Estrogens and Female Liver Health

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Steroids 133 (2018) 38–43

Contents lists available at ScienceDirect

Steroids
journal homepage: www.elsevier.com/locate/steroids

Estrogens and female liver health T


a a,b,⁎
Karen L. Chen , Zeynep Madak-Erdogan
a
Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
b
Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Due to declining estrogen levels during menopause, NAFLD prevalence is higher in postmenopausal women
Microbiome compared to in premenopausal women or in men. Postmenopausal women are more susceptible to weight gain,
Estrogens fat redistribution and dyslipidemia, all major hallmarks of metabolic syndrome associated with increased NAFLD
Botanical estrogens risk. Gut microbiota plays important roles in development of gastrointestinal tract, metabolism and immunity.
Dietary supplements
Host-microbe interactions allows regulation of a wide range of pathways that affect healthy and diseased
Estrogen receptor
physiology. Recent advances in – omics technologies, such as microbiome, transcriptome and metabolome
Metabolism
Menopause analysis, provided evidence that estrogens and intestinal microbiota (IM) can collectively influence obesity,
Non-alcoholic fatty liver disease inflammatory disease, diabetes, and cancers. By understanding underlying mechanisms of estrogens and mi-
crobiota crosstalk, we might design dietary and pharmacological interventions to alleviate the metabolic syn-
drome and NAFLD.

1. Metabolic health of postmenopausal women and hormone The loss of these benefits and the development of obesity after meno-
replacement therapy pause highlights the estrogen receptor, especially the receptor subtype
ERα, as a relevant therapeutic target. These effects, including insulin
The prevalence of obesity among women is increasing, yet there is resistance and altered glucose homeostasis, can be reversed by estradiol
no such significant trend for men. Currently, 70% of postmenopausal (E2) treatment [7,8]. The American Association of Clinical En-
women are overweight or obese. According to 2003–2012 National docrinologist, the American College of Endocrinology [9], the North
Health and Nutrition Examination Survey (NHANES) data, metabolic American Menopause Society [10] and the Endocrine Society [11] re-
syndrome is present in about 34% of the population and the rates of cognize the metabolic benefits of HRT in their guidelines but do not
metabolic syndrome is increasing among postmenopausal women [1,2]. recommend HRT use for prevention or treatment of these symptoms
Metabolic syndrome is defined as a collection of metabolic pathologies due to insufficient data and potential risks. Currently, HRT is only
that increase an individual’s risk of heart disease, diabetes, cancer and prescribed to women who display clinical symptoms, is given at the
nonalcoholic fatty liver disease (NAFLD) [3,4]. National Heart, Lung, lowest dose possible, and is continued only as long as the clinical
and Blood Institute (NHLBI) and the American Heart Association (AHA) symptoms are present. Due to potential risks associated with these
identified risk factors for metabolic syndrome as a fasting glucose products, they are not used long-term or for prevention. Moreover, the
≥100 mg/dL, blood pressure ≥130/85 mm Hg, triglycerides patient needs to be constantly monitored by a general practitioner to
≥150 mg/dL, HDL-C < 40 mg/dL, and waist circumference ≥102 cm catch any problems that might arise due to HRT use. Hence, there re-
in men or ≥88 cm in women [5]. Metabolic syndrome is diagnosed if mains a critical need for safer alternatives that target the signaling
an individual has 3 out of 5 of these risk factors. In addition, having pathways of the estrogen receptor (ER) in ways that maintain the
these symptoms is not associated with mortality in older men, yet postmenopausal health of metabolic tissues without stimulating re-
elevated glucose or low HDL levels are strongly associated with mor- productive tissues.
tality in older women [6]. Current HRT typically consists of an estrogen or estrogen in com-
Estrogens, which are necessary for the development and function of bination with a progestin, and may be taken to relieve hot flashes or to
reproductive tissues, have many beneficial effects in diverse, non-re- treat osteoporosis. Though the initial reason a woman would take HRT
productive target tissues, such as liver, fat pads, bone and vasculature. are usually not metabolic, there are many metabolic benefits to HRT

Abbreviations: ER, estrogen receptor; IM, intestinal microbiota; LPL, lipoprotein lipase; LPS, lipopolysaccharide; ODMA, O-desmethylangolensin; NAFLD, non-alcoholic fatty liver
disease

Corresponding author at: Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 905 S. Goodwin Ave, Urbana, IL 61801, USA.
E-mail address: zmadake2@illinois.edu (Z. Madak-Erdogan).

https://doi.org/10.1016/j.steroids.2017.10.015
Received 6 October 2017; Received in revised form 23 October 2017; Accepted 28 October 2017
Available online 01 November 2017
0039-128X/ © 2017 Elsevier Inc. All rights reserved.
K.L. Chen, Z. Madak-Erdogan Steroids 133 (2018) 38–43

