found that 30 g/d flaxseed significantly reduced HbA1c and glucose levels among those with central obesity at baseline[65]. A clinical trial in 55 hypercholesterolemic Chinese subjects found that 600 mg/d flaxseed lignan extract significantly lowered fasting glucose, particularly in those with a higher baseline glucose levels[66]. Another cross-over clinical trial in 22 healthy postmenopausal women reported that 500 mg/d flaxseed lignan extract significantly reduced C-reactive protein levels after 6 wk[67]. However, other studies have found null results[68-71].

Taken together, long-term intervention studies using isoflavone supplements have reported potential beneficial effects on glycemic parameters in postmenopausal women[51,53-56], while results from short-term small-size clinical trials are conflicting. Therefore, more high-quality long-term clinical trials are needed in men and premenopausal women, and to investigate the effect of lignans on glucose metabolism in humans.

Clinical trial evidence in patients with T2D

A number of clinical trials have been conducted in T2D patients to investigate the effects of phytoestrogens and related food sources on diabetes management. Jayagopal et al[72] found that 12-wk intervention of 30 g/d soy protein enriched with 132 mg isoflavones significantly reduced HbA1c (-0.6% vs 1.1% in placebo group), fasting insulin (-8.1% vs 9.9% in placebo group), and HOMA-IR (-6.5% vs 14.7% in placebo group) in postmenopausal women with T2D. Another long-term 4-year clinical trial among T2D patients with nephropathy reported a net change of -29 mg/dL in plasma glucose in the intervention group (n = 20; 0.8 g protein/kg body weight with 35% as soy protein, 35% as animal protein and 30% as vegetable protein) compared to the control group (n = 21; 70% as animal protein and 30% as vegetable protein)[73]. However, some small short-term trials among T2D patients failed to observe significant improvement for isoflavone-containing soy protein on glucose, insulin resistance or HbA1c[74-79]. On the other hand, clinical trials among T2D patients have reported improvement in lipid profiles[73,74,77,78,80], kidney function[73,78,81], endothelial function and blood pressures[76].

The effects of other isoflavone-enriched foods in T2D patients have also been tested: a 1-year intervention with 27 g/d flavonoid-enriched chocolate (containing 850 mg flavan-3-ols and 100 mg isoflavones) significantly reduced insulin resistance and improved insulin sensitivity and lipid profile compared to placebo in 93 postmenopausal women with T2D[82]. However, few studies have specifically investigated the effects of purified isoflavones supplements among T2D patients, and the available two interventions found no significant effects on glycemic control and lipid profiles[83,84], but the intervention periods were short (4 and 12 wk) and sample sizes were small (n = 16 and 32).

A few studies of flaxseed or lignans among diabetic patients also found promising results. Daily supplementation with 10 g flaxseed powder for 4 wk decreased fasting glucose by 19.7% and HbA1c by 15.6% in T2D patients[85], and also improved lipid profiles. Similarly, 5 g/d flaxseed gum for 12 wk significantly reduced serum glucose from 154 ± 8 mg/dL to 136 ± 7 mg/dL[86]. Moreover, 360 mg/d lignan for 12 wk slightly decreased HbA1c[36] and C-reactive protein[87], although fasting glucose and insulin and lipid profiles remained unchanged[36]. Another study using 600 mg/d lignan for 3 mo found decreased HbA1c and glucose levels, but the results were not statistically significant after multivariate adjustment[88].

In summary, isoflavone-enriched soy products and lignin-enriched flaxseeds provide promising benefits in glycemic control, lipid profiles and other cardiovascular markers in T2D patients, but the long-term effect of purified isoflavone or lignan supplements remains unknown.

POTENTIAL MECHANISMS LINKING PHYTOESTROGENS AND PREVENTION OF T2D

The potential mechanisms linking phytoestrogens and glucose metabolism and prevention of diabetes have been extensively reviewed elsewhere[3,40,89,90], here we briefly discuss some animal studies and potential mechanisms on this topic.

A study in male C57BL/KsJ-db/db mice found that both genistein (0.02%, w/w) and daidzein (0.02%, w/w) supplements significantly decreased blood glucose and HbA1c levels, and this effect might be due to the suppression of hepatic glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), fatty acid synthase, β-oxidation and carnitine palmitoyltransferase activities[91]. The same effects have been observed in the female non-obese diabetic mice as well, a T1D animal model[92]. Some other studies suggested that isoflavones may exert antidiabetic effect via peroxisome-proliferator activated receptors (PPAR) pathway. In the obese Zucker rats, a T2D model, high-isoflavone soy protein diet improved glucose tolerance relative to low-isoflavone soy protein and casein diets[93]. It was further found that genistein or daidzein significantly increased PPARα- and PPARγ-directed gene expression by 2-4 fold in RAW 264.7 cells[93]. The increased PPARα gene expression was also seen in another study[94]. In streptozotocin-induced diabetic rats, 3-wk genistein supplementation decreased HbA1c levels and G6Pase activity, while increased glucokinase level and antioxidant enzyme activities[95]. In an obese nongenetic T2D mouse model, dietary intake of genistein (250 mg/kg diet) improved hyperglycemia, glucose tolerance, and blood insulin level but did not affect insulin sensitivity, suggesting that genistein may increase the number of insulin-positive β-cells in islets, promote their survival, and preserve them by preventing apoptosis[96]. Numerous studies have suggested that genistein may have direct effects on β-cell proliferation, glucose-stimulated insulin secretion and protection against apoptosis[40]. Meanwhile, some other studies have shown insulin-sensitizing effect of genistein in male and female C57BL/6 mice[97], as well as ovariectomized rats[98].

Secoisolariciresinol diglucoside (SDG), the major dietary lignan in flaxseed, considerably reduced the incidence of diabetes in streptozotocin-induced diabetic rats[99], diabetes-prone BioBreeding rats, a T1D model[100], and ZDF rats, a T2D model[101]. In these experiments, SDG significantly decreased oxidative stress by reducing malondialdehyde and pancreatic-chemiluminescence level. Sesamin, the most abundant lignan in sesame seed, showed hypoglycemic effect in a dose-dependent manner in KK-Ay mice, a T2D model[102]. Sesamin was also found to attenuate vascular dysfunction and oxidative stress in streptozotocin-diabetic rats[103].

The effects of phytoestrogens on glucose metabolism are thought to be via estrogen-dependent pathway and non-estrogen dependent pathways. Estrogens have been shown to modulate lipid and glucose metabolism directly through lipogenesis, lipolysis, and adipogenesis, or indirectly through their effect on central nervous system influencing appetite and energy expenditure[104]. The relationship between endogenous sex hormones and development of T2D has been well established[105,106]. Because of structural similarity, phytoestrogens could act as estrogen agonists or antagonists, depending on the target tissues[107], doses[108-110], and endogenous circulating sex hormone profile[111]. Although the binding affinity to estrogen receptors (ERs) is much lower for phytoestrogens compared to 17β-estradiol[112], the concentration of phytoestrogens in blood is much higher than endogenous estrogens[113], making it still possible to compete with 17β-estradiol to bind the ERs. Therefore, it is hypothesized that phytoestrogens may influence glucose metabolism by directly modulating concentrations of circulating sex hormones, and this estrogenic effects of phytoestrogens have been supported by some human studies[114-119]. Oxidative stress is considered as one of the causes for T2D and phytoestrogens are known to have strong antioxidant activity[120]. For example, SDG, the major dietary lignan[121] and its mammalian metabolites enterodiol and enterolactone[122], were shown to have antioxidant activity even higher than that of vitamin E. Animal experiments found that lignans decreased lipid peroxidation in rats fed with docosahexaenoic acid[123], and flaxseed increased activities of catalase, superoxide dismutase, and peroxidase[124]. Similarly, isoflavones also showed antioxidant activity in vitro[125] and in vivo[95,126]. Several clinical trials in humans also found that high-isoflavone soy products increased antioxidant capacity[127-130].

