Skip to main content
Erschienen in: Advances in Therapy 6/2017

Open Access 19.05.2017 | Review

NAFLD as a Sexual Dimorphic Disease: Role of Gender and Reproductive Status in the Development and Progression of Nonalcoholic Fatty Liver Disease and Inherent Cardiovascular Risk

verfasst von: Stefano Ballestri, Fabio Nascimbeni, Enrica Baldelli, Alessandra Marrazzo, Dante Romagnoli, Amedeo Lonardo

Erschienen in: Advances in Therapy | Ausgabe 6/2017

Abstract

Nonalcoholic fatty liver disease (NAFLD) spans steatosis through nonalcoholic steatohepatis, cirrhosis, and hepatocellular carcinoma (HCC) associated with striking systemic features and excess cardiovascular and liver-related mortality. The pathogenesis of NAFLD is complex and multifactorial. Endocrine derangements are closely linked with dysmetabolic traits. For example, in animal and human studies, female sex is protected from dysmetabolism thanks to young individuals’ ability to partition fatty acids towards ketone body production rather than very low density lipoprotein (VLDL)-triacylglycerol, and to sex-specific browning of white adipose tissue. Ovarian senescence facilitates both the development of massive hepatic steatosis and the fibrotic progression of liver disease in an experimental overfed zebrafish model. Consistently, estrogen deficiency, by potentiating hepatic inflammatory changes, hastens the progression of disease in a dietary model of nonalcoholic steatohepatitis (NASH) developing in ovariectomized mice fed a high-fat diet. In humans, NAFLD more often affects men; and premenopausal women are equally protected from developing NAFLD as they are from cardiovascular disease. It would be expected that early menarche, definitely associated with estrogen activation, would produce protection against the risk of NAFLD. Nevertheless, it has been suggested that early menarche may confer an increased risk of NAFLD in adulthood, excess adiposity being the primary culprit of this association. Fertile age may be associated with more severe hepatocyte injury and inflammation, but also with a decreased risk of liver fibrosis compared to men and postmenopausal status. Later in life, ovarian senescence is strongly associated with severe steatosis and fibrosing NASH, which may occur in postmenopausal women. Estrogen deficiency is deemed to be responsible for these findings via the development of postmenopausal metabolic syndrome. Estrogen supplementation may at least theoretically protect from NAFLD development and progression, as suggested by some studies exploring the effect of hormonal replacement therapy on postmenopausal women, but the variable impact of different sex hormones in NAFLD (i.e., the pro-inflammatory effect of progesterone) should be carefully considered.
Hinweise

Enhanced content

To view enhanced content for this article go to http://​www.​medengine.​com/​Redeem/​2128F060377C0C79​.
Stefano Ballestri and Fabio Nascimbeni contributed equally and are joint first authors.

Introduction

Nonalcoholic fatty liver disease (NAFLD) encompasses the whole spectrum of (predominantly) steatogenic liver disorders spanning steatosis through nonalcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC) [13], associated with striking systemic features [4, 5] and excess cardiovascular and liver-related mortality [69]. Histologically indistinguishable from alcoholic liver disease, and nevertheless observed in the nonalcoholic individual [10], NAFLD is closely linked with insulin resistance (IR) [11] and, bidirectionally, with the metabolic syndrome (MetS) of which it may be both a cause and a consequence [12, 13]. A leading cause of chronic liver disease worldwide and affecting one out of four individuals in the European and north-American general populations [14], NAFLD is highly prevalent in certain groups of individuals carrying either the full-blown or individual traits of the MetS [15]. Moreover, NAFLD carries an excess of health-related expenditures owing to its close connections with progressive liver disease and cardio-metabolic morbi-mortality [9, 1619].
The pathogenesis of NAFLD has variably been conceptualized as two-hit [20], a one-hit [11], or a multistep process [21], and the last is presently the most widely accepted pathogenic model. In its original definition, a first hit leads to steatogenesis and a second one to fibrosis [22]. However, metabolic factors, and particularly IR, invariably account for most of the elementary NAFLD histologic lesions in humans, indirectly supporting the outdated and yet conceptually more parsimonious “one-hit” theory [11, 12].
Evidence both in animals and humans supports the notion that female sex is protected from dysmetabolic traits thanks to young individuals’ ability to partition fatty acids towards ketone body production rather than very low density lipoprotein (VLDL)-triacylglycerol [23], and to sex-specific browning of white adipose tissue which contributes in protecting female mice from experimental NAFLD associated with methionine choline deficient diet [24].
In 1980, Ludwig reported that NASH was common among elderly women with metabolic comorbidity [25]. However, we now know that NAFLD more often affects men [15, 26] and that premenopausal women are equally protected from NAFLD development as they are from cardiovascular disease (CVD) [27, 28]. Recent studies in the overnourished zebrafish model have shown that ovarian senescence, via hypoestrogenemia, facilitates both the development of massive hepatic steatosis and the fibrotic progression of liver disease [29]. Consistently, estrogen deficiency, by potentiating hepatic inflammatory changes, hastens the progression of disease in a dietary model of NASH developing in ovariectomized mice fed a cholesterol-rich hyperlipidic diet [30]. Collectively, these findings suggest that hormonal changes, rather than those multiple physiologic derangements associated with aging per se [31], are a major determinant of progressive NAFLD in human menopause.
Obesity and obesity-related diseases, such as type 2 diabetes (T2D), MetS, and atherosclerosis, are complex conditions driven by genetic and environmental factors, in which a sexual dimorphism has been clearly established. Here, we have reviewed current evidence suggesting that NAFLD is a sexually dimorphic condition, too. We hypothesized that the higher incidence of disease in men and the worse outcome in postmenopausal women, i.e., the “sexual dimorphism” of NAFLD, might offer clues useful in expanding our understanding of the pathogenesis and providing hints for prevention and treatment of NAFLD. Given the potential research and clinical implications of sexual dimorphism in NAFLD, we carried out a systematic review of the literature by using the following keywords: steatosis OR fatty liver AND gender OR sex OR menopause. On these grounds, this narrative review is aimed to highlight how sex modulates the development and progression of NAFLD and to pinpoint what the role of menarche and menopause is. Special emphasis is conferred to CVD risk. The relationship of polycystic ovary syndrome (PCOS) to NAFLD has recently been covered elsewhere [32] and is outside the scope of this review.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not involve any new studies of human or animal subjects performed by any of the authors.

Epidemiology of NAFLD

Gender and reproductive status modulate the risk of developing NAFLD and NASH with/without advanced fibrosis [15].