including diminishing insulin resistance and improving glucose home- steatohepatits at 65% and 20% respectively in obese individuals (BMI
ostasis [7,8]. HRT has also been shown to favorably influence liver 30.0–39.9 kg/m2) and 85% and 40% respectively in extremely obese
enzymes and potentially lower liver fat accumulation in women with patients (BMI ≥ 40 kg/m2) [46]. The prevalence of NAFLD is about
type 2 diabetes [12]. However, though HRT has many metabolic ben- 69% in patients with type 2 diabetes mellitus (T2DM) [47]. Risk factors
efits, they have been shown to increase risks of certain cancers in- for NAFLD include dietary fat consumption, insulin resistance, elevated
cluding breast cancer and endometrial cancers [13–16]. Therefore, it is serum free fatty acids, excess visceral adiposity, and pro-inflammatory
essential to find safer alternatives to HRT such as novel and synthetic markers [48]. These factors contribute to the accumulation of excessive
estrogens that can maintain the health of non-reproductive tissues hepatic fat and oxidative stress, that are components of the “two-hit
without stimulating reproductive tissues. hypothesis” that explain disease etiology [49–51]. Therefore, targeting
Selective estrogen receptor modulators (SERMs) are compounds one or both of these factors may be beneficial in treating NAFLD as
that can bind to estrogen receptors and can exert agonistic or antag- there are currently no FDA approved therapies for NAFLD.
onistic activity. Bazedoxifene (BZA) is one such SERM that has estrogen The present Dietary Guidelines indicate that more than 35% of daily
agonist activity in bone and reduces osteoporosis while possibly having energy consumed by Americans is from fat, particularly saturated fats,
antagonistic activity in the breast and uterus [17,18]. In mice, the an- and added sugar [52]. Excess consumption of saturated fat and sugar
tagonistic effects of BZA in the mammary gland can block E2 and in- contributes to increased body fatness and to the progression of chronic,
hibit the growth of tamoxifen sensitive and resistant tumors [19]. In cell low-level inflammation, thus, adversely impacting hepatic health and
culture models conjugated estrogen (CE) was much less potent in in- aggravating chronic diseases such as diabetes and hepatocellular car-
ducing breast cancer cell proliferation, and BZA entirely suppressed this cinogenesis. Hepatic cancer risk increases with the progression into
effect [20]. Although classic HRT uses an estrogen and progestin in NASH, which accounts for 20–25% of patients with NAFLD [53]. Since
combination, replacing progestin with SERMs could more effectively saturated fatty acids are not a priority substrate for β-oxidation, they
prevent side-effects. This new combination of estrogen and SERM has are a poor source of energy, and are directed toward storage in the liver
been described as the tissue-selective estrogen complexes (TSECs). Most [54]. Saturated fatty acids increase endoplasmic reticular stress and free
notably, the TSEC comprised of CE and BZA has been shown to prevent radical production in mitochondria, which promotes the cellular da-
osteoporosis with minimal effects on reproductive organs [21–23]. This mage and eventually, development of NASH [55]. Fructose, which is a
combination was approved by the FDA for the prevention of post- component of sucrose, is a highly lipogenic nutrient, enhancing hepatic
menopausal osteoporosis and the treatment of moderate-to-severe va- triglyceride accumulation through the de novo lipogenesis pathway
somotor symptoms caused by menopause. Recently, the possibility of [56]. Therefore, the components of Westernized diet disrupts lipid
this TSEC in preventing metabolic disorders is beginning to be explored. homeostasis and oxidative balance in the liver and enables the onset
It has been shown that the TSECs prevent visceral adiposity and had a and the progression of NAFLD.
significant impact on body weight increase compared to control in
ovariectomized mice [24,25]. CE + BZA caused a significant decrease 3. Mechanisms of metabolic regulation by estrogens
in both uterine and mesenteric white adipose tissue (WAT), but did not
influence the uterine weight, which CE only and E2 treatments did In humans, there are three major estrogens endogenous to the
[25]. TSEC decreases serum leptin levels, leptin/adiponectin ratio, and human body. These are estrone (E1), 17β-estradiol (E2), and estriol
levels of thiobarbituric acid reactive substances (TBARS) compared to (E3) with E2 being the most potent [57]. Estrogens are derived from
control mice suggesting normalization of serum adipokines and sys- cholesterol by reactions catalyzed by the P450 aromatase [57]. E2 has
temic inflammation [24]. Furthermore, the TSEC decreased hepatic the greatest affinity for estrogen receptors and a higher bioactivity [58].
fatty acid synthase (FAS) enzymatic activity [24] and significant de- Several tissues are able to generate estrogens in humans including the
creases in liver lipid deposition [25] suggesting benefits to liver health. reproductive organs of both males and females, the liver, osteoblasts,
muscle cells, skin, and adipose tissue [59]. In premenopausal women,
2. NAFLD and estrogens the ovaries produce the majority of the estrogen released into circula-
tion [57]. However, after menopause, ovaries stop functioning, dra-
NAFLD is the most common form of chronic liver disease in the matically decreasing the concentrations of estrogen in the body. Es-
United States and is characterized by histologic hepatitis in the absence trogen synthesis continues in adipose tissue and adipose tissue-derived
of alcohol abuse [26]. Though simple steatosis is considered benign, estrogen accounts for most of the circulating estrogens in post-
steatosis of the liver can progress to the more serious conditions of menopausal women [60]. A longitudinal study showed that trends in
nonalcoholic steatohepatitis (NASH) and cirrhosis [27,28]. Population obesity and hormone levels in women differed by menopausal status
studies have demonstrated that while premenopausal women have less [61]. Obese and overweight women had significantly lower con-
NAFLD compared to men, postmenopausal women have higher in- centrations of estradiol than non-obese premenopausal women how-
cidences of NAFLD than men [29–36], which suggests a protective role ever, this trend was reversed after menopause where obese and over-
for estrogens. The decrease in estrogens due to the onset of menopause weight women have higher estradiol levels than non-obese women, due
makes postmenopausal women more susceptible to weight gain, fat aromatization of estrogens in adipose tissue.
redistribution to abdominal areas, dyslipidemia, and insulin resistance, The molecular mechanism of action of estrogens in each tissue is
all of which are major hallmarks of metabolic syndrome and are asso- determined (a) by the ER composition of the tissue; (b) by the com-
ciated with NAFLD [37–39]. Hormone replacement therapy (HRT) use position of coregulators and partnering transcription factors, (c) and
reduces NAFLD incidence in postmenopausal women [35,40]. Use of notably, as we and others have shown, also by the activation of genomic
Tamoxifen, a selective estrogen receptor modulator, increases NAFLD vs. non-genomic pathways by estrogens. Our previous studies showed that
incidence [41–43]. In addition, both the aromatase knock-out mice reproductive tissues, including endometrium and the mammary gland,
[44], which are devoid of endogenous estrogens, and ERα knock-out which express dominantly ERα, are not stimulated by ligands that
mice [45] had hepatic steatosis. All this evidence suggest a role for ERα preferentially activate non-genomic ER signaling [62–64]. These tissues
signaling and estrogen deficiency in NAFLD progression in both human rely mostly on genomic signaling for the maintenance and proliferation
and mice. of these tissues and the cancers that arise in them [62,64]. Studies in-
The pathogenesis of NAFLD is complex, and there are significant volving mice expressing membrane-only ERα (MOER) showed defects
links between NAFLD and abnormal lipid metabolism as seen in obesity in uterus and mammary gland development [65]. Interestingly, liver
and metabolic syndrome. The prevalence of NAFLD increases with in- defects associated with lipid metabolism observed in ERα knockout
creasing body mass index (BMI), with the rates of steatosis and mice were not found in MOER mice [66,67], suggesting that membrane