Phytoestrogens may influence glucose metabolism and insulin resistance through other non-estrogen dependent mechanisms. For example, both lignans and isoflavones were found to suppress the PEPCK gene expression[92,131]. PEPCK enzyme catalyzes the first committed step in hepatic gluconeogenesis, and PEPCK gene transcription is induced by glucagon and glucocorticoids and inhibited by insulin. Thus, suppression of PEPCK gene will improve hyperglycemia through reduced gluconeogenesis[132]. Furthermore, phytoestrogens, mostly isoflavones, activate PPAR and increase the PPARα- and PPARγ-directed gene expression[93,94,133,134], which is implicated in the glucose homeostasis and lipid metabolism. In the yeast model, genistein was shown to be a reversible, slow-binding, non-competitive inhibitor of alpha-glucosidase[135], which breaks down starch and disaccharides to glucose. Therefore, the alpha-glucosidase inhibitors may reduce the postprandial glucose levels by slowing down the carbohydrate digestion and absorption. In the rabbit model, isoflavones were found to inhibit glucose uptake into rabbit intestinal brush border membrane vesicles in vitro[136]. Genistein also directly acted on pancreatic β-cells, leading to activation of the cAMP/PKA signaling cascade to increases rapid glucose-stimulated insulin secretion[137]. The increased insulin secretion was also reported elsewhere[138-140]. Other studies have found that isoflavones may inhibit tyrosine-specific protein kinases[141], induce adiponectin, leptin and GLUT4 gene expressions in 3T3-L1 adipocytes[142]; promote postprandial carbohydrate oxidation and energy expenditure[143], and protect against high glucose-induced pancreatic cell damage through ERβ and Bcl-2 dependent pathways[144].

IMPLICATIONS AND RECOMMENDATIONS FOR FUTURE STUDIES

As summarized in this review, there has been a long-lasting interest to examine the relation of phytoestrogens and related food sources with diabetes risk. Although the current evidence is promising, there are some knowledge gaps that should be addressed in future investigations.

The food composition databases of phytoestrogens have become the major concern in many epidemiological studies on the relation between phytoestrogens and diabetes risk. The phytoestrogen contents vary dramatically in different food items, and are also influenced by the geographic location, harvest time, and food preparation methods, etc. Therefore, it is urgent to establish accurate, up-to-date, and comprehensive databases in different countries. Particularly for lignans, there is a lack of databases available for research. To the best of our knowledge, there has been no prospective longitudinal study in Asian population investigating habitual intake of lignans and risk of developing T2D.

More prospective studies are needed to use objective biomarkers of phytoestrogens exposure, e.g., urinary excretion concentrations. One methodology challenge of the dietary assessment by questionnaire data is the large measurement error from incomplete inclusion of phytoestrogen-enriched food items in the questionnaire and lack of comprehensive food composition databases. Furthermore, phytoestrogen metabolism and circulating concentrations in human body can be influenced by many other factors (e.g., bioavailability and microflora function) in addition to dietary intake. In addition, phytoestrogen biomarker measurements can be easily done in epidemiological studies with archived biospecimen samples. Some large cohort studies have started to measure urinary concentrations of phytoestrogens and evaluate the relation with disease outcomes, but more investigations in different populations are still warranted. In these studies, repeated measures of phytoestrogen biomarkers are recommended to reduce measurement errors and address the issue of changes over time.

The results from clinical trials of the effects of phytoestrogens on glucose homeostasis are conflicting. Many trials have the limitations of small sample size and short intervention duration. Several recent trails in large sample size (n > 100) and longer duration (≥ 1 year) have produced more consistent and promising evidence to support the use of phytoestrogens. However, those trials were all in postmenopausal women and used isoflavones as the intervention supplements; thus, more high-quality long-term clinical trials are needed in men and premenopausal women, and to investigate the effect of lignans on glucose metabolism. Furthermore, clinical trials in T2D patients have supported the use of isoflavone-enriched soy products and lignin-enriched flaxseed for glycemic and lipid control, but whether the beneficial effects are due to phytoestrogens or other active components in soy or flaxseed remains unclear. Therefore, long-term and high-quality trials using purified phytoestrogen supplements are necessary to explore the possibility of their routine use for diabetes management.

Some studies hypothesized that the observed variations in effect of isoflavones on osteoporosis, cardiovascular disease, or some cancers could be attributed to the equol production ability in human[145]. More investigations in epidemiological studies and clinical trials are needed to test whether this hypothesis is also true for T2D. Furthermore, some studies found that the effects of phytoestrogens on lipid profile[146], endometrial cancer[8], or breast cancer[147] could be modified by various polymorphisms in genes relevant to estrogen or sex hormone binding globulin, like CYP1A1, CYP1B1, and COMT. However, there are few studies that directly assess gene-phytoestrogen interaction for the T2D outcome or glucose metabolism. This line of investigation is important to help understand the potential mechanisms and design personalized interventions.

Although there are many in vivo and in vitro studies to explore the potential pathways for the effects of phytoestrogens, the exact anti-diabetic mechanisms are still unclear. Furthermore, the effective doses used in many experimental studies far exceed the physiological concentrations in human circulation. Thus, it is recommended to consider dosages applicable for human in future animal studies.

CONCLUSION

In conclusion, the current evidence of phytoestrogen and T2D from different study designs are complex and inconsistent. Findings from some high-quality prospective cohort studies and clinical trials are promising, but more studies are needed to fill the aforementioned knowledge gaps. Although the widespread use of phytoestrogens could not be recommended due to the controversies, habitual consumption of phytoestrogens, particularly their intact food sources like soy and whole flaxseed, could be considered as a component of overall healthy dietary pattern for prevention and management of T2D.

Footnotes

P- Reviewer: Guerrero-Romero F, Hamad A S- Editor: Ji FF L- Editor: A E- Editor: Liu SQ

Conflict-of-interest: The authors declare no conflicts of interest regarding this manuscript.

Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/

Peer-review started: September 22, 2014

First decision: October 16, 2014

Article in press: December 17, 2014

References

  • 1.International Diabetes Federation. IDF Diabetes Atlas, Sixth Edition. Brussels, Belgium: International Diabetes Federation; 2013. Available from: http: //www.idf.org/diabetesatlas. [Google Scholar]
  • 2.Kwon DY, Daily JW, Kim HJ, Park S. Antidiabetic effects of fermented soybean products on type 2 diabetes. Nutr Res. 2010;30:1–13. doi: 10.1016/j.nutres.2009.11.004. [DOI] [PubMed] [Google Scholar]
  • 3.Bhathena SJ, Velasquez MT. Beneficial role of dietary phytoestrogens in obesity and diabetes. Am J Clin Nutr. 2002;76:1191–1201. doi: 10.1093/ajcn/76.6.1191. [DOI] [PubMed] [Google Scholar]
  • 4.Kurzer MS, Xu X. Dietary phytoestrogens. Annu Rev Nutr. 1997;17:353–381. doi: 10.1146/annurev.nutr.17.1.353. [DOI] [PubMed] [Google Scholar]
  • 5.Dixon RA. Phytoestrogens. Annu Rev Plant Biol. 2004;55:225–261. doi: 10.1146/annurev.arplant.55.031903.141729. [DOI] [PubMed] [Google Scholar]
  • 6.Bhagwat SH, David B, Holden JM. USDA-Iowa State University database on the isoflavone content of foods. US Department of Agriculture: 2008. Available from: http: //www.ars.usda.gov/SP2UserFiles/Place/12354500/Data/isoflav/Isoflav_R2.pdf. [Google Scholar]
  • 7.Cederroth CR, Nef S. Soy, phytoestrogens and metabolism: A review. Mol Cell Endocrinol. 2009;304:30–42. doi: 10.1016/j.mce.2009.02.027. [DOI] [PubMed] [Google Scholar]
  • 8.Xiao CW. Health effects of soy protein and isoflavones in humans. J Nutr. 2008;138:1244S–1249S. doi: 10.1093/jn/138.6.1244S. [DOI] [PubMed] [Google Scholar]
  • 9.Messina M, Nagata C, Wu AH. Estimated Asian adult soy protein and isoflavone intakes. Nutr Cancer. 2006;55:1–12. doi: 10.1207/s15327914nc5501_1. [DOI] [PubMed] [Google Scholar]
  • 10.Nagata C. Ecological study of the association between soy product intake and mortality from cancer and heart disease in Japan. Int J Epidemiol. 2000;29:832–836. doi: 10.1093/ije/29.5.832. [DOI] [PubMed] [Google Scholar]
  • 11.Pan A, Franco OH, Ye J, Demark-Wahnefried W, Ye X, Yu Z, Li H, Lin X. Soy protein intake has sex-specific effects on the risk of metabolic syndrome in middle-aged and elderly Chinese. J Nutr. 2008;138:2413–2421. doi: 10.3945/jn.108.097519. [DOI] [PubMed] [Google Scholar]
  • 12.Wu X, Cai H, Gao YT, Dai Q, Li H, Cai Q, Yang G, Franke AA, Zheng W, Shu XO. Correlations of urinary phytoestrogen excretion with lifestyle factors and dietary intakes among middle-aged and elderly Chinese women. Int J Mol Epidemiol Genet. 2012;3:18–29. [PMC free article] [PubMed] [Google Scholar]
  • 13.Talaei M, Koh WP, van Dam RM, Yuan JM, Pan A. Dietary soy intake is not associated with risk of cardiovascular disease mortality in Singapore Chinese adults. J Nutr. 2014;144:921–928. doi: 10.3945/jn.114.190454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zamora-Ros R, Jiménez C, Cleries R, Agudo A, Sánchez MJ, Sánchez-Cantalejo E, Molina-Montes E, Navarro C, Chirlaque MD, María Huerta J, et al. Dietary flavonoid and lignan intake and mortality in a Spanish cohort. Epidemiology. 2013;24:726–733. doi: 10.1097/EDE.0b013e31829d5902. [DOI] [PubMed] [Google Scholar]
  • 15.van der Schouw YT, Kreijkamp-Kaspers S, Peeters PH, Keinan-Boker L, Rimm EB, Grobbee DE. Prospective study on usual dietary phytoestrogen intake and cardiovascular disease risk in Western women. Circulation. 2005;111:465–471. doi: 10.1161/01.CIR.0000153814.87631.B0. [DOI] [PubMed] [Google Scholar]
  • 16.Mink PJ, Scrafford CG, Barraj LM, Harnack L, Hong CP, Nettleton JA, Jacobs DR. Flavonoid intake and cardiovascular disease mortality: a prospective study in postmenopausal women. Am J Clin Nutr. 2007;85:895–909. doi: 10.1093/ajcn/85.3.895. [DOI] [PubMed] [Google Scholar]
  • 17.Thompson LU, Robb P, Serraino M, Cheung F. Mammalian lignan production from various foods. Nutr Cancer. 1991;16:43–52. doi: 10.1080/01635589109514139. [DOI] [PubMed] [Google Scholar]
  • 18.Valsta LM, Kilkkinen A, Mazur W, Nurmi T, Lampi AM, Ovaskainen ML, Korhonen T, Adlercreutz H, Pietinen P. Phyto-oestrogen database of foods and average intake in Finland. Br J Nutr. 2003;89 Suppl 1:S31–S38. doi: 10.1079/BJN2002794. [DOI] [PubMed] [Google Scholar]
  • 19.Ley SH, Hamdy O, Mohan V, Hu FB. Prevention and management of type 2 diabetes: dietary components and nutritional strategies. Lancet. 2014;383:1999–2007. doi: 10.1016/S0140-6736(14)60613-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hallmans G, Zhang JX, Lundin E, Stattin P, Johansson A, Johansson I, Hultén K, Winkvist A, Aman P, Lenner P, et al. Rye, lignans and human health. Proc Nutr Soc. 2003;62:193–199. doi: 10.1079/pns2002229. [DOI] [PubMed] [Google Scholar]
  • 21.Stattin P, Bylund A, Biessy C, Kaaks R, Hallmans G, Adlercreutz H. Prospective study of plasma enterolactone and prostate cancer risk (Sweden) Cancer Causes Control. 2004;15:1095–1102. doi: 10.1007/s10552-004-1480-7. [DOI] [PubMed] [Google Scholar]
  • 22.Rowland I, Faughnan M, Hoey L, Wähälä K, Williamson G, Cassidy A. Bioavailability of phyto-oestrogens. Br J Nutr. 2003;89 Suppl 1:S45–S58. doi: 10.1079/BJN2002796. [DOI] [PubMed] [Google Scholar]
  • 23.Bredsdorff L, Obel T, Dethlefsen C, Tjønneland A, Schmidt EB, Rasmussen SE, Overvad K. Urinary flavonoid excretion and risk of acute coronary syndrome in a nested case-control study. Am J Clin Nutr. 2013;98:209–216. doi: 10.3945/ajcn.112.046169. [DOI] [PubMed] [Google Scholar]
  • 24.Lampe JW. Isoflavonoid and lignan phytoestrogens as dietary biomarkers. J Nutr. 2003;133 Suppl 3:956S–964S. doi: 10.1093/jn/133.3.956S. [DOI] [PubMed] [Google Scholar]
  • 25.Seow A, Shi CY, Franke AA, Hankin JH, Lee HP, Yu MC. Isoflavonoid levels in spot urine are associated with frequency of dietary soy intake in a population-based sample of middle-aged and older Chinese in Singapore. Cancer Epidemiol Biomarkers Prev. 1998;7:135–140. [PubMed] [Google Scholar]
  • 26.Chen Z, Zheng W, Custer LJ, Dai Q, Shu XO, Jin F, Franke AA. Usual dietary consumption of soy foods and its correlation with the excretion rate of isoflavonoids in overnight urine samples among Chinese women in Shanghai. Nutr Cancer. 1999;33:82–87. doi: 10.1080/01635589909514752. [DOI] [PubMed] [Google Scholar]
  • 27.Adlercreutz H, Honjo H, Higashi A, Fotsis T, Hämäläinen E, Hasegawa T, Okada H. Urinary excretion of lignans and isoflavonoid phytoestrogens in Japanese men and women consuming a traditional Japanese diet. Am J Clin Nutr. 1991;54:1093–1100. doi: 10.1093/ajcn/54.6.1093. [DOI] [PubMed] [Google Scholar]