Risk of NAFLD

Incidence

Evidence from longitudinal studies suggests that the incidence of NAFLD is higher in the male as compared to the female gender (Table 1) [3343]. One study investigating the incidence of NAFLD in women as a function of reproductive status found incidence to be higher in menopausal (7.5%)/postmenopausal (6.1%) women as compared to premenopausal (3.5%) women. However, postmenopausal status was associated with an increased risk of incident NAFLD at univariate but not at multivariate analysis adjusted for age, metabolic syndrome, and weight gain [37]. The incidence of NAFLD in women taking hormonal replacement therapy (HRT) (5.3%) was higher than in premenopausal (3.5%) women but lower than in menopausal women (7.5%). HRT was not associated with increased risk of incident NAFLD either at univariate or at multivariate analysis [37]. Indeed other longitudinal studies suggest that estrogens are a protective factor for the development of NAFLD. An Italian multicentric study on 5408 healthy women who had had hysterectomies, randomly assigned to receive tamoxifen (an estrogen inhibitor) or placebo for 5 years, showed that tamoxifen was associated with higher risk of development of NAFLD/NASH especially in overweight/obese women [44]. Finally a small double-blind, randomized placebo-controlled trial on 50 women with T2D showed that HRT containing low-dose estradiol and norethisterone significantly reduced serum levels of liver function enzymes, potentially owing to a reduced liver fat accumulation [45].
Table 1
Gender impacts on incident and prevalent NAFLD. Evidence from longitudinal and cross-sectional studies
Study
Method
Findings
Conclusion
Study population
Diagnosis of FL
Incidence (longitudinal studies)
Independent predictors
Kojima et al. [33]
35,519 Japanese subjects (health check-up), 65.2% M, mean age 45.8 years, F-up 12 years
US
14.3%
M sex (OR 1.7), metabolic factors (BMI OR 6.3)
M sex and metabolic factors are independent risk factors for the incidence of NAFLD
Hamaguchi et al. [34]
3147 healthy Japanese (1694 M), aged 21–80 years, mean F-up 414 days
US
10%; in M (14%) > F (5%)
Age (only in F), weight gain, and MetS
NAFLD is more incident in men than in women
Suzuki et al. [35]
1537 Japanese subjects (1352 M), mean age 35 years, mean F-up 60 months
AST, ALT
31/1000 person-years
M sex overall and in subjects <40 years (HR 4.6); metabolic factors
M sex and metabolic factors are independent risk factors for the incidence of NAFLD
Tsuneto et al. [36]
1635 Nagasaki atomic bomb survivors (606 M), mean age 63 years, F-up 11.6 years
US
19.9/1000 person-years (22.3 in M > 18.6 in F); M (38%) > F (25%) before 50 years
Obesity, hypertension, and high-TG (gender NS)
Gender is not an independent risk factor for incident NAFLD
Hamaguchi et al. [37]
1603 Japanese women, aged 21–80 years, mean F-up 414 days
US
5% in F; in postmenopausal (6.1–7.5%) and under HRT (5.3%) > premenopausal (3.5%)
Age (only in premenopausal), weight gain, and MetS
There is a gradient in the incidence of NAFLD in women: postmenopausal > HRT > premenopausal
Zhou et al. [38]
507 Chinese NAFLD-free participants, median F-up 4 years
US
9.1%; in M 7.3% > F 9.7%
Age, metabolic factors (gender NS)
Gender is not an independent risk factor for incident NAFLD
Zelber-Sagi et al. [39]
147 Israeli subjects, mean age 51.2 years, F-up 7 years
US
19.0% (2.7%/year)
Weight gain and HOMA (gender NS)
Gender is not an independent risk factor for incident NAFLD
Sung et al. [40]
2589 Korean subjects, mean F-up 4.37 years
US
34.7/1000 person-years; in M (23.4%) > F (9.7%)
MetS traits (gender NS)
Gender was not an independent risk factor for incident NAFLD
Xu et al. [41]
5562 non-obese Chinese subjects, mean age 43 years, F-up 5 years
US
8.9%
M sex, younger age, and metabolic factors
M sex is a risk factor for the incidence of NAFLD at multivariate analysis
Wong et al. [42]
565 Hong Kong subjects, mean age 48 years, median F-up 47 months
1H NMR
3.4%/year
MetS (M sex predictor only at univariate analysis)
M sex is a risk factor for the incidence of NAFLD at univariate analysis
Yun et al. [43]
37,130 Korean subjects, mean age (M 39.4, F 38.6 years), 46% M, F-up 2 years
US
M (44.5/1000 person-years) > F (20.4/1000 person-years)
WC gain in both sexes
Visceral obesity strongly predicts NAFLD in either gender
 
Study population
Diagnosis of FL
Prevalence (cross-sectional studies)
Independent predictors
 
Kojima et al. [33]
39,151 Japanese subjects (health check-up), mean age 45.8 years, 61% M
US
M (26%) > F (12.7%)
NA
NAFLD is more prevalent in M gender
Shen et al. [46]
4009 Chinese administrative officers non-drinkers, aged 20–81 years, 64% M
US
M (13.3%) > F (2.7%) before 50 years; similar after 50 years
M sex, age >50 years, and metabolic traits
M sex and metabolic factors are independent risk factors for the prevalence of NAFLD
Fan et al. [47]
3175 Chinese subjects from Shanghai, mean age 52years, 38.4% M
US
M (19.2%) > F (11.3%) before 50 years; F > M after 50 years
M sex (OR 2.7) and metabolic factors
M sex and metabolic factors are independent risk factors for the prevalence of NAFLD
Chen et al. [48]
3245 adults in a Taiwan rural village, aged ≥18 years, 45.3% M
US
M (19.7%) > F (10.7%)
M sex and metabolic factors; age ≥65 years negative predictor
M sex and metabolic factors are independent risk factors for the prevalence of NAFLD
Park et al. [49]
6648 Korean subjects, aged ≥20 years, 53% M
US
M (22.6%) > F (6.8%) before 50 years; similar after 50 years
Menopause status and increasing age in F
Menopause is a strong risk factor for prevalent NAFLD
Zelber-Sagi et al. [50]
352 Israeli subjects, mean age 51 years, 53.4% M
US
M (38%) > F (21%)
M sex (after adjustment for obesity)
M sex and metabolic factors are independent risk factors for prevalent NAFLD
Zhou et al. [51]
3543 inhabitants of 6 urban and rural areas in China, aged >7 years, 37% M
US
M (13.8%) > F (7.1%) before 50 years; the opposite after 50 years
M sex and metabolic factors
M sex and metabolic factors are independent risk factors for prevalent NAFLD
Li et al. [52]
9094 Chinese subjects (medical check-up), aged >18 years, 52% M
US
M (18.9%) > F (5.7%); increased with age in both sexes <50 years
M sex, increasing age, BMI, and other MetS features
M sex and metabolic factors are independent risk factors for prevalent NAFLD
Caballeria et al. [53]
766 Spanish individuals, aged 17–83 years, 33.4% M
US
M (42.2%) > F (20.3%)
M sex, increasing age, and metabolic factors
M sex and metabolic factors are independent risk factors for prevalent NAFLD
Hu et al. [54]
7152 employees of Shanghai work-units, aged 18–65 years, 60.5% M
US
M > F in the same age group, peaking at 50–65 years; in F > 50 years doubled
High TG level was the strongest predictor in M while obesity the strongest one in F
Risk factors for prevalent NAFLD are gender-specific
Wong et al. [55]
922 individuals from Hong Kong population, aged 19–72 years, 58% F
1H MRS
M (36.8%) > F (22.7%); in M peaking at 40 years; in F increasing after menopause
Metabolic factors (M sex and older age NS)
Gender is not an independent risk factor for prevalent NAFLD
Hamaguchi et al. [37]
4401 Japanese subjects (health check-up), aged 21-80 years, 51.6% men
US
M (24%) and postmenopausal F (15%) >remenopausal F (6%); increased with age in F only
Postmenopause and HRT NS (age and MetS adjusted); aging in premenopausal F only (weight gain and MetS adjusted)
Menopause and HRT are not independent risk factors for prevalent NAFLD
Eguchi et al. [56]
8352 Japanese subjects (health check-ups), aged 21–86 years, 51.8% M
US
M > F at all ages
Metabolic factors (age >50 years in F only)
Gender is not an independent risk factor for prevalent NAFLD
Younossi et al. [57]
11,613 American participants (from NHANES III)
US
19%
F sex and younger age for lean NAFLD; the opposite for NAFLD with BMI ≥25
F sex is a risk factor for lean NAFLD
Lazo et al. [58]
12,454 American subjects (from NHANES III), aged 20–74 years
US
M (20.2%) > F (15.8%)
Prevalence in M (20.2%) > F (15.8%) (adjusted for age, race, BMI, T2D)
M gender is associated with an increased NAFLD prevalence
Wang et al. [59]
4226 Chinese subjects >60 years vs 3145 controls <60 years from the same cohort
US
M (32%) > F (9%) before 60 years; similar after 60 years
M sex (only in those <60 years) and metabolic factors
M sex and metabolic factors are independent risk factors for prevalent NAFLD
Xu et al. [41]
6905 non-obese (BMI < 25) Chinese subjects, 63% M
US
7.3%
M sex, younger age, and metabolic factors
M sex and metabolic factors are independent risk factors for prevalent NAFLD
Yan et al. [60]
3762 Chinese residents, aged 20–92 years, 61.9% M
US
M (45%) > F (30%); higher before 50 years but similar after
Metabolic factors (M sex predicted only at univariate analysis)
M sex is a risk factor for prevalent NAFLD at univariate analysis only
Chiloiro et al. [61]
2974 South Italian subjects, aged 30–89 years, 56.5% M
US
M (37%) > F (26%)
MetS and its features, BMI, visceral, and subcutaneous fat in both M and F
Risk factors for prevalent NAFLD are not gender-specific
Foster et al. [62]
3056 American multiethnic subjects, mean age 63 years, 55% F
CT
NA
F sex protective in all and AAs; not in Caucasians and Hispanics. Younger age in all, Caucasians and Hispanics; not in AAs
F sex is spared from prevalent NAFLD
Yang et al. [63]
2256 Chinese subjects (health check-up), mean age 62.04 years, 87.9% M
US
M (28.4%) > F (20.3%)
Younger age, RDW and metabolic factors (gender NS)
Gender is not an independent risk factor for prevalent NAFLD
Saida et al. [64]
3110 Japanese subjects (health check-up), aged ≥30 years, 59.5% M
US
M (45%) > F (23%)
Weight gain ≥10 kg since the age of 20 years regardless of sex
Gender is not an independent risk factor for prevalent NAFLD
Wang et al. [65]
25,032 Chinese subjects (health check-up), aged 18–94 years, 62% M
US
M (32%) > F (13%); similar in both sexes >60 years
Metabolic parameters in both sexes; increasing age (≥60 years) only in F
Risk factors for prevalent NAFLD are not gender-specific
Schneider et al. [66]
4037 non-Hispanic white, 2746 non-Hispanic black, and 2892 Mexican–American adults in the NHANES III
US
M (15%) > F (10%) among non-Hispanic whites only (age-adjusted)
Gender difference still significant after adjustment for BMI and WC
M gender is associated with an increased NAFLD prevalence independent of visceral obesity
Xiao et al. [67]
18,676 Chinese subjects, mean age 40.6 years, 55.4% M
US
M (33%) > F (8%)
M sex and metabolic factors
M sex is an independent risk factor for prevalent NAFLD
Martínez-Alvarado et al. [68]
846 Mexican subjects, mean age 53 years, 49.3% M
CT
M (36.5%) > F (28.4%)
Metabolic factors (MetS in F only)
Gender is not an independent risk factor for prevalent NAFLD
Liu et al. [69]
11,200 Chinese gallstone-free subjects, 46.7% M
US
M (31%) > F (13%)
NA
NAFLD is more prevalent in M gender
Amirkalali et al. [70]
5023 Iranian participants, mean age 45.35 years, 56.7% M
US
F (46%) > M (42%); peaking in M 40–60 years, in F >60 years
Age, MetS, and its factors (higher ORs in F)
Gender is not an independent risk factor for prevalent NAFLD
Nishioji et al. [71]
3271 Japanese subjects (health check-up), 44.2% M
US
M > F for all ages, except in obese ≥70 years
Metabolic factors in M and F
Risk factors for prevalent NAFLD are not gender-specific
Huang et al. [72]
8626 Chinese subjects ≥40 years
US
M (28.7%) ~ F (28.1%)
NA
No gender specific difference in prevalent NAFLD
Fattahi et al. [73]
2980 Iranian subjects, aged ≥18 years, 31.3% M
US
M (32.9%) > F (27.4%)
NA
NAFLD is more prevalent in M gender
Motamed et al. [74]
5052 Iranian subjects, aged ≥18 years, 56.6% M
US
F (44.2%) > M (40.1%)
Age, metabolic factors; F sex protective
F sex is spared from prevalent NAFLD
AAs African-Americans, BMI body mass index, F female/s, FL fatty liver, F-up follow-up, 1 H MRS proton magnetic resonance spectroscopy, HOMA homeostasis model assessment, HRT hormone replacement therapy, M male/s, MetS metabolic syndrome, NA not assessed, NAFLD nonalcoholic fatty liver disease, NHANES III National Health and Nutrition Examination Survey III, NS not significant, OR odds ratio, RDW red blood cell distribution width, T2D type 2 diabetes, TG triglycerides, US ultrasound, WC waist circumference
Overall, data from epidemiological longitudinal studies have shown a key role of weight gain, presence of MetS and its single traits as independent predictors of the development of NAFLD [3343]. Notably, some studies have found that male sex was associated with incident NAFLD independently of age and metabolic factors [33, 35, 41]. Moreover, two Asian studies have reported a specific role of age according to gender and reproductive status in modulating the risk of incident NAFLD. One study showed that age was an independent predictor for developing NAFLD only in females [34] and the other one reported that age increased the risk for incident NAFLD in premenopausal but not in postmenopausal women [37].