39
K.L. Chen, Z. Madak-Erdogan Steroids 133 (2018) 38–43

ERα signaling is sufficient to suppress lipid synthesis in the liver. In regulating levels and activity of several adipokines including leptin,
addition, findings from mice with ERα that cannot bind estrogen re- adiponectin and NPY [82,83]. The levels of adipokines, such as adi-
sponse elements, named KIKO mice, was protected from ovariectomy ponectin and leptin may be affected by estrogen concentrations [84].
and high-fat diet induced obesity upon estrogen supplementation, Aiponectin is prodced by adipocytes and are involved in fatty acid
suggesting that direct ERE binding is not required for metabolic effects metabolism and glucose regulation [85]. Levels of adiponectin are in-
of ERα [68]. versely correlated with the concentrations of plasma estrogen levels in
ERα is more associated with obesity and maintaining adipose tissue females even though females have higher overall concentrations of
function [57]. ERα knockout (ERαKO) mice are more prone to obesity plasma adiponectin than males do. However, this might be due to other
and have a reduced energy expenditure regardless of gender. These hormones such as testosterone that results in the dimorphic differences
results differ from ERβ knockout mice, which do not have significant in adiponectin levels. strogen increases leptin production as well as
increase in visceral fat mass. Furthermore, ERαKO mice have increased leptin receptor expression in rats and humans [86]. Supplementation of
central adiposity. ERαKO mice have double the weight of fat pads estrogen in postmenopausal women caused an increase in circulating
compared to wild type animals even though energy intake are about leptin levels. In ovariectomized female rats injected with exogenous
equal. Studies in liver ERα knock-out mice (LERKO) showed that liver leptin, responsiveness to the anorexigenic effects of leptin were reduced
ERα is required for preventing aberrant hepatic lipid deposition in both and these mice had much higher food intake and increase in body
males and females [69,70]. weight than intact females. As leptin inhibits hunger and influences
Even though ERα have more direct effects in metabolic regulation, satiety, estrogens can aid in energy regulation and food intake to con-
ERβ is also reported to have effects on the mechanism of fat accumu- trol obesity through this mechanism. Other adipokines such as tumor
lation and distribution. In the absence of adipose ERα, ERβ prevents necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) were also shown
inflammation and fibrosis [71]. Interestingly, ERβ was suggested to to be increased following estrogen treatment of aromatase knockout
influence microbiota maintenance in the gut [72]. The gastrointestinal mice [79]. These data show that estrogens control the levels of different
microbiota have hydrolytic capabilities, which may expand and reduce adipokines associated with glucose and lipid metabolism which may
the ability of the host to absorb and intake dietary components. In lead to the higher instances of obesity after menopause when estrogen
addition to direct effects in the gut and microbiome, ERβ is expressed in levels decline.
the dorsal raphe, a region of the brain where cell bodies of serotonin Estrogens regulate expression levels of several genes that are im-
neurons originate [73]. Serotonin neurons are critical in the regulation portant for lipid deposition. FASN is fatty acid synthase which syn-
of food intake. It has also been proposed that ERβ is important for the thesizes fatty acids. Estradiol regulates FASN expression in liver and
maintenance of normal glucose and lipid metabolism in the body [74]. adipocytes by modulating downstream signaling pathway cross-talk
Estrogens regulate adipocyte metabolism and induce remodeling of [63,87,88]. In ovariectomized mice, FASN levels were increased, which
particular adipose depots, which are different between premenopausal were reversed by the administration of an estrogen receptor ligand.
women and men compared to post-menopausal women [59]. Estrogen Estrogen also appears to upregulate α2A-adrenergic receptors in human
Receptors partner with other transcription factors including LXR, adipose tissue which lowers the lipolytic response in subcutaneous
SREBP and especially cHREBP to regulate metabolism and readers are adipose tissue of healthy women [89]. By upregulating these receptors
encouraged to read the detailed review on this topic (REF. Ellis Levin only in subcutaneous adipose tissue and not in abdominal adipose
review on “Membrane estrogen receptors signal to determine tran- tissue, fat accumulation is higher in subcutaneous areas. This may ex-
scription factor function”). Estrogens can decrease lipogenesis by in- plain why females have higher deposits in these areas compared to the
hibiting lipoprotein lipase (LPL) [75]. LPL is responsible for hydro- higher abdominal fat accumulation in males. SREBP-1 is sterol reg-
lyzing triglycerides and facilitates adipose tissue storage. An in vitro ulatory element-binding protein 1 which is involved in glucose meta-
study using mouse adipocyte cell lines showed that fat accumulation bolism, fatty acid production, and cholesterol production. After ovar-
depended on the expression of this enzyme as E2 treatment decreased iectomy, expression of SREBP-1c, one of the isoforms of the SREBP, was
LPL mRNA, which blocked lipid deposition in turn. This effect is much higher than in intact mice [90]. Like FASN, administration of
regulated by an estrogen-response suppressive element in the LPL estrogens decreased expression of SREBP-1 to prevent fat accumulation
promoter gene. Increased levels of LPL have been shown in adipose [63,88].
tissue of obese human patients [76]. In obese women, plasma estradiol
activity has a strong negative correlation with LPL activity in adipose 4. Interaction of microbiota and estrogens
and postheparin plasma. These studies suggest that estrogen may di-
rectly influence fat storage and limit fat accumulation in adipocytes by Gut microbiota composition has a major impact on metabolic status
decreasing LPL action. of the host. Insulin sensitivity of mice can be improved by small in-
In ovariectomized mice fed a high fat diet, E2 administration im- testine infusion of gut microbiota samples from healthy donors in mice
proved glucose tolerance and insulin sensitivity. When estrogen pro- [91]. In a healthy gut, microbiota digests resistant starch and fibers,
duction decreases, as in menopause or following ovariectomy, the body synthesize vitamins, provide immunity, and prevent pathogen coloni-
weight increases. This increase can be prevented by estradiol treatment zation [92]. If gut microbiota is disrupted, bacterial metabolites and
[77–79]. In terms of glucose homeostasis, aromatase knockout mice bacterial cell components can reduce intestinal epithelial integrity and
had hepatic insulin resistance because of decreased Akt phosphoryla- access the liver through the portal vein. These microbial byproducts can
tion in the liver [79]. Akt is a key mediator in the insulin signaling contribute to inflammation, liver steatosis, and hepatic injury ulti-
pathways. With estrogen administration, Akt phosphorylation of ar- mately leading to NAFLD [93].
omatase knockout mice was restored reversing these effects. Therefore, High fat diet changes gut microbiota composition and increases the
estradiol treatment in estrogen deficient mice improved insulin sensi- concentration of gram negative bacteria, which produces endotoxin,
tivity by increasing the phosphorylation of the key mediators in insulin lipopolysaccharide (LPS) [94,95]. As gram negative bacteria produces
signaling [79,80]. endotoxins increase inflammation as associated diseases such as type 2
In addition to direct effects of estrogens on metabolic tissues, ERs diabetes [96] and NAFLD [97–100]. NAFLD is strongly influenced by
are also expressed in regions of central nervous system (CNS) that obesity and insulin resistance [101]. In fact, gut microbiota composi-
regulates food intake and energy expenditure [81]. ERα is the main tion is altered in NAFLD where certain bacterial overgrowths contribute
mediator of estrogen action in CNS and play important roles in main- to disease development and progression [102]. Insulin sensitivity of
taining function of specialized neuronal circuits such as pro-opiomela- mice can be improved by small intestine infusion of gut microbiota
nocortin (POMC) neurons or ventral medial hypothalamus (VMH) and samples from healthy donors in mice [91]. Manipulating the gut