  • 28.Tseng M, Olufade T, Kurzer MS, Wahala K, Fang CY, van der Schouw YT, Daly MB. Food frequency questionnaires and overnight urines are valid indicators of daidzein and genistein intake in U.S. women relative to multiple 24-h urine samples. Nutr Cancer. 2008;60:619–626. doi: 10.1080/01635580801993751. [DOI] [PubMed] [Google Scholar]
  • 29.Chun OK, Chung SJ, Song WO. Urinary isoflavones and their metabolites validate the dietary isoflavone intakes in US adults. J Am Diet Assoc. 2009;109:245–254. doi: 10.1016/j.jada.2008.10.055. [DOI] [PubMed] [Google Scholar]
  • 30.Jaceldo-Siegl K, Fraser GE, Chan J, Franke A, Sabaté J. Validation of soy protein estimates from a food-frequency questionnaire with repeated 24-h recalls and isoflavonoid excretion in overnight urine in a Western population with a wide range of soy intakes. Am J Clin Nutr. 2008;87:1422–1427. doi: 10.1093/ajcn/87.5.1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fraser GE, Franke AA, Jaceldo-Siegl K, Bennett H. Reliability of serum and urinary isoflavone estimates. Biomarkers. 2010;15:135–139. doi: 10.3109/13547500903335927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Krogholm KS, Bysted A, Brantsæter AL, Jakobsen J, Rasmussen SE, Kristoffersen L, Toft U. Evaluation of flavonoids and enterolactone in overnight urine as intake biomarkers of fruits, vegetables and beverages in the Inter99 cohort study using the method of triads. Br J Nutr. 2012;108:1904–1912. doi: 10.1017/S0007114512000104. [DOI] [PubMed] [Google Scholar]
  • 33.Grace PB, Taylor JI, Low YL, Luben RN, Mulligan AA, Botting NP, Dowsett M, Welch AA, Khaw KT, Wareham NJ, et al. Phytoestrogen concentrations in serum and spot urine as biomarkers for dietary phytoestrogen intake and their relation to breast cancer risk in European prospective investigation of cancer and nutrition-norfolk. Cancer Epidemiol Biomarkers Prev. 2004;13:698–708. [PubMed] [Google Scholar]
  • 34.Nurmi T, Mursu J, Peñalvo JL, Poulsen HE, Voutilainen S. Dietary intake and urinary excretion of lignans in Finnish men. Br J Nutr. 2010;103:677–685. doi: 10.1017/S0007114509992261. [DOI] [PubMed] [Google Scholar]
  • 35.Kim MK, Chung BC, Yu VY, Nam JH, Lee HC, Huh KB, Lim SK. Relationships of urinary phyto-oestrogen excretion to BMD in postmenopausal women. Clin Endocrinol (Oxf) 2002;56:321–328. doi: 10.1046/j.1365-2265.2002.01470.x. [DOI] [PubMed] [Google Scholar]
  • 36.Pan A, Sun J, Chen Y, Ye X, Li H, Yu Z, Wang Y, Gu W, Zhang X, Chen X, et al. Effects of a flaxseed-derived lignan supplement in type 2 diabetic patients: a randomized, double-blind, cross-over trial. PLoS One. 2007;2:e1148. doi: 10.1371/journal.pone.0001148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kunisue T, Tanabe S, Isobe T, Aldous KM, Kannan K. Profiles of phytoestrogens in human urine from several Asian countries. J Agric Food Chem. 2010;58:9838–9846. doi: 10.1021/jf102253j. [DOI] [PubMed] [Google Scholar]
  • 38.Yang G, Shu XO, Jin F, Elasy T, Li HL, Li Q, Huang F, Zhang XL, Gao YT, Zheng W. Soyfood consumption and risk of glycosuria: a cross-sectional study within the Shanghai Women’s Health Study. Eur J Clin Nutr. 2004;58:615–620. doi: 10.1038/sj.ejcn.1601855. [DOI] [PubMed] [Google Scholar]
  • 39.Goodman-Gruen D, Kritz-Silverstein D. Usual dietary isoflavone intake is associated with cardiovascular disease risk factors in postmenopausal women. J Nutr. 2001;131:1202–1206. doi: 10.1093/jn/131.4.1202. [DOI] [PubMed] [Google Scholar]
  • 40.Gilbert ER, Liu D. Anti-diabetic functions of soy isoflavone genistein: mechanisms underlying its effects on pancreatic β-cell function. Food Funct. 2013;4:200–212. doi: 10.1039/c2fo30199g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Shi L, Ryan HH, Jones E, Simas TA, Lichtenstein AH, Sun Q, Hayman LL. Urinary isoflavone concentrations are inversely associated with cardiometabolic risk markers in pregnant U.S. women. J Nutr. 2014;144:344–351. doi: 10.3945/jn.113.184069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.van der Schouw YT, Sampson L, Willett WC, Rimm EB. The usual intake of lignans but not that of isoflavones may be related to cardiovascular risk factors in U.S. men. J Nutr. 2005;135:260–266. doi: 10.1093/jn/135.2.260. [DOI] [PubMed] [Google Scholar]
  • 43.de Kleijn MJ, van der Schouw YT, Wilson PW, Grobbee DE, Jacques PF. Dietary intake of phytoestrogens is associated with a favorable metabolic cardiovascular risk profile in postmenopausal U.S.women: the Framingham study. J Nutr. 2002;132:276–282. doi: 10.1093/jn/132.2.276. [DOI] [PubMed] [Google Scholar]
  • 44.Villegas R, Gao YT, Yang G, Li HL, Elasy TA, Zheng W, Shu XO. Legume and soy food intake and the incidence of type 2 diabetes in the Shanghai Women’s Health Study. Am J Clin Nutr. 2008;87:162–167. doi: 10.1093/ajcn/87.1.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mueller NT, Odegaard AO, Gross MD, Koh WP, Yu MC, Yuan JM, Pereira MA. Soy intake and risk of type 2 diabetes in Chinese Singaporeans [corrected] Eur J Nutr. 2012;51:1033–1040. doi: 10.1007/s00394-011-0276-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Nanri A, Mizoue T, Takahashi Y, Kirii K, Inoue M, Noda M, Tsugane S. Soy product and isoflavone intakes are associated with a lower risk of type 2 diabetes in overweight Japanese women. J Nutr. 2010;140:580–586. doi: 10.3945/jn.109.116020. [DOI] [PubMed] [Google Scholar]
  • 47.Morimoto Y, Steinbrecher A, Kolonel LN, Maskarinec G. Soy consumption is not protective against diabetes in Hawaii: the Multiethnic Cohort. Eur J Clin Nutr. 2011;65:279–282. doi: 10.1038/ejcn.2010.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zamora-Ros R, Forouhi NG, Sharp SJ, González CA, Buijsse B, Guevara M, van der Schouw YT, Amiano P, Boeing H, Bredsdorff L, et al. The association between dietary flavonoid and lignan intakes and incident type 2 diabetes in European populations: the EPIC-InterAct study. Diabetes Care. 2013;36:3961–3970. doi: 10.2337/dc13-0877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sun Q, Wedick NM, Pan A, Townsend MK, Cassidy A, Franke AA, Rimm EB, Hu FB, van Dam RM. Gut microbiota metabolites of dietary lignans and risk of type 2 diabetes: a prospective investigation in two cohorts of U.S. women. Diabetes Care. 2014;37:1287–1295. doi: 10.2337/dc13-2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Liu ZM, Chen YM, Ho SC. Effects of soy intake on glycemic control: a meta-analysis of randomized controlled trials. Am J Clin Nutr. 2011;93:1092–1101. doi: 10.3945/ajcn.110.007187. [DOI] [PubMed] [Google Scholar]
  • 51.Ho SC, Chen YM, Ho SS, Woo JL. Soy isoflavone supplementation and fasting serum glucose and lipid profile among postmenopausal Chinese women: a double-blind, randomized, placebo-controlled trial. Menopause. 2007;14:905–912. doi: 10.1097/GME.0b013e318032b2d3. [DOI] [PubMed] [Google Scholar]