Prevalence

Despite preliminary studies reporting a higher risk of NAFLD in females, a large body of evidence now definitely supports the notion that the prevalence of NAFLD is higher in men than in women and that gender-specific differences exist in relation to age (Table 1) [33, 37, 41, 4677]. Conflicting with the above notion, two large sample community Iranian studies reported a higher prevalence of NAFLD in women than in men [70, 74]. However, these findings might, at least in part, be accounted for by women having markedly higher rates of prevalence of obesity compared to men in these studies. Consistent with the view reported above, a higher prevalence of fatty liver has been also observed in male obese children and adolescents than in female ones [78].
Men commonly display an increasing prevalence of NAFLD during adulthood from young to middle-age, describing an “inverted U-shaped curve” which starts declining after the age of 50–60 years [47, 56]. In women of fertile age, the prevalence of NAFLD is lower than in men owing to the putative protective effect of estrogens which, however, wanes after the menopause. Accordingly, the prevalence of NAFLD in women rises after the age of 50 years, peaks at 60–69 years, and declines after 70 years [47, 54, 56]. As a result of this, after the fifth decade, postmenopausal women compared to men of the same age have a similar [46, 49, 60, 65] or even higher [47, 51] prevalence of NAFLD. In agreement with these findings, a multicenter study from northern Italy found that men with NAFLD were approximately 10 years younger than women with this condition [79], a finding compatible with the hypothesis that premenopausal women are “protected” from developing NAFLD. Conversely, NAFLD is more prevalent in postmenopausal [37, 8082] and PCOS-affected women than premenopausal ones [80]. Consistently, a large cross-sectional population-based survey in northeast Germany on 808 women aged 40–59 years showed that menopause status was independently associated with hepatic steatosis after adjustment for metabolic factors [83]. Interestingly, women with NAFLD exhibit a significantly lower concentration of serum estradiol, which is the principal active estrogen, than NAFLD-free (premenopausal, postmenopausal, and PCOS) controls [80]. A lower prevalence of NAFLD, as well as of MetS, has been reported in postmenopausal women receiving HRT compared to those not receiving it, which suggests that HRT probably protects from NAFLD. Moreover, in this study postmenopausal women with NAFLD who received HRT had lower frequency of insulin resistance as well as MetS and showed reduced serum levels of liver enzymes and ferritin [84].
Most studies using multivariate analysis have shown that male sex is associated with an increased prevalence of NAFLD independently of age and metabolic conditions [41, 46, 48, 50, 51, 53, 58, 62, 63, 66, 67, 74]. Some studies have shown an independent, positive association between NAFLD and increasing age in both sexes [46, 5053, 70, 74] or in females only [65], while others reported an inverse association [41, 48, 62, 63]. At variance, other studies have shown that sex differences in the prevalence of NAFLD were mostly accounted for by metabolic factors [36, 37, 55, 56, 60, 63, 64]. Finally, some studies failed to investigate the role of gender given that a separate multivariate analysis according to sex was not performed [34, 49, 54, 56, 61, 65, 68, 70, 71].
In addition to and independent of the role of MetS and its components, menopausal status [49, 83, 85] and increasing age [49, 56, 82] have all been consistently identified as strong risk factors for NAFLD in women. Only in a few studies was the association between menopausal status and NAFLD no longer significant after adjustment for age and metabolic factors [37, 81], indicating that menopause predisposes to developing NAFLD via incident dysmetabolic traits which typically appear in the postmenopausal age.
A recent cross-sectional study conducted in a population of postmenopausal women concluded that MetS, abdominal obesity, and IR are risk factors for the development of NAFLD while higher adiponectin levels protect from developing it [86]. These findings indicate that NAFLD modifiers in this specific population of women closely mirror those found in the general population.
In lean subjects [body mass index (BMI) less than 25 kg/m2)] NAFLD has been associated with younger age [41, 57] and either male [41] or female gender [57].