40
K.L. Chen, Z. Madak-Erdogan Steroids 133 (2018) 38–43

microbiota composition with probiotics and prebiotics, such as Lacto- therapies that work independently or synergistically to provide a more
bacillus and Bifidobacterium strains also improve weight and fat mass holistic treatment approach to prevent metabolic disease.
development. Mice fed a high fat diet had improved glucose-insulin
homeostasis and attenuated weight gain when supplemented with Funding
Lactobacillus and Bifidobacterium probiotics.
Gut microorganism composition can be altered with the addition of This work was supported by grants from the University of Illinois,
various chemical substrates and dietary administration of estrogen-like Office of the Vice Chancellor for Research, Arnold O. Beckman award
compounds were shown to promote the proliferation and growth of RB15150 (to ZME), National Institute of Food and Agriculture, U.S.
certain types of bacteria [103]. Dietary consumption of soy isoflavones Department of Agriculture, award ILLU-698-909 (to ZME), and in-
such as daidzein, genistein, and glycitin can significantly alter fecal vestigator initiated grant-WI196180 from Pfizer (to ZME).
bacterial community structure and composition in postmenopausal
women [104]. Specifically, soy phytoestrogen consumption increases References
the concentration of Bifidobacterium, a bacteria beneficial to gut
health, and suppresses Clostridiaceae, an opportunistic pathogens [1] K.M. Flegal, D. Kruszon-Moran, M.D. Carroll, C.D. Fryar, C.L. Ogden, Trends in
which cause of several human diseases [56]. As estrogens influence the obesity among adults in the United States, 2005 to 2014, JAMA 315 (21) (2016)
2284–2291.
microbiota, some bacteria also metabolize estrogen-like compounds or [2] M. Aguilar, T. Bhuket, S. Torres, B. Liu, R.J. Wong, Prevalence of the metabolic
phytoestrogens, such as soy isoflavones to produce more biologically syndrome in the United States, 2003–2012, JAMA 313 (19) (2015) 1973–1974.
active forms that have high affinity for human estrogen receptors [105]. [3] M. Rodriguez-Monforte, E. Sanchez, F. Barrio, B. Costa, G. Flores-Mateo, Metabolic
syndrome and dietary patterns: a systemic review and meta-analysis of observa-
Higher serum levels of gut microbial metabolites of phytoestrogen lig- tional studies, Eur. J. Nutr. 56 (3) (2016) 925–947.
nans were associated with a lower weight in postmenopausal women [4] G.A. Bray, The Metabolic Syndrome and Obesity, Humana Press, 2007.
[106]. Some gut bacteria are known to metabolize phytoestrogens into [5] S.M. Grundy, H.B. Brewer, J.I. Cleeman, S.C. Smith, C. Lenfant, Definition of
metabolic syndrome: report of the national heart, lung, and blood institute/
O-desmethylangolensin (ODMA) and equol, which are structurally si-
american heart association conference on scientific issues related to definition,
milar to mammalian estrogen [107]. ODMA producing individuals are Arterioscler Thromb. Vasc. Biol. 24 (2004) e13–e18.
leaner [108,109]. Equol supplementation improves glycemic control [6] J.L. Kulk, C.I. Ardem, Age and sex differences in the clustering of metabolic syn-
drome factors: association with mortality risk, Diabetes Care 33 (11) (2010)
and lowers LDL levels [107,109]. Recently we showed that dietary li-
2457–2461.
corice root supplementation, which contain isoliquiritegenin, a low- [7] S. Kumagai, A. Holmang, P. Bjorntorp, The effects of oestrogen and progesterone
affinity ER ligand, improved ovariectomy associated weight gain and on insulin sensitivity in female rats, Acta Physiol. Scand. 149 (1993) 91–97.
metabolic syndrome in mice [110]. Administration of this botanical [8] J.D. Wagner, M.K. Thomas, J.K. Williams, L. Zhang, K.A. Greaves, W.T. Cefalu,
Insulin sensitivity and cardiovascular risk factors in ovariectomized monkeys with
estrogen did not stimulate reproductive tissues. However, how iso- estradiol alone or combined with nomegestrol acetate, J. Clin. Endocrinol. Metab.
liquiritegenin or any other low affinity estrogen supplementation would 83 (1998) 896–901.
alter gut microbiota and blood metabolites is not known. [9] N.F. Goodman, R.H. Cobin, S.B. Ginzburg, I.A. Katz, D.E. Woode, E. American
Association of Clinical, American Association of Clinical Endocrinologists Medical
As estrogens are metabolized by the liver through first-pass meta- Guidelines for Clinical Practice for the diagnosis and treatment of menopause,
bolism and biotransformed through methylation, hydroxylation, and Endocr. Pract. 17 (Suppl 6) (2011) 1–25.
conjugation. The estrobolome, collection of estrogen metabolizing en- [10] The 2017 hormone therapy position statement of The North American Menopause
Society, Menopause 24(7) (2017) 728–753.
zymes associated with gut microbioata, could further alter these [11] C.A. Stuenkel, S.R. Davis, A. Gompel, M.A. Lumsden, M.H. Murad, J.V. Pinkerton,
transformed estrogens and generate novel ligands of varying potency R.J. Santen, Treatment of symptoms of the menopause: an endocrine society
for estrogen receptors [111]. For example, some bacterial species with clinical practice guideline, J. Clin. Endocrinol. Metab. 100 (11) (2015)
3975–4011.
β-glucosidase activity can deconjugate estrogens, producing estrogens [12] J. MacKenzie, B.M. Fisher, A.J. Jaap, A. Stanley, K. Paterson, N. Sattar, Effects of
with higher biological activity [112]. Higher gut microbial diversity is HRT on liver enzyme levels in women with type 2 diabetes: a randomized placebo-
associated with more estrogen metabolites in urine [112]. Increased controlled trial, Clin. Endocrinol. 65 (1) (2006) 40–44.
[13] Z. Bai, R. Gust, Breast cancer, estrogen receptor and ligands, Arch. Pharm. 342
parent estrogen concentrations compared to estrogen metabolites in
(2009) 133–149.
serum have been associated with a low gut microbial diversity and [14] B.J. Deroo, K.S. Korach, Estrogen receptors and human disease, J. Clin. Invest. 116
increased breast cancer risk [113,114]. Estrogen metabolite to parent (2006) 561–570.
estrogen ratio may also be an important indicator of metabolic dys- [15] E.M. Fox, J. Davis, M.A. Shupnik, ERB in breast cancer–onlooker, passive player,
or active protector? Steroids 73 (2008) 1039–1051.
function. These findings suggest gut microbiome as a potential bio- [16] C. Osipo, H. Liu, K. Meeke, V.C. Jordan, The consequences of exhaustive anti-
marker for identifying metabolic disease risk and eventually preventing estrogen therapy in breast cancer: estrogen-induced tumor cell death, Exp. Biol.
these diseases using probiotic interventions. Med. 229 (2004) 722–731.
[17] J.V. Pinkerton, J.A. Harvey, R. Lindsay, K. Pan, A.A. Chines, S. Mirkin,
D.F. Archer, Effects of bazedoxifene/conjugated estrogens on the endometrium
5. Conclusions and future research and bone: a randomized trial, J. Clin. Endocrinol. Metab. 99 (2) (2014)
E189–E198.
[18] A. Itabashi, K. Yoh, A.A. Chines, T. Miki, M. Takada, H. Sato, I. Goria, T. Sugimoto,
Estrogens are key in maintaining homeostasis in healthy pre- H. Mizunuma, H. Ochi, G.D. Constantine, H. Ohta, Effects of bazedoxifene on bone
menopausal individuals. When estrogen levels decrease with meno- mineral density. bone turnover, and safety in postmenopausal japanese women
pause, metabolic rate decreases, causing increased weight and obesity. with osteoporosis, J. Bone Miner. Res. 26 (3) (2011) 519–529.
[19] S.E. Wardell, E.R. Nelson, C.A. Chao, D.P. McDonnell, Bazedoxifene exhibits an-
However, we still do not know how certain estrogens may alter the tiestrogenic activity in animal models of tamoxifen resistant breast cancer; im-
microbiome to resemble normal weight phenotypes or how gut estro- plications for treatment of advanced disease, Clin. Cancer Res. 19 (9) (2013)
genic byproducts produced by gut bacteria may act as ER ligands to 2420–2431.
[20] Z. Madak-Erdogan, P. Gong, B.S. Katzenellenbogen, Differential utilization of
mediate metabolic functions. It may be possible that the microbiota
nuclear and extranuclear receptor signaling pathways in the actions of estrogens,
metabolize endogenous or dietary estrogens to produce novel ER li- SERMs, and a tissue-selective estrogen complex (TSEC), J. Steroid Biochem. Mol.
gands that can also be used to mitigate symptoms associated with Biol. 158 (2016) 198–206.
menopause and obesity. Alternatively, the microbiota may be ma- [21] R. Lindsey, Preventing osteoporosis with a tissue selective estrogen complex
(TSEC) containing bazedoxifene/conjugated estrogens (BZA/CE), Osteoporos Int.
nipulated to produce less potent estrogens that could maintain meta- 22 (2011) 447–451.
bolic function but reduce the risk of other diseases such as cancer. By [22] R.A. Lobo J.V. Pinkerton M.L. Gass M.H. Dorin S. Ronkin J.H. Pickar G.
identifying and analyzing other forms of estrogen that may be less Constantine Evaluation of bazedoxifene/conjugated estrogens for the treatment of
menopausal symptoms and effects on metabolic parameters and overal safety
potent and bind loosely to receptors, it may be possible to find more profile Fetil. Steril. 92(3) (2009) 1025-1038.
effective forms of estrogen replacement therapies. Treatment may also [23] R.J. Santen, Y. Song, W. Yue, J.P. Wang, D.F. Heitjan, Effects of menopausal
involve using microbiota therapies in combination with estrogen hormonal therapy on occult breast tumors, J. Steroid Biochem. Mol. Biol. 137