  • 52.Zhang YB, Chen WH, Guo JJ, Fu ZH, Yi C, Zhang M, Na XL. Soy isoflavone supplementation could reduce body weight and improve glucose metabolism in non-Asian postmenopausal women--a meta-analysis. Nutrition. 2013;29:8–14. doi: 10.1016/j.nut.2012.03.019. [DOI] [PubMed] [Google Scholar]
  • 53.Atteritano M, Marini H, Minutoli L, Polito F, Bitto A, Altavilla D, Mazzaferro S, D’Anna R, Cannata ML, Gaudio A, et al. Effects of the phytoestrogen genistein on some predictors of cardiovascular risk in osteopenic, postmenopausal women: a two-year randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab. 2007;92:3068–3075. doi: 10.1210/jc.2006-2295. [DOI] [PubMed] [Google Scholar]
  • 54.Llaneza P, Gonzalez C, Fernandez-Iñarrea J, Alonso A, Diaz-Fernandez MJ, Arnott I, Ferrer-Barriendos J. Soy isoflavones, Mediterranean diet, and physical exercise in postmenopausal women with insulin resistance. Menopause. 2010;17:372–378. doi: 10.1097/gme.0b013e3181ba56fa. [DOI] [PubMed] [Google Scholar]
  • 55.Llaneza P, González C, Fernández-Iñarrea J, Alonso A, Díaz F, Pérez-López FR. Soy isoflavones improve insulin sensitivity without changing serum leptin among postmenopausal women. Climacteric. 2012;15:611–620. doi: 10.3109/13697137.2011.631062. [DOI] [PubMed] [Google Scholar]
  • 56.Squadrito F, Marini H, Bitto A, Altavilla D, Polito F, Adamo EB, D’Anna R, Arcoraci V, Burnett BP, Minutoli L, et al. Genistein in the metabolic syndrome: results of a randomized clinical trial. J Clin Endocrinol Metab. 2013;98:3366–3374. doi: 10.1210/jc.2013-1180. [DOI] [PubMed] [Google Scholar]
  • 57.Choquette S, Riesco É, Cormier É, Dion T, Aubertin-Leheudre M, Dionne IJ. Effects of soya isoflavones and exercise on body composition and clinical risk factors of cardiovascular diseases in overweight postmenopausal women: a 6-month double-blind controlled trial. Br J Nutr. 2011;105:1199–1209. doi: 10.1017/S0007114510004897. [DOI] [PubMed] [Google Scholar]
  • 58.Llaneza P, González C, Fernandez-Iñarrea J, Alonso A, Diaz F, Arnott I, Ferrer-Barriendos J. Soy isoflavones, diet and physical exercise modify serum cytokines in healthy obese postmenopausal women. Phytomedicine. 2011;18:245–250. doi: 10.1016/j.phymed.2010.07.011. [DOI] [PubMed] [Google Scholar]
  • 59.Barsalani R, Riesco E, Lavoie JM, Dionne IJ. Effect of exercise training and isoflavones on hepatic steatosis in overweight postmenopausal women. Climacteric. 2013;16:88–95. doi: 10.3109/13697137.2012.662251. [DOI] [PubMed] [Google Scholar]
  • 60.Kim J, Lee H, Lee O, Lee KH, Lee YB, Young KD, Jeong YH, Choue R. Isoflavone supplementation influenced levels of triglyceride and luteunizing hormone in Korean postmenopausal women. Arch Pharm Res. 2013;36:306–313. doi: 10.1007/s12272-013-0059-9. [DOI] [PubMed] [Google Scholar]
  • 61.Usui T, Tochiya M, Sasaki Y, Muranaka K, Yamakage H, Himeno A, Shimatsu A, Inaguma A, Ueno T, Uchiyama S, et al. Effects of natural S-equol supplements on overweight or obesity and metabolic syndrome in the Japanese, based on sex and equol status. Clin Endocrinol (Oxf) 2013;78:365–372. doi: 10.1111/j.1365-2265.2012.04400.x. [DOI] [PubMed] [Google Scholar]
  • 62.Pan A, Yu D, Demark-Wahnefried W, Franco OH, Lin X. Meta-analysis of the effects of flaxseed interventions on blood lipids. Am J Clin Nutr. 2009;90:288–297. doi: 10.3945/ajcn.2009.27469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Rhee Y, Brunt A. Flaxseed supplementation improved insulin resistance in obese glucose intolerant people: a randomized crossover design. Nutr J. 2011;10:44. doi: 10.1186/1475-2891-10-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hutchins AM, Brown BD, Cunnane SC, Domitrovich SG, Adams ER, Bobowiec CE. Daily flaxseed consumption improves glycemic control in obese men and women with pre-diabetes: a randomized study. Nutr Res. 2013;33:367–375. doi: 10.1016/j.nutres.2013.02.012. [DOI] [PubMed] [Google Scholar]
  • 65.Wu H, Pan A, Yu Z, Qi Q, Lu L, Zhang G, Yu D, Zong G, Zhou Y, Chen X, et al. Lifestyle counseling and supplementation with flaxseed or walnuts influence the management of metabolic syndrome. J Nutr. 2010;140:1937–1942. doi: 10.3945/jn.110.126300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zhang W, Wang X, Liu Y, Tian H, Flickinger B, Empie MW, Sun SZ. Dietary flaxseed lignan extract lowers plasma cholesterol and glucose concentrations in hypercholesterolaemic subjects. Br J Nutr. 2008;99:1301–1309. doi: 10.1017/S0007114507871649. [DOI] [PubMed] [Google Scholar]
  • 67.Hallund J, Tetens I, Bügel S, Tholstrup T, Bruun JM. The effect of a lignan complex isolated from flaxseed on inflammation markers in healthy postmenopausal women. Nutr Metab Cardiovasc Dis. 2008;18:497–502. doi: 10.1016/j.numecd.2007.05.007. [DOI] [PubMed] [Google Scholar]
  • 68.Cornish SM, Chilibeck PD, Paus-Jennsen L, Biem HJ, Khozani T, Senanayake V, Vatanparast H, Little JP, Whiting SJ, Pahwa P. A randomized controlled trial of the effects of flaxseed lignan complex on metabolic syndrome composite score and bone mineral in older adults. Appl Physiol Nutr Metab. 2009;34:89–98. doi: 10.1139/H08-142. [DOI] [PubMed] [Google Scholar]
  • 69.Dodin S, Cunnane SC, Mâsse B, Lemay A, Jacques H, Asselin G, Tremblay-Mercier J, Marc I, Lamarche B, Légaré F, et al. Flaxseed on cardiovascular disease markers in healthy menopausal women: a randomized, double-blind, placebo-controlled trial. Nutrition. 2008;24:23–30. doi: 10.1016/j.nut.2007.09.003. [DOI] [PubMed] [Google Scholar]
  • 70.Billinsky J, Glew RA, Cornish SM, Whiting SJ, Thorpe LU, Alcorn J, Paus-Jenssen L, Hadjistavropoulos T, Chilibeck PD. No evidence of hypoglycemia or hypotension in older adults during 6 months of flax lignan supplementation in a randomized controlled trial: a safety evaluation. Pharm Biol. 2013;51:778–782. doi: 10.3109/13880209.2013.766220. [DOI] [PubMed] [Google Scholar]
  • 71.Wu JH, Hodgson JM, Puddey IB, Belski R, Burke V, Croft KD. Sesame supplementation does not improve cardiovascular disease risk markers in overweight men and women. Nutr Metab Cardiovasc Dis. 2009;19:774–780. doi: 10.1016/j.numecd.2009.01.003. [DOI] [PubMed] [Google Scholar]
  • 72.Jayagopal V, Albertazzi P, Kilpatrick ES, Howarth EM, Jennings PE, Hepburn DA, Atkin SL. Beneficial effects of soy phytoestrogen intake in postmenopausal women with type 2 diabetes. Diabetes Care. 2002;25:1709–1714. doi: 10.2337/diacare.25.10.1709. [DOI] [PubMed] [Google Scholar]
  • 73.Azadbakht L, Atabak S, Esmaillzadeh A. Soy protein intake, cardiorenal indices, and C-reactive protein in type 2 diabetes with nephropathy: a longitudinal randomized clinical trial. Diabetes Care. 2008;31:648–654. doi: 10.2337/dc07-2065. [DOI] [PubMed] [Google Scholar]