Risk of NASH and Fibrosis Progression

One large sample study based on an electronic health file database reported that the risk of “recorded” NAFLD/NASH diagnosis increased linearly with BMI, was higher in males than females and in those with T2D [87]. However, the proportion of true NASH cases among the “recorded” diagnosis of NAFLD/NASH was unclear.
A longitudinal study in subjects with biopsy-proven NAFLD found that sex was not an independent risk factor for the progression of fibrosis [88]. In agreement, a systematic review has shown that age and hepatic necroinflammation are the only independent predictors of the development of advanced fibrosis in NASH patients, while other parameters such as MetS features and sex are not [89].
However, conflicting with the above studies, data from cross-sectional studies (Table 2) [29, 57, 65, 9097], which are based, in the majority of cases, on a histological diagnosis of NASH, tend to suggest that the risk of NASH and advanced fibrosis is indeed higher in females than males independent of metabolic factors [65, 90, 93, 94], and only a few studies conflict with the above findings [91, 92, 95].
Table 2
Impact of gender on NASH/fibrosis prevalence
Study
Method
Cross-sectional studies
Findings
Conclusion
Study population
Diagnosis of NASH
Prevalence
Independent predictors
Singh et al. [90]
71 consecutive Asian-Indian NASH patients, aged 9–57 years, 76.1% M
Biopsy
NA
F sex for fibrosis stage
F sex is an independent risk factor for fibrosis
Hossain et al. [91]
432 American NAFLD patients, mean age 43.6 years, 22.9% M
Biopsy
26.8% NASH and 17.4% moderate-to severe fibrosis
M sex for NASH and moderate-to-severe fibrosis in addition to ethnicity, ALT, AST, metabolic factors
M gender is a strong independent risk factor for both NASH and fibrosis
Al-hamoudi et al. [92]
1312 Saudi Arabian inpatients, mean age 44.7 years, 51.0% M
US
16.6% NASH (by ALT >60 U/L)
M sex, young age and low total CH predicted high ALT in NAFLD
M gender is a strong independent risk factor for hypertransaminasemic NAFLD
Bambha et al. [93]
1026 adults (NASH CRN Database), mean age 48.8 years, 37% M
Biopsy
61% NASH and 29% advanced fibrosis
F sex for NASH and advanced fibrosis; increasing age for advanced fibrosis; plus metabolic factors
F sex is an independent risk factor for NASH and fibrosis
Younossi et al. [57]
11,613 American participants (from NHANES III)
US
12% NASH (by grade 2–3 US-FL + high ALT, AST, and/or T2D/IR)
Younger age and metabolic factors (gender NS)
Gender is not an independent risk factor for NASH
Tapper et al. [94]
358 NAFLD patients, 62.9% M
Biopsy
NASH and advanced fibrosis > in F vs M (45% vs 30%; 23% vs 14%)
F sex, BMI, and ALT for NASH; age, AST, and APRI for advanced fibrosis
F sex is an independent risk factor for NASH and fibrosis
Wang et al. [65]
25,032 Chinese subjects (health check-up), aged 18–94 years, 62% M
US
NASH with advanced fibrosis (by BAAT score, AST/ALT) > F vs M
NA
Prevalence of NASH with advanced fibrosis is higher in F sex
Yang et al. [95]
541 American patients with NASH, mean age 48 years, 35.1% M
Biopsy
100% NASH; 22% advanced fibrosis
M sex, postmenopausal F status (premenopausal F reference; borderline P), pre-T2D/T2D for fibrosis
M sex and postmenopausal status are independent risk factors for fibrosing NASH
Turola et al. [29]
244 females and 244 age-matched males with NAFLD
Biopsy
F2–F4 fibrosis
Menopause, metabolic factors, and NASH for F2–F4 fibrosis in F with NAFLD
Menopause is strongly associated with fibrosing NASH
Klair et al. [96]
488 postmenopausal NAFLD subjects
Biopsy
Advanced fibrosis 38.4% in premature menopause F vs 32.7% in other F
Premature menopause and time from menopause for advanced fibrosis (adjusted for age, race, metabolic factors)
The longer the duration of postmenopausal status the higher the risk of NASH
Yang et al. [97]
1112 American NAFLD patients (160 premenopausal and 489 postmenopausal F; 463 M)
Biopsy
NASH > in pre/postmenopausal F (62%) vs M (50%)
Lobular inflammation risk increased in (1) premenopausal F > M and postmenopausal F, (2) oral contraceptives and HRT users (adjusted for covariates of liver metabolic stress)
Fertile age and estrogen use may predispose to more necroinflammatory NASH variants
ALT alanine aminotransferase, AST aspartate aminotransferase, APRI AST-to-platelet ratio index, BMI body mass index, F female/s, FL fatty liver, F-up follow-up, HRT hormone replacement therapy, M male/s, MetS metabolic syndrome, NA not assessed, NAFLD nonalcoholic fatty liver disease, NASH nonalcoholic steatohepatitis, NHANES III National Health and Nutrition Examination Survey III, NS not significant, T2D type 2 diabetes, US ultrasound, WC waist circumference, years years
As is easily foreseeable, obese and postmenopausal women, compared to premenopausal and non-obese women, suffer from a remarkably higher prevalence of NAFLD and NASH as a result of a worse metabolic profile [82]. At variance, a recent study reported that, compared to men and postmenopausal women, the risk of lobular inflammation and hepatocyte injury was significantly increased both in premenopausal women and in those taking synthetic hormones such as oral contraceptives and HRT [97]. Given the supposed effects of estrogens on metabolic health and liver injury, the findings of this study appear counterintuitive. It is noteworthy that the study by Yang et al. has several limitations, including some important sources of potential bias, such as the restricted enrollment at tertiary academic centers, menopausal category and synthetic hormone use were self-reported, and information on cumulative estrogen and/or progesterone exposure and serum hormonal levels were lacking. Of note, the authors highlight that, despite increased liver injury and inflammation, premenopausal women were at decreased risk of liver fibrosis compared with men and postmenopausal women. Moreover, sensitivity analyses separately assessing the impact of progesterone use and estrogen use clearly suggested that only the former was associated with liver damage. Collectively these findings provide novel hints regarding a potential multifaceted impact of sex hormones in NAFLD.
Consistent with the hypothesis that estrogens do exert a beneficial effect on NAFLD, menopause has been independently associated with significant fibrosis both in women with NAFLD and in an experimental zebrafish steatosis model [29]. A recent study has shown that men display a higher risk of advanced fibrosis compared to premenopausal women, while after menopause both sexes show a similar severity of liver fibrosis, suggesting that estrogens protect from the development of fibrosis [95]. A subsequent study limited to non-obese women with biopsy-proven NAFLD confirmed that, even in non-obese NAFLD patients, postmenopausal women still had more severe fibrosis, when compared to premenopausal subjects [98]. Accordingly, a study conducted in 488 postmenopausal women with biopsy-proven NAFLD has shown that, independently of age and metabolic factors, the longer the estrogen deficiency in postmenopausal status is (i.e., premature menopause and time from menopause), the higher the risk of fibrosis is [96].
The role of gender in influencing liver-related mortality in NAFLD is still uncertain, although some studies have reported that men are at an increased risk [99101].

Risk of Hepatocellular Carcinoma

A universal feature of HCC is the striking male prevalence, with a male/female ratio averaging 2:1 to 7:1; the latter proportion is more often found in HBV-related HCC [102]. Nevertheless, compared to viral etiologies of HCC, NAFLD-related HCC was found to be associated with the lowest male/female ratios in one study [103]. NAFLD-related HCC may also occur in non-cirrhotic livers, but this seems to be more likely in men [104]. The sexual dimorphism in HCC is also maintained regarding prognosis, with women showing better survival rates [102, 105]. However, menopause seems to attenuate these advantages [102, 105].
In summary, the incidence of NAFLD is higher in men than in women, and some longitudinal studies indicate that male gender is an independent predictor of NAFLD development. The prevalence of NAFLD is globally higher in men than women, but after menopause women display a similar or even a higher prevalence compared to men, a finding supporting a protective effect of estrogens. In cross-sectional studies, male gender and menopausal status have often been associated with the risk of NAFLD, independent of age and metabolic factors. The prevalence of NASH and advanced fibrosis has been found to be higher in postmenopausal women than in men; however, longitudinal studies have failed to support a role for gender in influencing the progression of liver fibrosis. Finally, HCC is definitely more common in men than in women in cases due to viral etiology; NAFLD may probably lower the male/female ratio in the risk of developing HCC and it is possible that gender affects the prognosis of HCC.

NAFLD and CVD

NAFLD is increasingly recognized as a multisystem disease [5]. A growing body of evidence suggests that NAFLD (assessed by liver enzymes, imaging, or biopsy) is associated with increased incidence and prevalence of subclinical and clinical CVD, mainly coronary heart disease (CHD), independently of age, gender, and metabolic factors, as recently reviewed in detail elsewhere [106, 107]. In the general population, male sex is more prone to incident CHD under the age of 50 years compared to women but, after menopause, the incidence in women dramatically increases to approach that of men [108].

Gender-Specific Risk of CVD in Studies on NAFLD and CVD

Several longitudinal studies investigating the association between CVD and NAFLD have reported multivariate analysis models, which have been adjusted for multiple confounders, including sex; as a result of this, the influence of gender in the risk of CVD cannot be evaluated in such studies (reviewed in [106, 107]). A large population-based American study (NHANES III) found that NAFLD assessed with ultrasound, together with male sex, age, race, and metabolic factors independently predicted incident CVD [109]. Another study addressing CHD as a prespecified outcome found that patients with NAFLD had a higher 10-year risk for CHD, as calculated by the Framingham risk score, than the matched control population. This estimated risk was higher in men than in women and more CVD events were reported among men than women at the end of 10 year follow-up, although this finding was not statistically significant [110].
Many cross-sectional studies in which the diagnosis of NAFLD was based on liver imaging studies (either ultrasound or CT scanning) showed an independent association of male sex with coronary artery calcifications [111] as well as significant CHD [112115].