41
K.L. Chen, Z. Madak-Erdogan Steroids 133 (2018) 38–43

(2013) 150–156. [51] K. Yamaguchi, L. Yang, S. McCall, J. Huang, X.X. Yu, S.K. Pandey, S. Bhanot,
[24] J.H. Kim, M.S. Meyers, S.S. Khuder, S.L. Abdallah, H.T. Muturi, L. Russo, C.R. Tate, B.P. Monia, Y.X. Li, A.M. Diehl, Inhibiting triglyceride synthesis improves hepatic
A.L. Hevener, S.M. Najjar, C. Leloup, F. Mauvais-Jarvis, Tissue-selective estrogen steatosis but exacerbates liver damage and fibrosis in obese mice with nonalco-
complexes with bazedoxifene prevent metabolic dysfunction in female mice, Mol. holic steatohepatitis, Hepatology 45 (6) (2007) 1366–1374.
Metab. 3 (2014) 177–190. [52] U.S.D.o.H.a.H.S. U.S. Department of Agriculture, Dietary Guidelines for
[25] J. Barrera, K.L. Chambliss, M. Ahmed, K. Tanigaki, B. Thompson, J.G. McDonald, Americans, 7 ed., U.S. Government Printing Office, Washington, DC, 2010.
C. Mineo, P.W. Shaul, Bazedoxifene and conjugated estrogen prevent diet-induced [53] P. Angulo, L.M. Alba, L.A. Adams, K.D. Lindor, M.D. Jensen, Leptin, insulin re-
obesity, hepatic steatosis, and type 2 diabetes in mice without impacting the re- sistance, and liver fibrosis in human nonalcoholic fatty liver disease, J. Hepatol. 41
productive tract, Am. J. Physiol. Endocrinol. Metab. 307 (2014) E345–E354. (2004) 943–949.
[26] J. Henao-Mejia, E. Elinav, C. Kin, L. Hao, W.Z. Mehal, T. Strowig, C.A. Thaiss, [54] L.H. Storlien, X.F. Huang, S. Lin, X. Xin, H.Q. Wang, P.L. Else, Dietary fat subtypes
A.L. Kau, S.C. Eisenbarth, M.J. Jurczak, J.-P. Camporez, G.I. Shulmanm, and obesity, World Rev. Nutr. Dietetics 88 (2001) 148–154.
J.I. Gordon, H.M. Hoffman, R.A. Flavell, Inflammasome-mediated dysbiosi reg- [55] A.K. Leamy, R.A. Egnatchik, J.D. Young, Molecular mechanisms and the role of
ulates progression of NAFLD and obesity, Nature 482 (2012) 179–185. saturated fatty acids in the progression of non-alcoholic fatty liver disease, Prog.
[27] E. Scorletti, P.C. Calder, C.D. Byrne, Non-alcoholic fatty liver disease and cardi- Lipid Res. 52 (1) (2013) 165–174.
ovascular risk: metabolic aspects and novel treatments, Endocrine 40 (3) (2011) [56] M.J. Dekker, Q.Z. Su, C. Baker, A.C. Rutledge, K. Adeli, Fructose: a highly lipo-
332–343. genic nutrient implicated in insulin resistance, hepatic steatosis, and the metabolic
[28] C. Loguercio, T. De Simone, M.V. D'Auria, I. de Sio, A. Federico, C. Tuccillo, syndrome, Am. J. Physiol.-Endoc. M 299 (5) (2010) E685–E694.
A.M. Abbatecola, C. Del Vecchio Blnco, Non-alcoholic fatty liver disease: a mul- [57] C.J. Park, Z. Zhao, C. Glidewell-Kenney, M. Lazic, P. Chambon, A. Krust, J. Weiss,
ticentre clinical study by the Italian Association for the Study of the Liver, Dig. D.J. Clegg, A. Dunaif, J.L. Jameson, J.E. Levine, Genetic rescue of nonclassical
Liver Dis. 36 (6) (2004) 398–405. ERalpha signaling normalizes energy balance in obese Eralpha-null mutant mice,
[29] S.-I. Kojima, N. Watanabe, M. Numata, T. Ogawa, S. Matsuzaki, Increase in the J. Clin. Invest. 121 (2) (2011) 604–612.
prevalence of fatty liver in Japan over the past 12 years: analysis of clinical [58] A. Foryst-Ludwig, U. Kintscher, Metabolic impact of estrogen signalling through
background, J. Gastroenterol. 38 (10) (2003) 954–961. ERalpha and ERbeta, J. Steroid Biochem. Mol. Biol. 122 (1–3) (2010) 74–81.
[30] S.H. Park, W.K. Jeon, S.H. Kim, H.J. Kim, D.I. Park, Y.K. Cho, I.K. Sung, C.I. Sohn, [59] C.J. Gruber, W. Tschugguel, C. Schneeberger, J.C. Huber, Production and actions
D.K. Keum, B.I. Kim, Prevalence and risk factors of non-alcoholic fatty liver disease of estrogens, N. Engl. J. Med. 346 (5) (2002) 340–352.
among Korean adults, J. Gastroenterol. Hepatol. 21 (1) (2006) 138–143. [60] C. Stocco, Tissue physiology and pathology of aromatase, Steroids 77 (1–2) (2012)
[31] Y.-J. Zhou, Y.-Y. Li, Y.-Q. Nie, J.-X. Ma, L.-G. Lu, S.-L. Shi, M.-H. Chen, P.-J. Hu, 27–35.
Prevalence of fatty liver disease and its risk factors in the population of South [61] E.R. Simpson, Y. Zhao, V.R. Agarwal, M.D. Michael, S.E. Bulun,
China, World J. Gastroenterol. 13 (47) (2007) 6419–6424. M.M. Hinshelwood, S. Graham-Lorence, T. Sun, C.R. Fisher, K. Qin,
[32] H. Nomura, S. Kashiwagi, J. Hayashi, W. Kajiyama, S. Tani, M. Goto, Prevalence of C.R. Mendelson, Aromatase expression in health and disease, Recent Prog. Horm.
fatty liver in a general population of Okinawa, Japan, Jpn. J. Med. 27 (2) (1988) Res. 52 (1997) 185–213 discussion 213-4.
142–149. [62] K.L. Chambliss, Q. Wu, S. Oltmann, E.S. Konaniah, M. Umetani, K.S. Korach,
[33] Y. Gutierrez-Grobe, G. Ponciano-Rodriguez, M.H. Ramos, M. Uribe, N. Mendez- G.D. Thomas, C. Mineo, I.S. Yuhanna, S.H. Kim, Z. Madak-Erdogan, A. Maggi,
Sanchez, Prevalence of non alcoholic fatty liver disease in premenopausal, post- S.P. Dineen, C.L. Roland, D.Y. Hui, R.A. Brekken, J.A. Katzenellenbogen,
menopausal and polycystic ovary syndrome women. The role of estrogens, Ann. B.S. Katzenellenbogen, P.W. Shaul, Non-nuclear estrogen receptor alpha signaling
Heptaology 9 (4) (2010) 402–409. promotes cardiovascular protection but not uterine or breast cancer growth in
[34] Z.X. Wang, M. Xu, Z. Hu, U.K. Shrestha, Prevalence of nonalcoholic fatty liver mice, J. Clin. Invest. 120 (7) (2010) 2319–2330.
disease and its metabollic risk factors in women of different ages and body mass [63] Z. Madak-Erdogan, S.H. Kim, P. Gong, Y.C. Zhao, K.L. Chambliss, K.E. Carlson,
index, Menopause 22 (6) (2014) 667–673. C.G. Mayne, P.W. Shaul, K.S. Korach, J.A. Katzenellenbogen,
[35] G.S.d.A. Florentino, H.P. Cotrim, C.P. Vilar, A.V.d.A. Florentino, B.S. Katzenellenbogen, Design of pathway preferential estrogens that provide
G.M.A. Guimaraes, V.S.T. Barreto, Nonalcoholic fatty liver disease in menopausal beneficial metabolic and vascular effects without stimulating reproductive tissues,
women, Arquivos de Gastroenterologia 50 (2013) 180–185. Sci. Signal 9 (429) (2016) ra53.