  • 74.Hermansen K, Søndergaard M, Høie L, Carstensen M, Brock B. Beneficial effects of a soy-based dietary supplement on lipid levels and cardiovascular risk markers in type 2 diabetic subjects. Diabetes Care. 2001;24:228–233. doi: 10.2337/diacare.24.2.228. [DOI] [PubMed] [Google Scholar]
  • 75.Liu ZM, Chen YM, Ho SC, Ho YP, Woo J. Effects of soy protein and isoflavones on glycemic control and insulin sensitivity: a 6-mo double-blind, randomized, placebo-controlled trial in postmenopausal Chinese women with prediabetes or untreated early diabetes. Am J Clin Nutr. 2010;91:1394–1401. doi: 10.3945/ajcn.2009.28813. [DOI] [PubMed] [Google Scholar]
  • 76.Clerici C, Nardi E, Battezzati PM, Asciutti S, Castellani D, Corazzi N, Giuliano V, Gizzi S, Perriello G, Di Matteo G, et al. Novel soy germ pasta improves endothelial function, blood pressure, and oxidative stress in patients with type 2 diabetes. Diabetes Care. 2011;34:1946–1948. doi: 10.2337/dc11-0495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Anderson JW, Blake JE, Turner J, Smith BM. Effects of soy protein on renal function and proteinuria in patients with type 2 diabetes. Am J Clin Nutr. 1998;68:1347S–1353S. doi: 10.1093/ajcn/68.6.1347S. [DOI] [PubMed] [Google Scholar]
  • 78.Teixeira SR, Tappenden KA, Carson L, Jones R, Prabhudesai M, Marshall WP, Erdman JW. Isolated soy protein consumption reduces urinary albumin excretion and improves the serum lipid profile in men with type 2 diabetes mellitus and nephropathy. J Nutr. 2004;134:1874–1880. doi: 10.1093/jn/134.8.1874. [DOI] [PubMed] [Google Scholar]
  • 79.Gobert CP, Pipe EA, Capes SE, Darlington GA, Lampe JW, Duncan AM. Soya protein does not affect glycaemic control in adults with type 2 diabetes. Br J Nutr. 2010;103:412–421. doi: 10.1017/S0007114509991802. [DOI] [PubMed] [Google Scholar]
  • 80.Pipe EA, Gobert CP, Capes SE, Darlington GA, Lampe JW, Duncan AM. Soy protein reduces serum LDL cholesterol and the LDL cholesterol: HDL cholesterol and apolipoprotein B: apolipoprotein A-I ratios in adults with type 2 diabetes. J Nutr. 2009;139:1700–1706. doi: 10.3945/jn.109.109595. [DOI] [PubMed] [Google Scholar]
  • 81.Azadbakht L, Esmaillzadeh A. Soy-protein consumption and kidney-related biomarkers among type 2 diabetics: a crossover, randomized clinical trial. J Ren Nutr. 2009;19:479–486. doi: 10.1053/j.jrn.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 82.Curtis PJ, Sampson M, Potter J, Dhatariya K, Kroon PA, Cassidy A. Chronic ingestion of flavan-3-ols and isoflavones improves insulin sensitivity and lipoprotein status and attenuates estimated 10-year CVD risk in medicated postmenopausal women with type 2 diabetes: a 1-year, double-blind, randomized, controlled trial. Diabetes Care. 2012;35:226–232. doi: 10.2337/dc11-1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.González S, Jayagopal V, Kilpatrick ES, Chapman T, Atkin SL. Effects of isoflavone dietary supplementation on cardiovascular risk factors in type 2 diabetes. Diabetes Care. 2007;30:1871–1873. doi: 10.2337/dc06-1814. [DOI] [PubMed] [Google Scholar]
  • 84.Howes JB, Tran D, Brillante D, Howes LG. Effects of dietary supplementation with isoflavones from red clover on ambulatory blood pressure and endothelial function in postmenopausal type 2 diabetes. Diabetes Obes Metab. 2003;5:325–332. doi: 10.1046/j.1463-1326.2003.00282.x. [DOI] [PubMed] [Google Scholar]
  • 85.Mani UV, Mani I, Biswas M, Kumar SN. An open-label study on the effect of flax seed powder (Linum usitatissimum) supplementation in the management of diabetes mellitus. J Diet Suppl. 2011;8:257–265. doi: 10.3109/19390211.2011.593615. [DOI] [PubMed] [Google Scholar]
  • 86.Thakur G, Mitra A, Pal K, Rousseau D. Effect of flaxseed gum on reduction of blood glucose and cholesterol in type 2 diabetic patients. Int J Food Sci Nutr. 2009;60 Suppl 6:126–136. doi: 10.1080/09637480903022735. [DOI] [PubMed] [Google Scholar]
  • 87.Pan A, Demark-Wahnefried W, Ye X, Yu Z, Li H, Qi Q, Sun J, Chen Y, Chen X, Liu Y, et al. Effects of a flaxseed-derived lignan supplement on C-reactive protein, IL-6 and retinol-binding protein 4 in type 2 diabetic patients. Br J Nutr. 2009;101:1145–1149. doi: 10.1017/S0007114508061527. [DOI] [PubMed] [Google Scholar]
  • 88.Barre DE, Mizier-Barre KA, Stelmach E, Hobson J, Griscti O, Rudiuk A, Muthuthevar D. Flaxseed lignan complex administration in older human type 2 diabetics manages central obesity and prothrombosis-an invitation to further investigation into polypharmacy reduction. J Nutr Metab. 2012;2012:585170. doi: 10.1155/2012/585170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Behloul N, Wu G. Genistein: a promising therapeutic agent for obesity and diabetes treatment. Eur J Pharmacol. 2013;698:31–38. doi: 10.1016/j.ejphar.2012.11.013. [DOI] [PubMed] [Google Scholar]
  • 90.Jungbauer A, Medjakovic S. Phytoestrogens and the metabolic syndrome. J Steroid Biochem Mol Biol. 2014;139:277–289. doi: 10.1016/j.jsbmb.2012.12.009. [DOI] [PubMed] [Google Scholar]
  • 91.Ae Park S, Choi MS, Cho SY, Seo JS, Jung UJ, Kim MJ, Sung MK, Park YB, Lee MK. Genistein and daidzein modulate hepatic glucose and lipid regulating enzyme activities in C57BL/KsJ-db/db mice. Life Sci. 2006;79:1207–1213. doi: 10.1016/j.lfs.2006.03.022. [DOI] [PubMed] [Google Scholar]
  • 92.Choi MS, Jung UJ, Yeo J, Kim MJ, Lee MK. Genistein and daidzein prevent diabetes onset by elevating insulin level and altering hepatic gluconeogenic and lipogenic enzyme activities in non-obese diabetic (NOD) mice. Diabetes Metab Res Rev. 2008;24:74–81. doi: 10.1002/dmrr.780. [DOI] [PubMed] [Google Scholar]
  • 93.Mezei O, Banz WJ, Steger RW, Peluso MR, Winters TA, Shay N. Soy isoflavones exert antidiabetic and hypolipidemic effects through the PPAR pathways in obese Zucker rats and murine RAW 264.7 cells. J Nutr. 2003;133:1238–1243. doi: 10.1093/jn/133.5.1238. [DOI] [PubMed] [Google Scholar]
  • 94.Banz WJ, Davis J, Peterson R, Iqbal MJ. Gene expression and adiposity are modified by soy protein in male Zucker diabetic fatty rats. Obes Res. 2004;12:1907–1913. doi: 10.1038/oby.2004.238. [DOI] [PubMed] [Google Scholar]
  • 95.Lee JS. Effects of soy protein and genistein on blood glucose, antioxidant enzyme activities, and lipid profile in streptozotocin-induced diabetic rats. Life Sci. 2006;79:1578–1584. doi: 10.1016/j.lfs.2006.06.030. [DOI] [PubMed] [Google Scholar]