Gender Differences in the Association Between NAFLD and CVD

Studies in which the diagnosis of NAFLD was based on surrogate indices, such as otherwise unexplained raised liver enzymes, found that sex modulates the association of NAFLD with incident CVD/mortality. For example, most population-based cohort and meta-analytic studies reported an independent association between raised GGT and incident CVD in both sexes [116]; conversely, a population-based cohort study from Germany found that increased GGT was associated with higher risk of all-cause and CVD mortality only in men but not in women, and this association was stronger in men who also had ultrasound scanning findings compatible with steatosis [101]. Increased ALT has been variably associated with incident CVD either in both sexes [117, 118] or in men only [119]. A recent study found that ALT levels independently predicted insulin sensitivity only in women, suggesting that this gender-specific association might explain the sex difference in the predictive role of increased ALT for CVD [120]. Finally, a recent large Korean cross-sectional study reported that men had more prevalent ultrasonographic fatty liver disease, carotid plaques, and increased carotid intima-media thickness (IMT) values than women, but ultrasonographic fatty liver disease independently predicted subclinical carotid atherosclerosis (IMT and plaques) in women only [121].
No gender difference has been reported in the association between NAFLD assessed with liver ultrasound and incident CVD/mortality. A study based on a national Danish registry showed that patients with a hospital discharge diagnosis of NAFLD had a higher all-cause mortality, including liver and CVD related, which was similar among sexes [122]. A study conducted in a community-based Japanese cohort of 1637 apparently healthy subjects found that the diagnosis of NAFLD based on ultrasound findings was a predictor of CVD in both men and women [123].
The only study carried out in postmenopausal women found a significant correlation between NAFLD (based on CT scanning findings) and prevalence of coronary artery calcification (CAC); however, NAFLD was not independently associated with CAC in these postmenopausal women [124].
In summary, male patients carry an increased risk of CVD; moreover, NAFLD seems to be associated with CVD independently of metabolic factors in both sexes. Few data are available in postmenopausal women and studies should specifically be conducted to ascertain whether NAFLD is a specific/independent cardiovascular risk factor in this population of patients.

Role of Gender, Reproductive Status, and Age in the Heterogeneity of NAFLD Pathobiology

Although NAFLD may be found in either gender from infancy to old adulthood, gender and reproductive status modulate the susceptibility to development and progression of disease [125]. Indeed age, sex, and fertility exert a variable impact on those general pathogenic mechanisms which are involved in NAFLD. Here we will specifically examine how body fat distribution, obesity and local hypercorticolism, steatogenesis and lipidomics, oxidative stress and antioxidant mechanisms, endotoxins, immune response, and fibrogenesis are affected by gender and sex hormones as a result of reproductive status and age.

Factors Associated with NAFLD are Different in Men and in Women

A pioneering study supporting the notion that gender-dimorphic risk factors are associated with NAFLD was published in 2000. This study, by evaluating 199 individuals, found that elevated BMI was an independent predictor of fatty liver in either sex. Glucose area under the curve and a central-type body fat distribution predicted fatty liver only in women [126]. Similarly, insulin sensitivity has also been reported to be strongly associated with gender-dimorphic risk factors, i.e., fasting insulin and leptin levels (but none of the liver enzymes) in men versus BMI and ALT in women [120]. Of note, Suzuki et al. demonstrated that the associations between anthropometric measures (regional adiposity) and degree of fibrosis clearly differ between premenopausal women and postmenopausal women/men [127].

NAFLD Epidemiology and Physiopathology are Modulated by Age at Menarche and Postmenopausal Status

In women, a complex interaction including genetic polymophisms, dietary habits, endogenous sex hormones, age at menarche, menopausal status, dysmetabolic traits, and HRT modulates the risk of developing NAFLD, NASH, and fibrosis [84, 128130].
Early menarche has been associated with higher alanine aminotransferase, C-reactive protein, triglyceride levels, BMI, waist circumference, adult diabetes, cardiovascular morbidity and mortality, advanced liver disease, and HCC [131]. A recent Chinese study conducted in postmenopausal women has identified early menarche as a potential risk factor for NAFLD later in life; consistently, late menarche protects from NAFLD [130]. These associations were significantly attenuated after adjustment for current BMI or HOMA-IR, suggesting that obesity and insulin resistance may partly mediate the association between age at menarche and NAFLD [130]. The biological mechanism linking early menarche with increased risk of NAFLD is far from being clearly elucidated. It has been suggested that early maturation may determine a longer duration of positive energy balance and a greater accumulation of body fat [132]. Consistently, a large cross-sectional study among middle-aged Korean women confirmed that the inverse association between age at menarche and NAFLD was partially mediated by adiposity [133]. Again, a recent study from the American CARDIA cohort showed that early menarche was associated with NAFLD and visceral and subcutaneous abdominal ectopic fat depots in middle adulthood; these associations were attenuated after adjustment for weight gain between young and middle adulthood [131]. Finally, a Chinese study suggested that the presence of central obesity and MetS, but not NAFLD, after menopause was predicted by longer duration of menstruation and early menarche [134].
Compared to the fertile age, menopause increases the risk of NAFLD and liver fibrosis [135] via long-standing estrogen deficiency associated with ovarian senescence and dysmetabolic features such as T2D, hypertriglyceridemia, and central obesity [29, 81]. Consistently, HRT protects from NAFLD development [84], and oophorectomy in young women with endometrial cancer independently increases the risk of NAFLD together with the development of T2D and hypercholesterolemia [136]. These findings are in agreement with a study suggesting that, in HCV infection, increasing severity of fibrosis is associated with a higher BMI, advanced steatosis, and the menopause and that, conversely, menopausal women receiving HRT exhibit a lower stage of fibrosis [137]. Collectively, these data support the notion that estrogens have antifibrogenic properties in humans. This antifibrotic activity may occur by triggering anti-inflammatory, antioxidant, and antiapoptotic molecular pathways [138], which are mimicked by exercise training [139].
Of concern, however, young women in their reproductive age and those exposed to female synthetic hormones (oral contraceptive or HRT) are not completely spared the risk of developing NAFLD and, indeed, they tend to have more severe hepatocyte injury and inflammation. Notably, despite the possibility of enhanced hepatocellular damage, premenopausal women have been consistently reported at decreased risk of liver fibrosis compared to men and postmenopausal women. Moreover, sex hormones exert complex and variable effects on human NAFLD; indeed, the detrimental pro-inflammatory impact may be conveyed by progesterone, but not estrogen [97].
Table 3 summarizes the physiological role of estrogen, progesterone, and androgens according to gender, age, reproductive status, and obesity [140157].
Table 3
Physiological role of hormones
 
Men
Women
Both sexes
Obesity/type 2 diabetes
Estradiol
Estradiol in men is essential for modulating libido, erectile function, and spermatogenesis [140]
A study conducted in healthy men suggests that estradiol protects from NAFLD [141]
Endogenous estrogens are master regulators of lipid metabolism and inhibit inflammation, vascular cell growth, and plaque progression in premenopausal women [142]
The loss of estrogens which occurs postmenopausally is associated with a modest increase in LDL cholesterol with either no change or a small decrease in HDL cholesterol. Estrogen administration decreases LDL cholesterol and Lp(a) levels while increasing triglycerides and HDL cholesterol levels, but these effects are dependent on the dose and route of administration [143]
Estrogens improve inflammation related to metabolic dysfunction (“metaflammation”). Further to a direct downregulation of inflammatory pathways, this effect is also mediated by metabolic amelioration [144]
Obesity is associated with hyperestrogenism which, in turn, increases the risk of breast cancer in men [145]
Progesterone
Progesterone has important effects in regulating male fertility by affecting the energetic homeostasis of sperm [146]
Progesterone has a major role in the ovarian and menstrual cycle; moreover it exerts an immuno regulatory function; regulates the contraction of human intestinal smooth muscle cells and the motility of various human cell types [147]
Progesterone is an independent predictor of insulin resistance in girls [148]
Progesterone has been potentially implicated as a therapeutic adjunct in many clinical conditions such as traumatic brain injury, Alzheimer’s disease, and diabetic neuropathy [147]
Increasing levels of progesterone have been associated with the development of systemic insulin resistance [149]
Little is known regarding the role, if any, of serum progesterone in NAFLD
Androgens
The synthesis of testosterone is key to male fertility. A negative feedback finely regulates the secretion of hormones at the levels of hypothalamic-pituitary–gonadal axis. Congenital or acquired disturbances of this axis will lead to hypogonadism and thus impair male fertility [150]. Testosterone has no significant correlation with NAFLD in a study from China [141]. However, it is an independent predictor of insulin resistance in boys [148]
Androgens have important biological roles in young women, influencing bone and muscle mass, vascular health, cognition, mood, well-being, and libido [151]
However, testosterone deficiency in young women may pass underdiagnosed because of generally nonspecific symptoms and inaccuracy of testosterone measurement [152]. The primary indication for the prescription of testosterone for women is loss of libido [153]
Sarcopenia, namely the decline in muscle mass and strength which occurs with ageing, has been associated with a deficiency in both 17β-estradiol and testosterone, two sex hormones which act on satellite cells. These remain quiescent throughout life and are activated in response to stressful events, enabling them to guide repair and regeneration of the skeletal muscle [154]
Obese men tend to be hypogonadic as a result of the functional suppression of the hypothalamic–pituitary–testicular axis [155]. However, weight loss obtained with either a low-calorie diet or bariatric surgery is associated with the normalization of sex hormone levels exhibiting a significant increase in bound and unbound testosterone and gonadotropin levels and a decrease in estradiol [156]. A study from Japan reported that testosterone levels were inversely associated with diabetes among men but not among women [157]

Obesity and Local Hypercortisolism

Obesity, which is closely linked with NAFLD, mimics hypercortisolism. It is of interest that despite the prevalence of obesity being higher among women than men, the former are somewhat protected from the associated cardiometabolic consequences; however, this wanes after menopause, suggesting a role for estrogens. Mouse models suggest that sexually dimorphic expression and activity of glucocorticoid metabolizing enzymes may have a role in the differential metabolic responses to obesity in males and females [158].
Biochemical, genetic, and therapeutic studies have provided robust evidence for 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) being key in the pathogenesis of NAFLD [159161]. It is of interest, therefore, that 11β-HSD1 gene expression is regulated in a tissue-specific and sexually dimorphic manner. In particular, intact rats exhibit hepatic 11β-HSD1 mRNA levels 18-fold lower in the female than the male [162].