[36] C.W. Brady, Liver disease in menopause, World J. Gastroenterol. 21 (25) (2015) [64] Z. Madak-Erdogan, K.J. Kieser, S.H. Kim, B. Komm, J.A. Katzenellenbogen,
7613–7620. B.S. Katzenellenbogen, Nuclear and extranuclear pathway inputs in the regulation
[37] F. Mauvais-Jarvis, D.J. Clegg, A.L. Hevener, The role of estrogens in control of of global gene expression by estrogen receptors, Mol. Endocrinol. 22 (9) (2008)
energy balance and glucose homeostasis, Endocr. Rev. 34 (3) (2013) 309–338. 2116–2127.
[38] G. Pagano, G. Pacini, G. Musso, R. Gambino, F. Mecca, N. Depetris, M. Cassader, [65] A. Pedram, M. Razandi, J.K. Kim, F. O'Mahony, E.Y. Lee, U. Luderer, E.R. Levin,
E. David, P. Cavallo-Perin, M. Rizzetto, Nonalcoholic steatohepatitis, insulin re- Developmental phenotype of a membrane only estrogen receptor alpha (MOER)
sistance, and metabolic syndrome: Further evidence for an etiologic association, mouse, J. Biol. Chem. 284 (6) (2009) 3488–3495.
Hepatology 35 (2) (2002) 367–372. [66] K.J. Hamilton, Y. Arao, K.S. Korach, Estrogen hormone physiology: reproductive
[39] G. Marchesini, E. Bugianesi, G. Forlani, F. Cerrelli, M. Lenzi, R. Manini, S. Natale, findings from estrogen receptor mutant mice, Reprod. Biol. 14 (1) (2014) 3–8.
E. Vanni, N. Villanova, N. Melchionda, M. Rizzetto, Nonalcoholic fatty liver, [67] A. Pedram, M. Razandi, F. O'Mahony, H. Harvey, B.J. Harvey, E.R. Levin, Estrogen
steatohepatitis, and the metabolic syndrome, Hepatology 37 (4) (2003) 917–923. reduces lipid content in the liver exclusively from membrane receptor signaling,
[40] J. McKenzie, B.M. Fisher, A.J. Jaap, A. Stanley, K. Paterson, N. Sattar, Effects of Sci. Signal 6 (276) (2013) ra36.
HRT on liver enzyme levels in women with type 2 diabetes: a randomized placebo- [68] A. Yasrebi, J.A. Rivera, E.A. Krumm, J.A. Yang, T.A. Roepke, Activation of es-
controlled trial, Clin. Endocrinol. 65 (1) (2006) 40–44. trogen response element-independent ERα signaling protects female mice from
[41] K.A. Oien, D. Moffat, G.W. Curry, J. Dickson, T. Habeshaw, P.R. Mills, diet-induced obesity, Endocrinology 158 (2) (2017) 319–334.
R.N.M. MacSween, Cirrhosis with steatohepatitis after adjuvant tamoxifen, The [69] S. Della Torre, N. Mitro, R. Fontana, M. Gomaraschi, E. Favari, C. Recordati,
Lancet 353 (9146) (1999) 36–37. F. Lolli, F. Quagliarini, C. Meda, C. Ohlsson, M. Crestani, Nina H. Uhlenhaut,
[42] D.S. Pratt, T.A. Knox, J. Erban, TAmoxifen-induced steatohepatitis, Ann. Intern. L. Calabresi, A. Maggi, An essential role for liver ERα in coupling hepatic meta-
Med. 123 (3) (1995) 236–236. bolism to the reproductive cycle, Cell Rep. 15 (2) (2016) 360–371.
[43] S. Bruno, P. Maisonneuve, P. Castellana, N. Rotmensz, S. Rossi, M. Maggioni, [70] S. Qiu, J.T. Vazquez, E. Boulger, H. Liu, P. Xue, M.A. Hussain, A. Wolfe, Hepatic
M. Persico, A. Colombo, F. Monasterolo, D. Casadei-Giunchi, F. Desiderio, estrogen receptor α is critical for regulation of gluconeogenesis and lipid meta-
T. Stroffolini, V. Sacchini, A. Decensi, U. Veronesi, Incidence and risk factors for bolism in males, Sci. Rep. 7 (1) (2017) 1661.
non-alcoholic steatohepatitis: prospective study of 5408 women enrolled in Italian [71] K.E. Davis, M.D. Neinast, K. Sun, W.M. Skiles, J.D. Bills, J.A. Zehr, D. Zeve,
tamoxifen chemoprevention trial, BMJ 330 (7497) (2005) 932. L.D. Hahner, D.W. Cox, L.M. Gent, Y. Xu, Z.V. Wang, S.A. Khan, D.J. Clegg, The
[44] J.D.Y. Chow, M.E.E. Jones, K. Prelle, E.R. Simpson, W.C. Boon, A selective es- sexually dimorphic role of adipose and adipocyte estrogen receptors in modulating
trogen receptor α agonist ameliorates hepatic steatosis in the male aromatase adipose tissue expansion, inflammation, and fibrosis, Mol. Metab. 2 (3) (2013)
knockout mouse, J. Endocrinol. 210 (3) (2011) 323–334. 227–242.
[45] G. Rando, W. Wahli, Sex differences in nuclear receptor-regulated liver metabolic [72] A. Pedram, M. Razandi, E.R. Levin, Nature of functional estrogen receptors at the
pathways, Biochimica et Biophysica Acta (BBA) – Molecular Basis Disease 1812 plasma membrane, Mol. Endocrinol. 20 (9) (2006) 1996–2009.
(8) (2011) 964–973. [73] P.A. Heine, J.A. Taylor, G.A. Iwamoto, D.B. Lubahn, P.S. Cooke, Increased adipose
[46] E. Fabbrini, S. Sullivan, S. Klein, Obesity and nonalcoholic fatty liver disease: tissue in male and female estrogen receptor-alpha knockout mice, Proc. Natl.
biochemical, metabolic and clinical implications, Heptaology 51 (2) (2010) Acad. Sci. USA 97 (23) (2000) 12729–12734.
679–689. [74] P.J. Shughrue, M.V. Lane, I. Merchenthaler, Comparative distribution of estrogen
[47] N.C. Leite, G.F. Salles, A.L. Araujo, C.A. Villeda-Nogueira, C.R. Cardoso, receptor-alpha and -beta mRNA in the rat central nervous system, J. Comp.
Prevalence and assocaited factors of non-alcoholic fatty liver disease in patients Neurol. 388 (4) (1997) 507–525.
with type-2 diabetes mellitus, Liver Int. 29 (2009) 113–119. [75] H. Homma, H. Kurachi, Y. Nishio, T. Takeda, T. Yamamoto, K. Adachi,
[48] M.E. Rinella, Nonalcoholic fatty liver disease: a systematic review, J. Am. Med. K. Morishige, M. Ohmichi, Y. Matsuzawa, Y. Murata, Estrogen suppresses tran-
Assoc. 313 (22) (2015) 2263–2273. scription of lipoprotein lipase gene. Existence of a unique estrogen response ele-
[49] A.E. Feldstein, N.W. Werneburg, A. Canbay, M.E. Guicciardi, S.F. Bronk, ment on the lipoprotein lipase promoter, J. Biol. Chem. 275 (15) (2000)
R. Rydzewski, L.J. Burgart, G.J. Gores, Free fatty acids promote hepatic lipotoxi- 11404–11411.
city by stimulating TNF-alpha expression via ysosomal pathway, Hepatology 40 [76] J.M. Ong, P.A. Kern, Effect of feeding and obesity on lipoprotein lipase activity,
(2004) 184–194. immunoreactive protein, and messenger RNA levels in human adipose tissue, J.
[50] C.P. Day, Steatohepatitis: a tale of two ‘hits’? Gastroenterology 114 (1998) Clin. Invest. 84 (1) (1989) 305–311.
1349–1354. [77] C. Tagliaferri, J. Salles, J.F. Landrier, C. Giraudet, V. Patrac, P. Lebecque,