  • 96.Fu Z, Gilbert ER, Pfeiffer L, Zhang Y, Fu Y, Liu D. Genistein ameliorates hyperglycemia in a mouse model of nongenetic type 2 diabetes. Appl Physiol Nutr Metab. 2012;37:480–488. doi: 10.1139/h2012-005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Penza M, Montani C, Romani A, Vignolini P, Pampaloni B, Tanini A, Brandi ML, Alonso-Magdalena P, Nadal A, Ottobrini L, et al. Genistein affects adipose tissue deposition in a dose-dependent and gender-specific manner. Endocrinology. 2006;147:5740–5751. doi: 10.1210/en.2006-0365. [DOI] [PubMed] [Google Scholar]
  • 98.Cao YK, Zhang SF, Zou SE, Xia X. Daidzein improves insulin resistance in ovariectomized rats. Climacteric. 2013;16:111–116. doi: 10.3109/13697137.2012.664831. [DOI] [PubMed] [Google Scholar]
  • 99.Prasad K, Mantha SV, Muir AD, Westcott ND. Protective effect of secoisolariciresinol diglucoside against streptozotocin-induced diabetes and its mechanism. Mol Cell Biochem. 2000;206:141–149. doi: 10.1023/a:1007018030524. [DOI] [PubMed] [Google Scholar]
  • 100.Prasad K. Oxidative stress as a mechanism of diabetes in diabetic BB prone rats: effect of secoisolariciresinol diglucoside (SDG) Mol Cell Biochem. 2000;209:89–96. doi: 10.1023/a:1007079802459. [DOI] [PubMed] [Google Scholar]
  • 101.Prasad K. Secoisolariciresinol diglucoside from flaxseed delays the development of type 2 diabetes in Zucker rat. J Lab Clin Med. 2001;138:32–39. doi: 10.1067/mlc.2001.115717. [DOI] [PubMed] [Google Scholar]
  • 102.Hong L, Yi W, Liangliang C, Juncheng H, Qin W, Xiaoxiang Z. Hypoglycaemic and hypolipidaemic activities of sesamin from sesame meal and its ability to ameliorate insulin resistance in KK-Ay mice. J Sci Food Agric. 2013;93:1833–1838. doi: 10.1002/jsfa.5974. [DOI] [PubMed] [Google Scholar]
  • 103.Baluchnejadmojarad T, Roghani M, Jalali Nadoushan MR, Vaez Mahdavi MR, Kalalian-Moghaddam H, Roghani-Dehkordi F, Dariani S, Raoufi S. The sesame lignan sesamin attenuates vascular dysfunction in streptozotocin diabetic rats: involvement of nitric oxide and oxidative stress. Eur J Pharmacol. 2013;698:316–321. doi: 10.1016/j.ejphar.2012.09.031. [DOI] [PubMed] [Google Scholar]
  • 104.Cooke PS, Naaz A. Role of estrogens in adipocyte development and function. Exp Biol Med (Maywood) 2004;229:1127–1135. doi: 10.1177/153537020422901107. [DOI] [PubMed] [Google Scholar]
  • 105.Ding EL, Song Y, Malik VS, Liu S. Sex differences of endogenous sex hormones and risk of type 2 diabetes: a systematic review and meta-analysis. JAMA. 2006;295:1288–1299. doi: 10.1001/jama.295.11.1288. [DOI] [PubMed] [Google Scholar]
  • 106.Barros RP, Gustafsson JÅ. Estrogen receptors and the metabolic network. Cell Metab. 2011;14:289–299. doi: 10.1016/j.cmet.2011.08.005. [DOI] [PubMed] [Google Scholar]
  • 107.Safe SH, Pallaroni L, Yoon K, Gaido K, Ross S, Saville B, McDonnellc D. Toxicology of environmental estrogens. Reprod Fertil Dev. 2001;13:307–315. doi: 10.1071/rd00108. [DOI] [PubMed] [Google Scholar]
  • 108.Dang ZC. Dose-dependent effects of soy phyto-oestrogen genistein on adipocytes: mechanisms of action. Obes Rev. 2009;10:342–349. doi: 10.1111/j.1467-789X.2008.00554.x. [DOI] [PubMed] [Google Scholar]
  • 109.Dang ZC, Audinot V, Papapoulos SE, Boutin JA, Löwik CW. Peroxisome proliferator-activated receptor gamma (PPARgamma) as a molecular target for the soy phytoestrogen genistein. J Biol Chem. 2003;278:962–967. doi: 10.1074/jbc.M209483200. [DOI] [PubMed] [Google Scholar]
  • 110.Hsieh CY, Santell RC, Haslam SZ, Helferich WG. Estrogenic effects of genistein on the growth of estrogen receptor-positive human breast cancer (MCF-7) cells in vitro and in vivo. Cancer Res. 1998;58:3833–3838. [PubMed] [Google Scholar]
  • 111.Glazier MG, Bowman MA. A review of the evidence for the use of phytoestrogens as a replacement for traditional estrogen replacement therapy. Arch Intern Med. 2001;161:1161–1172. doi: 10.1001/archinte.161.9.1161. [DOI] [PubMed] [Google Scholar]
  • 112.Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, van der Saag PT, van der Burg B, Gustafsson JA. Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology. 1998;139:4252–4263. doi: 10.1210/endo.139.10.6216. [DOI] [PubMed] [Google Scholar]
  • 113.Adlercreutz H, Markkanen H, Watanabe S. Plasma concentrations of phyto-oestrogens in Japanese men. Lancet. 1993;342:1209–1210. doi: 10.1016/0140-6736(93)92188-y. [DOI] [PubMed] [Google Scholar]
  • 114.Nagata C, Inaba S, Kawakami N, Kakizoe T, Shimizu H. Inverse association of soy product intake with serum androgen and estrogen concentrations in Japanese men. Nutr Cancer. 2000;36:14–18. doi: 10.1207/S15327914NC3601_3. [DOI] [PubMed] [Google Scholar]
  • 115.Nagata C, Takatsuka N, Shimizu H, Hayashi H, Akamatsu T, Murase K. Effect of soymilk consumption on serum estrogen and androgen concentrations in Japanese men. Cancer Epidemiol Biomarkers Prev. 2001;10:179–184. [PubMed] [Google Scholar]
  • 116.Wu AH, Stanczyk FZ, Seow A, Lee HP, Yu MC. Soy intake and other lifestyle determinants of serum estrogen levels among postmenopausal Chinese women in Singapore. Cancer Epidemiol Biomarkers Prev. 2002;11:844–851. [PubMed] [Google Scholar]
  • 117.Low YL, Dunning AM, Dowsett M, Folkerd E, Doody D, Taylor J, Bhaniani A, Luben R, Khaw KT, Wareham NJ, et al. Phytoestrogen exposure is associated with circulating sex hormone levels in postmenopausal women and interact with ESR1 and NR1I2 gene variants. Cancer Epidemiol Biomarkers Prev. 2007;16:1009–1016. doi: 10.1158/1055-9965.EPI-06-0899. [DOI] [PubMed] [Google Scholar]
  • 118.Persky VW, Turyk ME, Wang L, Freels S, Chatterton R, Barnes S, Erdman J, Sepkovic DW, Bradlow HL, Potter S. Effect of soy protein on endogenous hormones in postmenopausal women. Am J Clin Nutr. 2002;75:145–153. doi: 10.1093/ajcn/75.1.145. [DOI] [PubMed] [Google Scholar]
  • 119.Pino AM, Valladares LE, Palma MA, Mancilla AM, Yáñez M, Albala C. Dietary isoflavones affect sex hormone-binding globulin levels in postmenopausal women. J Clin Endocrinol Metab. 2000;85:2797–2800. doi: 10.1210/jcem.85.8.6750. [DOI] [PubMed] [Google Scholar]
  • 120.Mitchell JH, Gardner PT, McPhail DB, Morrice PC, Collins AR, Duthie GG. Antioxidant efficacy of phytoestrogens in chemical and biological model systems. Arch Biochem Biophys. 1998;360:142–148. doi: 10.1006/abbi.1998.0951. [DOI] [PubMed] [Google Scholar]
  • 121.Prasad K. Hydroxyl radical-scavenging property of secoisolariciresinol diglucoside (SDG) isolated from flax-seed. Mol Cell Biochem. 1997;168:117–123. doi: 10.1023/a:1006847310741. [DOI] [PubMed] [Google Scholar]
  • 122.Prasad K. Antioxidant Activity of Secoisolariciresinol Diglucoside-derived Metabolites, Secoisolariciresinol, Enterodiol, and Enterolactone. Int J Angiol. 2000;9:220–225. doi: 10.1007/BF01623898. [DOI] [PubMed] [Google Scholar]
  • 123.Ikeda S, Kagaya M, Kobayashi K, Tohyama T, Kiso Y, Higuchi N, Yamashita K. Dietary sesame lignans decrease lipid peroxidation in rats fed docosahexaenoic acid. J Nutr Sci Vitaminol (Tokyo) 2003;49:270–276. doi: 10.3177/jnsv.49.270. [DOI] [PubMed] [Google Scholar]