Body Fat Distribution

Regional adiposity displays a typical sexual dimorphism in humans. Women have a larger capacity to store fat in the subcutaneous compartment [163]. Men generally have twice as much visceral fat compared to women for any given fat mass value [164]. This is relevant given that, compared to subcutaneous adipose tissue depots, visceral adipose tissue depots in general display greater secretory capacity and a more pro-inflammatory profile [165]. Moreover, visceral adipose depots release free fatty acids (FFAs) directly into the portal blood and thus potentially overload the liver [166]. Not surprisingly, those phenotypes which occur whenever, due to hormonal effect, peripheral adiposity is relatively restricted and visceral adiposity is expanded (i.e., male obesity and postmenopausal women) have all been associated with NASH, and fibrosing NASH [127, 167169].

Steatogenesis and Lipidomics are Affected by Sex Hormones

Steatosis will invariably occur as a result of an imbalance among enhanced steatogenesis and decreased capacity of oxidation of fatty acids [170]. Moreover, qualitative changes in the hepatic lipidomics may concur with lipotoxicity. Data suggest that all these mechanisms are under the control of sex hormones. For example ovariectomy in rats is associated with increased intrahepatic steatogenesis which occurs through a decreased synthesis of peroxisome proliferator-activated receptor (PPAR), and an increased transcription of genes encoding sterol regulatory element-binding protein 1 [(SREBP-1), a nuclear transcription factor and master regulator of the endogenous synthesis of cholesterol, fatty acids, triglycerides, and phospholipids] and stearoyl coenzyme A desaturase 1 [(SCD1), the rate-limiting enzyme for generating monounsaturated fatty acids (MUFA) from saturated fatty acids protects from hepatocyte lipotoxicity]; all these effects of ovariectomy are prevented by 177β-estradiol replacement, indicating a role for estrogens in the prevention of hepatic fat accumulation [171]. Consistently, hypoestrogenemia is associated with hepatic steatosis through changes in gene expression of molecules related to fat oxidation and lipogenesis; resistance and endurance training prevent this both in rats and human models [172175]. Moreover, a normal activity of stearoyl-CoA desaturase (SCD), the rate-limiting enzyme for generating monounsaturated fatty acids from saturated fatty acids, protects from hepatocyte lipotoxicity. Indeed, in mouse models of NAFLD, either genetic manipulation or dietary changes that inhibit the activity of SCD promote fibrosing NASH via hepatocyte lipotoxicity, despite inhibiting obesity and improving insulin resistance [176, 177]. Interestingly, ovarian hormones are also involved in MUFA biosynthesis, via SCD1 [178].
Excess 16:0 fatty acids associated with de novo lipogenesis from high carbohydrate diet inhibits the synthesis of highly unsaturated fatty acids; this may potentially account for the improved metabolic profile which results from supplementing a hypercaloric diet with preformed eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) [179]. For example, findings from a genetically engineered NASH mouse model fed a Western diet have shown that dietary DHA was superior to EPA in attenuating Western diet-induced hyperlipidemia and hepatic injury and at reversing those deleterious metabolic and histological effects induced in the liver by a Western diet [180]. Interestingly, human (women using the contraceptive pill or HRT and transsexual subjects) and experimental data in rats consistently indicate that sex hormones act to modify plasma and tissue n-3 PUFA content, possibly by altering the expression of desaturase and elongase enzymes in the liver [181]. Collectively, these findings suggest that enzyme activities which are key in the development of a potentially hepatotoxic lipidomic signature are critically controlled by sex hormones. Finally, it is of clinical importance that the therapeutic activity of PPAR-α agonists in obesity and fatty acid oxidation is modulated by sex and estrogens [182].

Oxidative Stress and Antioxidant Mechanisms

Reactive oxygen species (ROS) result from the oxidation of fatty acids within mitochondria and peroxisomes. The antioxidant defenses which physiologically constrain oxidative stress are hindered by nutritional deficiencies or changes in the intestinal microbiome that limit availability of choline [183]; by aging and the content of cysteine in diet, which, in turn, will affect the intrahepatic synthesis of glutathione [184]; and by sex and menopause which both affect the normal metabolism of choline [185].

Endotoxin

Quali-quantitative changes in intestinal microbiota (dysbiosis), which are often associated with dietary indiscretions, have consistently been associated with increased gut permeability which could lead to increased translocation of bacterial products from the intestinal lumen into the portal circulation, thereby triggering chronic inflammation [170]. It this context, it is relevant that in an experimental model of liver failure due to endotoxemia after hepatectomy, sexually mature female rats are more exposed than males to endotoxin-induced liver injury and that ovariectomy abrogates this sexual dimorphism [186]. Whether this also applies to human NAFLD, however, remains to be proven.

Interindividual Variation in Immune Responses is Another Key Liver Disease Modifier in NAFLD

Whether metabolic inflammation is a gender-dimorphic phenomenon has not been explored in a systematic and organized manner. Obesity, however, will typically exhibit changes in the innate and adaptive immune mechanisms [187]. However, how mechanistically such obesity-related dysregulation of immune defenses will impact on the pathogenesis of NASH is not fully elucidated. Experimental data obtained in the ob/ob mouse model suggest a role for natural killer T cells (NKT cells) which may at least partly account for the interindividual variation in immune responses being a key modifier in the development and progression of NAFLD [125]. In this respect, it should be highlighted that sex is a major determinant in the immune response. Supporting this notion, one study in young children reported that immune responses to vaccines were consistently higher or equivalent in girls compared with boys [188].

Liver Fibrosis

Liver fibrosis is the end-stage result of various liver injuries. In recent times, importance has been given to the Hedgehog signaling pathway on the grounds that activation of this pathway will stimulate the proliferation and differentiation of hepatic stellate cells (HSCs) as myofibroblasts (MF-HSCs), and, conversely, inhibiting Hedgehog activity in myofibroblasts derived from HSCs causes them to revert to a more quiescent (namely less fibrogenic) phenotype [125]. Accordingly, Hedgehog ligands and other factors that control fate decisions in HSCs are critical determinants of the development of cirrhosis due to various causes as well of the course of NASH [125]. For example, hepatic expression of Hedgehog ligands and Hedgehog pathway activity progressively increase from simple steatosis, to NASH, and reach highest levels in NASH-cirrhosis [189]. Of major significance to the pathogenesis of NAFLD, the Hedgehog pathway is strongly regulated by lipids [190], and conversely, Hedgehog signaling is a master regulator of body fat distribution and glucose metabolism [191, 192]. These findings suggest that interindividual variation in Hedgehog signaling might contribute to variability in both hepatic and extrahepatic outcomes of the metabolic syndrome [125].
Experimental overexpression of Hedgehog ligand in hepatocytes is able to induce a fibrogenic liver response and to promote hepatocarcinogenesis in a transgenic mouse model [193]. At least in part by modulating intermediary metabolism [194, 195], Hedgehog also interacts with pathways which regulate growth and differentiation [194, 196198], such as Wnt/β-catenin signaling, which are of potential significance for the development of hepatocellular carcinoma [199].
Treatment with compounds, e.g., vitamin E, that suppress Hedgehog ligand production and reduce the hepatic accumulation of Hedgehog-responsive myofibroblasts has proven beneficial in human NASH [200].
Further studies are necessary to establish whether and to what extent sex hormones affect Hedgehog signaling and how manipulation of cellular energy homeostasis might be exploited to prevent and manage fibrosing NASH, cirrhosis, and HCC in individuals with NAFLD.

Can Gender Dimorphism of NAFLD be Exploited for Therapeutic Purposes?

In principle, sex differences found in NAFLD may be accounted for by the effects of sex chromosomes; sex hormones; and by differences in dietary and lifestyle habits [201, 202]. A better understanding of the physiopathological peculiarities of NAFLD in women may contribute to developing tailored therapeutic interventions [203]. Accordingly, the roles of estrogen and HRT, the metabolism of choline, and the effect of weight reduction and exercising in women are discussed in detail hereafter.