42
K.L. Chen, Z. Madak-Erdogan Steroids 133 (2018) 38–43

M.J. Davicco, A. Chanet, C. Pouyet, A. Dhaussy, A. Huertas, Y. Boirie, Y. Wittrant, mice through lipopolysaccharides (LPS) receptor signaling: protective action of
V. Coxam, S. Walrand, Increased body fat mass and tissue lipotoxicity associated estrogens, PLoS One 7 (11) (2012) e48220.
with ovariectomy or high-fat diet differentially affects bone and skeletal muscle [97] G. Paolella, C. Mandato, L. Pierri, M. Poeta, M. Di Stasi, P. Vajro, Gut-liver axis and
metabolism in rats, Eur. J. Nutr. 54 (7) (2015) 1139–1149. probiotics: their role in non-alcoholic fatty liver disease, World J.
[78] R.E. Stubbins, V.B. Holcomb, J. Hong, N.P. Nunez, Estrogen modulates abdominal Gastroenterology: WJG 20 (42) (2014) 15518–15531.
adiposity and protects female mice from obesity and impaired glucose tolerance, [98] J. Henao-Mejia, E. Elinav, C. Jin, L. Hao, W.Z. Mehal, T. Strowig, C.A. Thaiss,
Eur. J. Nutr. 51 (7) (2012) 861–870. A.L. Kau, S.C. Eisenbarth, M.J. Jurczak, J.P. Camporez, G.I. Shulman, J.I. Gordon,
[79] M.L. Van Sinderen, G.R. Steinberg, S.B. Jorgensen, J. Honeyman, J.D. Chow, H.M. Hoffman, R.A. Flavell, Inflammasome-mediated dysbiosis regulates pro-
K.A. Herridge, A.L. Winship, E. Dimitriadis, M.E. Jones, E.R. Simpson, W.C. Boon, gression of NAFLD and obesity, Nature 482 (7384) (2012) 179–185.
Effects of estrogens on adipokines and glucose homeostasis in female aromatase [99] W. Jiang, N. Wu, X. Wang, Y. Chi, Y. Zhang, X. Qiu, Y. Hu, J. Li, Y. Liu, Dysbiosis
knockout mice, PLoS One 10 (8) (2015) e0136143. gut microbiota associated with inflammation and impaired mucosal immune
[80] M.L. Van Sinderen, G.R. Steinberg, S.B. Jorgensen, J. Honeyman, J.D. Chow, function in intestine of humans with non-alcoholic fatty liver disease, Sci. Rep. 5
K.A. Herridge, A.L. Winship, E. Dimitriadis, M.E.E. Jones, E.R. Simpson, (2015) 8096.
W.C. Boon, Effects of estrogens on adipokines and glucose homeostasis in female [100] L.C. Kong, B.A. Holmes, A. Cotillard, F. Habi-Rachedi, R. Brazeilles, S. Gougis,
aromatase knockout mice, PLoS One 10 (8) (2015) e0136143. N. Gausseres, P.D. Cani, S. Fellahi, J.P. Bastard, S.P. Kennedy, J. Dore,
[81] A.L. Hevener, D.J. Clegg, F. Mauvais-Jarvis, Impaired estrogen receptor action in S.D. Ehrlich, J.D. Zucker, S.W. Rizkalla, K. Clement, Dietary patterns differently
the pathogenesis of the metabolic syndrome, Mol. Cell. Endocrinol. 418 (Part 3) associate with inflammation and gut microbiota in overweight and obese subjects,
(2015) 306–321. PLoS One 9 (10) (2014) e109434.
[82] A. Frank, L.M. Brown, D.J. Clegg, The role of hypothalamic estrogen receptors in [101] B.W. Smith, L.A. Adams, Nonalcoholic fatty liver disease and diabetes mellitus:
metabolic regulation, Front. Neuroendocrinol. 35 (4) (2014) 550–557. pathogenesis and treatment, Nat. Rev. Endocrinol. 7 (8) (2011) 456–465.
[83] M. López, M. Tena-Sempere, Estrogens and the control of energy homeostasis: a [102] S.M. Ferolla, G.N. Armiliato, C.A. Couto, T.C. Ferrari, The role of intestinal bac-
brain perspective, Trends Endocrinol. Metab. 26 (8) (2015) 411–421. teria overgrowth in obesity-related nonalcoholic fatty liver disease, Nutrients 6
[84] P.H. Iverius, J.D. Brunzell, Relationship between lipoprotein lipase activity and (12) (2014) 5583–5599.
plasma sex steroid level in obese women, J. Clin. Invest. 82 (3) (1988) 1106–1112. [103] C.L. Frankenfeld, Relationship of obesity and high urinary enterolignan con-
[85] T.P. Combs, A.H. Berg, M.W. Rajala, S. Klebanov, P. Iyengar, J.C. Jimenez- centrations in 6806 children and adults: analysis of National Health and Nutrition
Chillaron, M.E. Patti, S.L. Klein, R.S. Weinstein, P.E. Scherer, Sexual differentia- Examination Survey data, Eur. J. Clin. Nutr. 67 (8) (2013) 887–889.
tion, pregnancy, calorie restriction, and aging affect the adipocyte-specific secre- [104] S. Possemiers, S. Bolca, W. Verstraete, A. Heyerick, The intestinal microbiome: a
tory protein adiponectin, Diabetes 52 (2) (2003) 268–276. separate organ inside the body with the metabolic potential to influence the
[86] H. Shimizu, Y. Shimomura, Y. Nakanishi, T. Futawatari, K. Ohtani, N. Sato, bioactivity of botanicals, Fitoterapia 82 (1) (2011) 53–66.
M. Mori, Estrogen increases in vivo leptin production in rats and human subjects, [105] C.L. Frankenfeld, C. Atkinson, K. Wahala, J.W. Lampe, Obesity prevalence in re-
J. Endocrinol. 154 (2) (1997) 285–292. lation to gut microbial environments capable of producing equol or O-desmethy-
[87] M.F. Santolla, R. Lappano, P. De Marco, M. Pupo, A. Vivacqua, D. Sisci, langolensin from the isoflavone daidzein, Eur. J. Clin. Nutr. 68 (4) (2014)
S. Abonante, D. Iacopetta, A.R. Cappello, V. Dolce, M. Maggiolini, G protein- 526–530.
coupled estrogen receptor mediates the up-regulation of fatty acid synthase in- [106] T. Usui, M. Tochiya, Y. Sasaki, K. Muranaka, H. Yamakage, A. Himeno,