  • 124.Rajesha J, Murthy KN, Kumar MK, Madhusudhan B, Ravishankar GA. Antioxidant potentials of flaxseed by in vivo model. J Agric Food Chem. 2006;54:3794–3799. doi: 10.1021/jf053048a. [DOI] [PubMed] [Google Scholar]
  • 125.Rüfer CE, Kulling SE. Antioxidant activity of isoflavones and their major metabolites using different in vitro assays. J Agric Food Chem. 2006;54:2926–2931. doi: 10.1021/jf053112o. [DOI] [PubMed] [Google Scholar]
  • 126.Valsecchi AE, Franchi S, Panerai AE, Rossi A, Sacerdote P, Colleoni M. The soy isoflavone genistein reverses oxidative and inflammatory state, neuropathic pain, neurotrophic and vasculature deficits in diabetes mouse model. Eur J Pharmacol. 2011;650:694–702. doi: 10.1016/j.ejphar.2010.10.060. [DOI] [PubMed] [Google Scholar]
  • 127.Azadbakht L, Kimiagar M, Mehrabi Y, Esmaillzadeh A, Hu FB, Willett WC. Dietary soya intake alters plasma antioxidant status and lipid peroxidation in postmenopausal women with the metabolic syndrome. Br J Nutr. 2007;98:807–813. doi: 10.1017/S0007114507746871. [DOI] [PubMed] [Google Scholar]
  • 128.Jenkins DJ, Kendall CW, Garsetti M, Rosenberg-Zand RS, Jackson CJ, Agarwal S, Rao AV, Diamandis EP, Parker T, Faulkner D, et al. Effect of soy protein foods on low-density lipoprotein oxidation and ex vivo sex hormone receptor activity--a controlled crossover trial. Metabolism. 2000;49:537–543. doi: 10.1016/s0026-0495(00)80022-0. [DOI] [PubMed] [Google Scholar]
  • 129.Tikkanen MJ, Wähälä K, Ojala S, Vihma V, Adlercreutz H. Effect of soybean phytoestrogen intake on low density lipoprotein oxidation resistance. Proc Natl Acad Sci USA. 1998;95:3106–3110. doi: 10.1073/pnas.95.6.3106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Mitchell JH, Collins AR. Effects of a soy milk supplement on plasma cholesterol levels and oxidative DNA damage in men--a pilot study. Eur J Nutr. 1999;38:143–148. doi: 10.1007/s003940050055. [DOI] [PubMed] [Google Scholar]
  • 131.Prasad K. Suppression of phosphoenolpyruvate carboxykinase gene expression by secoisolariciresinol diglucoside (SDG), a new antidiabetic agent. Int J Angiol. 2002;11:107–109. [Google Scholar]
  • 132.Quinn PG, Yeagley D. Insulin regulation of PEPCK gene expression: a model for rapid and reversible modulation. Curr Drug Targets Immune Endocr Metabol Disord. 2005;5:423–437. doi: 10.2174/156800805774912962. [DOI] [PubMed] [Google Scholar]
  • 133.Davis J, Higginbotham A, O’Connor T, Moustaid-Moussa N, Tebbe A, Kim YC, Cho KW, Shay N, Adler S, Peterson R, et al. Soy protein and isoflavones influence adiposity and development of metabolic syndrome in the obese male ZDF rat. Ann Nutr Metab. 2007;51:42–52. doi: 10.1159/000100820. [DOI] [PubMed] [Google Scholar]
  • 134.Mezei O, Li Y, Mullen E, Ross-Viola JS, Shay NF. Dietary isoflavone supplementation modulates lipid metabolism via PPARalpha-dependent and -independent mechanisms. Physiol Genomics. 2006;26:8–14. doi: 10.1152/physiolgenomics.00155.2005. [DOI] [PubMed] [Google Scholar]
  • 135.Lee DS, Lee SH. Genistein, a soy isoflavone, is a potent alpha-glucosidase inhibitor. FEBS Lett. 2001;501:84–86. doi: 10.1016/s0014-5793(01)02631-x. [DOI] [PubMed] [Google Scholar]
  • 136.Vedavanam K, Srijayanta S, O’Reilly J, Raman A, Wiseman H. Antioxidant action and potential antidiabetic properties of an isoflavonoid-containing soyabean phytochemical extract (SPE) Phytother Res. 1999;13:601–608. doi: 10.1002/(sici)1099-1573(199911)13:7<601::aid-ptr550>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 137.Liu D, Zhen W, Yang Z, Carter JD, Si H, Reynolds KA. Genistein acutely stimulates insulin secretion in pancreatic beta-cells through a cAMP-dependent protein kinase pathway. Diabetes. 2006;55:1043–1050. doi: 10.2337/diabetes.55.04.06.db05-1089. [DOI] [PubMed] [Google Scholar]
  • 138.Jonas JC, Plant TD, Gilon P, Detimary P, Nenquin M, Henquin JC. Multiple effects and stimulation of insulin secretion by the tyrosine kinase inhibitor genistein in normal mouse islets. Br J Pharmacol. 1995;114:872–880. doi: 10.1111/j.1476-5381.1995.tb13285.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Lu MP, Wang R, Song X, Chibbar R, Wang X, Wu L, Meng QH. Dietary soy isoflavones increase insulin secretion and prevent the development of diabetic cataracts in streptozotocin-induced diabetic rats. Nutr Res. 2008;28:464–471. doi: 10.1016/j.nutres.2008.03.009. [DOI] [PubMed] [Google Scholar]
  • 140.Wagner JD, Zhang L, Shadoan MK, Kavanagh K, Chen H, Tresnasari K, Kaplan JR, Adams MR. Effects of soy protein and isoflavones on insulin resistance and adiponectin in male monkeys. Metabolism. 2008;57:S24–S31. doi: 10.1016/j.metabol.2008.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, Itoh N, Shibuya M, Fukami Y. Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem. 1987;262:5592–5595. [PubMed] [Google Scholar]
  • 142.Fukumitsu S, Aida K, Ueno N, Ozawa S, Takahashi Y, Kobori M. Flaxseed lignan attenuates high-fat diet-induced fat accumulation and induces adiponectin expression in mice. Br J Nutr. 2008;100:669–676. doi: 10.1017/S0007114508911570. [DOI] [PubMed] [Google Scholar]
  • 143.Ishihara K, Oyaizu S, Fukuchi Y, Mizunoya W, Segawa K, Takahashi M, Mita Y, Fukuya Y, Fushiki T, Yasumoto K. A soybean peptide isolate diet promotes postprandial carbohydrate oxidation and energy expenditure in type II diabetic mice. J Nutr. 2003;133:752–757. doi: 10.1093/jn/133.3.752. [DOI] [PubMed] [Google Scholar]
  • 144.Zhong WW, Liu Y, Li CL. Mechanisms of genistein protection on pancreas cell damage in high glucose condition. Intern Med. 2011;50:2129–2134. doi: 10.2169/internalmedicine.50.5320. [DOI] [PubMed] [Google Scholar]
  • 145.Shor D, Sathyapalan T, Atkin SL, Thatcher NJ. Does equol production determine soy endocrine effects? Eur J Nutr. 2012;51:389–398. doi: 10.1007/s00394-012-0331-7. [DOI] [PubMed] [Google Scholar]
  • 146.Vafeiadou K, Hall WL, Williams CM. Does genotype and equol-production status affect response to isoflavones? Data from a pan-European study on the effects of isoflavones on cardiovascular risk markers in post-menopausal women. Proc Nutr Soc. 2006;65:106–115. doi: 10.1079/pns2005483. [DOI] [PubMed] [Google Scholar]
  • 147.Wang Q, Li H, Tao P, Wang YP, Yuan P, Yang CX, Li JY, Yang F, Lee H, Huang Y. Soy isoflavones, CYP1A1, CYP1B1, and COMT polymorphisms, and breast cancer: a case-control study in southwestern China. DNA Cell Biol. 2011;30:585–595. doi: 10.1089/dna.2010.1195. [DOI] [PubMed] [Google Scholar]

Articles from World Journal of Diabetes are provided here courtesy of Baishideng Publishing Group Inc

RESOURCES