Estrogens

As detailed in Table 3, estrogens exert several beneficial metabolic effects. Experimental studies suggest that estrogens promote the accumulation of peripheral gluteofemoral subcutaneous adipose tissue and, within the liver, promote FFA beta-oxidation and prevent the accumulation of triglycerides; moreover, they regulate energy homeostasis, enhance insulin sensitivity, and exert a protective role on the function of pancreatic beta-cells [201]. Finally, estrogens seem to have antisteatotic, antioxidant, and antifibrogenic properties in the liver [204, 205]. Estrogens may protect from liver steatosis and fibrosis in female mice via upregulation of miRNA-125b and miRNA-29, respectively [206, 207]. A recent experimental study showed that the estrogen/estrogen receptor alpha signaling plays essential roles in ROS detoxification and in the reduction of oxidative damage in the liver via partnering with hepatic PPARG coactivator 1 alpha, thus halting the transition from simple steatosis to steatohepatitis [205]. Estrogens are also known to inhibit stellate cell activation and fibrogenesis in experimental models [204]. Of note, a study showed that the natural estrogen 17β-estradiol prevents deoxycholic acid-induced hepatocellular damage in several cell lines. This hepatoprotective effect of estrogen was sustained by mechanisms which were unlikely to be mediated by nuclear estrogen receptor alpha [208]. Interestingly, it has recently been demonstrated that a non-nuclear estrogen receptor plays a key role in reducing hepatic steatosis in female mice [209]. Similarly, estrogen deficiency leads to fat redistribution toward visceral fat accumulation and its inherently unfavorable metabolic derangements, and is thus able to induce the development and progression of NAFLD/NASH both in mice and humans. Of note, both aromatase knockout mice, which are unable to synthesize endogenous estrogen, and estrogen receptor alpha knockout mice develop hepatic steatosis [210212]. Patients with breast cancer treated with tamoxifen, a potent estrogen antagonist, develop progressive liver steatosis and NASH [213]. Surgical and physiological menopausal status have been invariably associated with excess risk of NAFLD progression and liver fibrosis, and duration of estrogen deficiency has been directly related to increased fibrosis risk in postmenopausal women with NAFLD [95, 96, 98, 136].

Hormonal Replacement Therapy

Despite the above premises, the role of HRT in reverting the metabolic alterations associated with low levels of estrogens is a matter of dispute. In animal models, both estrogen and raloxifene, a second-generation selective estrogen receptor modulator, have improved inflammation and ballooning so halting the progression of liver fibrosis in diet-induced NASH in female ovariectomized mice [30, 214]. However, the theoretical benefits of HRT in liver disease remain to be proven in humans. A seminal randomized placebo-controlled study suggested that HRT containing low-dose estradiol and norethisterone was able to reduce liver enzyme concentrations in women with T2D, potentially through the reduction of accumulation of fat in the liver [45]. Accordingly, a north American study using data of the NHANES III survey showed that postmenopausal women who received HRT had a significantly lower risk of NAFLD than postmenopausal women who did not (OR 0.69, 95% CI 0.48–0.99) [215]. However, whether HRT reduces the risk of NASH and/or fibrosis among postmenopausal women remains uncertain. Two studies showed no, or borderline, beneficial effects of HRT on fibrosis among postmenopausal women with NAFLD [95, 96]. Worryingly, a recent study reported that synthetic hormone use was associated with more severe hepatocellular injury and lobular inflammation. Further analysis showed that progesterone, but not estrogen use, was associated with worse liver histological lesions, suggesting a multifaceted impact of sex hormones on NASH features [97].

Choline

Several lines of evidence support a key role of choline as an essential nutrient and cellular component. Choline is the major source of methyl groups in the diet and is a precursor of phosphatidylcholine, which is an important component of cell membranes and VLDL, required for the export of lipids from the liver into the bloodstream. Thus, depletion of choline inhibits hepatic triglyceride export and induces fatty liver in both experimental models and humans [185, 203, 216, 217]. Of note, postmenopausal women are more prone than premenopausal women and men to develop NAFLD as a consequence of choline deficiency [128, 185, 217], and decreased dietary choline intake has been significantly associated with increased fibrosis in postmenopausal women with NAFLD [218]. Indeed, menopausal status determines a differential susceptibility to choline deficiency arising from the estrogen-inducible expression and activity of phosphatidylethanolamine-N-methyltransferase (PEMT), a key enzyme in de novo choline biosynthetic pathway. Following menopause, the endogenous supply of choline decreases as a result of estrogen deficiency; consequently, the dietary requirement of choline is increased [185, 203, 216, 217]. Consistently, choline supplementation has been reported to improve or revert liver steatosis in patients receiving long-term total parenteral nutrition [219]. However, no interventional studies have addressed whether choline supplementation is able to prevent NAFLD or reduce NAFLD progression; moreover, future studies investigating the role of preventive or therapeutic choline supplementation in postmenopausal women are eagerly awaited.

Weight Reduction and Exercise

Lifestyle changes, i.e., physical activity and balanced diet, are the milestone of intervention for NAFLD treatment. In general, weight loss induced by lifestyle changes improves IR and features of the MetS, including NAFLD. Of note, a modest weight loss of as little as 2–3 kg is associated with NAFLD reversal [39], and weight loss of greater than 7–10% significantly improves histological features of NASH, including fibrosis [220]. However, significant weight reduction achieved through dietary restrictions results in negative effects on lean muscle and bone mass in postmenopausal women. Therefore, while weight reduction in postmenopausal women with metabolic abnormalities and risk factors for NAFLD should be strongly recommended, special attention should be paid to how weight loss is obtained in older women to prevent the worsening of loss of lean mass [139, 203]. An integrated approach consisting of dietary changes along with regular exercise is mandatory to prevent the untoward effect of losing lean mass. Both resistance training and aerobic exercise are effective in preventing muscle and bone loss during weight loss [203], and equally reduce liver fat content [221, 222]. Postmenopausal women with higher levels of physical activity seem to have lower total body and visceral fat and to be less likely to gain fat mass during menopause [139, 223]. However, only few studies have specifically assessed the role of physical activity in NAFLD prevention/reversal in postmenopausal women. Data have shown that exercise training effectively reduces liver enzymes in overweight postmenopausal women, probably as a reflection of liver fat content [224], and aerobic physical activity reduces cardiovascular risk factors in postmenopausal women with NAFLD [225]. Further studies are needed to define the ideal structure and duration of exercise-based interventions in postmenopausal women with NAFLD or at risk of developing it.
In summary, there are no codified therapeutic interventions approved for tackling NAFLD in women. Given the theoretical beneficial metabolic and hepatic effects of estrogens, it is intriguing to speculate that HRT might possibly play a role in the prevention and treatment of metabolic alterations associated with menopause, including NAFLD. However, studies in humans are lacking and the potential metabolic benefits should be weighed, in clinical practice, against the detrimental cardiovascular and neoplastic risk associated with long-term HRT [226, 227]. Moreover, the multifaceted and variable impact of different sex hormones in NAFLD should be carefully considered. Dietary changes associated with either aerobic or resistance physical exercise should be considered as the milestone of treatment of NAFLD. Future studies will have to evaluate the effects on NAFLD prevention/treatment of supplementing choline to a balanced diet in postmenopausal women. Finally, the physiopathological peculiarities of NAFLD in women and the gender-based differences in the kinetics and dynamics of drugs and xenobiotics should be taken into account when developing new treatment strategies and proposing interventional trials for NAFLD.

Conclusion

Consistent with the notion that NAFLD and NASH are strongly linked with hormonal influences [228], this narrative review article was driven by the hypothesis that what we called the “sexual dimorphism” of NAFLD might be useful in identifying clues of pathogenic significance and providing hints for prevention and treatment of NAFLD. On these grounds, a systematic research of the literature was conducted. What we found was that not only are men at an increased risk of developing NAFLD (Fig. 1) but also significant age-related changes in NAFLD epidemiology in women may potentially bear physiopathological, clinical, and therapeutic significance. NAFLD epidemiology and physiopathology are modulated by age at menarche and postmenopausal status (Fig. 2). It would be expected that early menarche, definitely associated with estrogen activation, would produce protection against the risk of NAFLD. Nevertheless, it has been suggested that early menarche may confer an increased risk of NAFLD in adulthood, excess adiposity being the primary culprit of this association. Fertile age may be associated with more severe hepatocyte injury and inflammation, but also with a decreased risk of liver fibrosis compared to men and postmenopausal status. Ovarian senescence is strongly associated with severe steatosis and fibrosing NASH which may occur in postmenopausal women. Estrogen deficiency is deemed to be responsible for these findings via the development of postmenopausal metabolic syndrome. Estrogen supplementation may at least theoretically protect from NAFLD development and progression, as suggested by some studies exploring the effect of HRT on postmenopausal women, but the variable impact of different sex hormones in NAFLD (i.e., the pro-inflammatory effect of progesterone) should be carefully taken into account when proposing treatment with synthetic hormones.
Taken collectively, these data may generate innovative hypotheses to be tested in appropriate clinical and experimental studies on NAFLD physiopathology and treatment.