duced by 17beta-estradiol in cancer cells and cancer-associated fibroblasts, J. Biol. A. Shimatsu, A. Inaguma, T. Ueno, S. Uchiyama, N. Satoh-Asahara, Effects of
Chem. 287 (52) (2012) 43234–43245. natural S-equol supplements on overweight or obesity and metabolic syndrome in
[88] Z. Madak-Erdogan, P. Gong, Y. Chen Zhao, L. Xu, K.U. Wrobel, J.A. Hartman, the Japanese, based on sex and equol status, Clin. Endocrinol. (Oxf) 78 (3) (2013)
M. Wang, A. Cam, U.T. Iwaniec, R.T. Turner, N.C. Twaddle, D.R. Doerge, 365–372.
I.A. Khan, J.A. Katzenellenbogen, B.S. Katzenellenbogen, W.G. Helferich, Dietary [107] V.C. Antharam, E.C. Li, A. Ishmael, A. Sharma, V. Mai, K.H. Rand, G.P. Wang,
licorice root supplementation reduces diet-induced weight gain, lipid deeposition, Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile
and heptaic steatosis in ovariectomized mice without stimulating reproductive infection and nosocomial diarrhea, J. Clin. Microbiol. 51 (9) (2013) 2884–2892.
tissues and mammary gland, Mol. Nutr. Food Res. 60 (2) (2015) 369–380. [108] R. Menon, S.E. Watson, L.N. Thomas, C.D. Allred, A. Dabney, M.A. Azcarate-Peril,
[89] S.B. Pedersen, K. Kristensen, P.A. Hermann, J.A. Katzenellenbogen, B. Richelsen, J.M. Sturino, Diet complexity and estrogen receptor beta status affect the com-
Estrogen controls lipolysis by up-regulating alpha2A-adrenergic receptors directly position of the murine intestinal microbiota, Appl. Environ. Microbiol. 79 (18)
in human adipose tissue through the estrogen receptor alpha. Implications for the (2013) 5763–5773.
female fat distribution, J. Clin. Endocrinol. Metab. 89 (4) (2004) 1869–1878. [109] C.H. Nakatsu, A. Armstrong, A.P. Clavijo, B.R. Martin, S. Barnes, C.M. Weaver,
[90] A. Paquette, D. Wang, M. Jankowski, J. Gutkowska, J.M. Lavoie, Effects of ovar- Fecal bacterial community changes associated with isoflavone metabolites in
iectomy on PPAR alpha, SREBP-1c, and SCD-1 gene expression in the rat liver, postmenopausal women after soy bar consumption, PLoS One 9 (10) (2014)
Menopause 15 (6) (2008) 1169–1175. e108924.
[91] A. Vrieze, E. Van Nood, F. Holleman, J. Salojarvi, R.S. Kootte, J.F. Bartelsman, [110] Z. Madak-Erdogan, P. Gong, Y.C. Zhao, L. Xu, K.U. Wrobel, J.A. Hartman,
G.M. Dallinga-Thie, M.T. Ackermans, M.J. Serlie, R. Oozeer, M. Derrien, M. Wang, A. Cam, U.T. Iwaniec, R.T. Turner, N.C. Twaddle, D.R. Doerge,
A. Druesne, J.E. Van Hylckama Vlieg, V.W. Bloks, A.K. Groen, H.G. Heilig, I.A. Khan, J.A. Katzenellenbogen, B.S. Katzenellenbogen, W.G. Helferich, Dietary
E.G. Zoetendal, E.S. Stroes, W.M. de Vos, J.B. Hoekstra, M. Nieuwdorp, Transfer of licorice root supplementation reduces diet-induced weight gain, lipid deposition,
intestinal microbiota from lean donors increases insulin sensitivity in individuals and hepatic steatosis in ovariectomized mice without stimulating reproductive
with metabolic syndrome, Gastroenterology 143 (4) (2012) 913-6 e7. tissues and mammary gland, Mol. Nutr. Food Res. 60 (2) (2016) 369–380.
[92] H.J. Flint, K.P. Scott, P. Louis, S.H. Duncan, The role of the gut microbiota in [111] M. Kwa, C.S. Plottel, M.J. Blaser, S. Adams, The intestinal microbiome and es-
nutrition and health, Nat. Rev. Gastroenterol. Hepatol. 9 (2012) 577–589. trogen receptor-positive female breast cancer, J. Natl. Cancer Inst. 108 (8) (2016)
[93] M.R. Charlton, J.M. Burns, R.A. Pedersen, K.D. Watt, J.K. Heimbach, djw029.
R.A. Dierkhising, Frequency and outcomes of liver transplantation for nonalco- [112] R. Flores, J. Shi, B. Fuhrman, X. Xu, T.D. Veenstra, M.H. Gail, P. Gajer, J. Ravel,
holic steatohepatitis in the United States, Gastroenterology 141 (2011) J.J. Goedert, Fecal microbial determinants of fecal and systemic estrogens and
1249–1253. estrogen metabolites: a cross sectional study, J. Transl. Med. 10 (2012) 253.
[94] P.J. Turnbaugh, F. Backhed, L. Fulton, J.I. Gordon, Diet-induced obesity is linked [113] R.T. Falk, L.A. Brinton, J.F. Dorgan, B.J. Fuhrman, T.D. Veenstra, X. Xu,
to marked but reversible alterations in the mouse distal gut microbiome, Cell Host G.L. Gierach, Relationship of serum estrogens and estrogen metabolites to post-
Microbe. 3 (4) (2008) 213–223. menopausal breast cancer risk: a nested case-control study, Breast Cancer Res. 15
[95] P.J. Turnbaugh, V.K. Ridaura, J.J. Faith, F.E. Rey, R. Knight, J.I. Gordon, The (2) (2015) R34.
effect of diet on the human gut microbiome: a metagenomic analysis in humanized [114] B.J. Fuhrman, H.S. Feigelson, R. Flores, M.H. Gail, X. Xu, J. Ravel, J.J. Goedert,
gnotobiotic mice, Sci. Transl. Med. 1 (6) (2009) 6ra14. Associations of the Fecal Microbiome With Urinary Estrogens and Estrogen
[96] V. Blasco-Baque, M. Serino, J.N. Vergnes, E. Riant, P. Loubieres, J.F. Arnal, Metabolites in Postmenopausal Women, The J. Clin. Endocrinol. Metab. 99 (12)
P. Gourdy, M. Sixou, R. Burcelin, P. Kemoun, High-fat diet induces periodontitis in (2014) 4632–4640.

43

You might also like