Acknowledgements

No funding or sponsorship was received for this study or publication of this article.
All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval for the version to be published.
Stefano Ballestri, Fabio Nascimbeni, and Amedeo Lonardo conceived the idea of this article and wrote its first draft. All the authors have read, edited, and approved the various versions of this article.

Disclosures

Stefano Ballestri and Enrica Baldelli have nothing to disclose. Fabio Nascimbeni is involved as a researcher in the “Phase 3, Double Blind, Randomized, Long-Term, Placebo-controlled, Multicenter study evaluating the Safety and Efficacy of Obethicolic Acid in Subjects with NASH” (EudraCT 2015-002560-16). Alessandra Marrazzo is involved as a researcher in the “Phase 3, Double Blind, Randomized, Long-Term, Placebo-controlled, Multicenter study evaluating the Safety and Efficacy of Obethicolic Acid in Subjects with NASH” (EudraCT 2015-002560-16). Amedeo Lonardo is involved as a researcher in the “Phase 3, Double Blind, Randomized, Long-Term, Placebo-controlled, Multicenter study evaluating the Safety and Efficacy of Obethicolic Acid in Subjects with NASH” (EudraCT 2015-002560-16). Dante Romagnoli serves as a consultant for AbbVie.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not involve any new studies of human or animal subjects performed by any of the authors.

Data Availability

All data generated or analyzed during this study are included in this published article.

Open Access

This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://​creativecommons.​org/​licenses/​by/​4.​0), which permits use, duplication, adaptation, distribution, and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Brunt EM. Nonalcoholic fatty liver disease: pros and cons of histologic systems of evaluation. Int J Mol Sci 2016;17. Brunt EM. Nonalcoholic fatty liver disease: pros and cons of histologic systems of evaluation. Int J Mol Sci 2016;17.
2.
Zurück zum Zitat Ballestri S, Nascimbeni F, Romagnoli D, et al. The role of nuclear receptors in the pathophysiology, natural course, and drug treatment of NAFLD in humans. Adv Ther. 2016;33:291–319.PubMedCrossRef Ballestri S, Nascimbeni F, Romagnoli D, et al. The role of nuclear receptors in the pathophysiology, natural course, and drug treatment of NAFLD in humans. Adv Ther. 2016;33:291–319.PubMedCrossRef
3.
Zurück zum Zitat Piscaglia F, Svegliati-Baroni G, Barchetti A, et al. Clinical patterns of hepatocellular carcinoma in nonalcoholic fatty liver disease: a multicenter prospective study. Hepatology. 2016;63:827–38.PubMedCrossRef Piscaglia F, Svegliati-Baroni G, Barchetti A, et al. Clinical patterns of hepatocellular carcinoma in nonalcoholic fatty liver disease: a multicenter prospective study. Hepatology. 2016;63:827–38.PubMedCrossRef
4.
Zurück zum Zitat Petta S, Valenti L, Bugianesi E, et al. A “systems medicine” approach to the study of non-alcoholic fatty liver disease. Dig Liver Dis. 2016;48:333–42.PubMedCrossRef Petta S, Valenti L, Bugianesi E, et al. A “systems medicine” approach to the study of non-alcoholic fatty liver disease. Dig Liver Dis. 2016;48:333–42.PubMedCrossRef
6.
Zurück zum Zitat Lonardo A, Sookoian S, Chonchol M, et al. Cardiovascular and systemic risk in nonalcoholic fatty liver disease—atherosclerosis as a major player in the natural course of NAFLD. Curr Pharm Des. 2013;19:5177–92.PubMedCrossRef Lonardo A, Sookoian S, Chonchol M, et al. Cardiovascular and systemic risk in nonalcoholic fatty liver disease—atherosclerosis as a major player in the natural course of NAFLD. Curr Pharm Des. 2013;19:5177–92.PubMedCrossRef
7.
Zurück zum Zitat Loria P, Marchesini G, Nascimbeni F, et al. Cardiovascular risk, lipidemic phenotype and steatosis. A comparative analysis of cirrhotic and non-cirrhotic liver disease due to varying etiology. Atherosclerosis. 2014;232:99–109.PubMedCrossRef Loria P, Marchesini G, Nascimbeni F, et al. Cardiovascular risk, lipidemic phenotype and steatosis. A comparative analysis of cirrhotic and non-cirrhotic liver disease due to varying etiology. Atherosclerosis. 2014;232:99–109.PubMedCrossRef
8.
Zurück zum Zitat Lonardo A, Ballestri S, Targher G, et al. Diagnosis and management of cardiovascular risk in nonalcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol. 2015;9:629–50.PubMedCrossRef Lonardo A, Ballestri S, Targher G, et al. Diagnosis and management of cardiovascular risk in nonalcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol. 2015;9:629–50.PubMedCrossRef
9.
Zurück zum Zitat Targher G, Byrne CD, Lonardo A, et al. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: a meta-analysis. J Hepatol. 2016;65:589–600.PubMedCrossRef Targher G, Byrne CD, Lonardo A, et al. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: a meta-analysis. J Hepatol. 2016;65:589–600.PubMedCrossRef
10.
Zurück zum Zitat Nascimbeni F, Pais R, Bellentani S, et al. From NAFLD in clinical practice to answers from guidelines. J Hepatol. 2013;59:859–71.PubMedCrossRef Nascimbeni F, Pais R, Bellentani S, et al. From NAFLD in clinical practice to answers from guidelines. J Hepatol. 2013;59:859–71.PubMedCrossRef
11.
Zurück zum Zitat Lonardo A, Bellentani S, Ratziu V, et al. Insulin resistance in nonalcoholic steatohepatitis: necessary but not sufficient - death of a dogma from analysis of therapeutic studies? Expert Rev Gastroenterol Hepatol. 2011;5:279–89.PubMedCrossRef Lonardo A, Bellentani S, Ratziu V, et al. Insulin resistance in nonalcoholic steatohepatitis: necessary but not sufficient - death of a dogma from analysis of therapeutic studies? Expert Rev Gastroenterol Hepatol. 2011;5:279–89.PubMedCrossRef
12.
Zurück zum Zitat Ballestri S, Nascimbeni F, Romagnoli D, et al. The independent predictors of non-alcoholic steatohepatitis and its individual histological features. Insulin resistance, serum uric acid, metabolic syndrome, alanine aminotransferase and serum total cholesterol are a clue to pathogenesis and candidate targets for treatment. Hepatol Res. 2016;46:1074–87.PubMedCrossRef Ballestri S, Nascimbeni F, Romagnoli D, et al. The independent predictors of non-alcoholic steatohepatitis and its individual histological features. Insulin resistance, serum uric acid, metabolic syndrome, alanine aminotransferase and serum total cholesterol are a clue to pathogenesis and candidate targets for treatment. Hepatol Res. 2016;46:1074–87.PubMedCrossRef
13.
Zurück zum Zitat Ballestri S, Zona S, Targher G, et al. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J Gastroenterol Hepatol. 2016;31:936–44.PubMedCrossRef Ballestri S, Zona S, Targher G, et al. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J Gastroenterol Hepatol. 2016;31:936–44.PubMedCrossRef
14.
Zurück zum Zitat Younossi ZM, Koenig AB, Abdelatif D, et al. Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64:73–84.PubMedCrossRef Younossi ZM, Koenig AB, Abdelatif D, et al. Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64:73–84.PubMedCrossRef
15.
Zurück zum Zitat Non-Alcoholic Fatty Liver Disease Study Group, Lonardo A, Bellentani S, et al. Epidemiological modifiers of non-alcoholic fatty liver disease: focus on high-risk groups. Dig Liver Dis. 2015;47:997–1006.CrossRef Non-Alcoholic Fatty Liver Disease Study Group, Lonardo A, Bellentani S, et al. Epidemiological modifiers of non-alcoholic fatty liver disease: focus on high-risk groups. Dig Liver Dis. 2015;47:997–1006.CrossRef
16.
Zurück zum Zitat Baumeister SE, Volzke H, Marschall P, et al. Impact of fatty liver disease on health care utilization and costs in a general population: a 5-year observation. Gastroenterology. 2008;134:85–94.PubMedCrossRef Baumeister SE, Volzke H, Marschall P, et al. Impact of fatty liver disease on health care utilization and costs in a general population: a 5-year observation. Gastroenterology. 2008;134:85–94.PubMedCrossRef
18.
Zurück zum Zitat Lonardo A, Sookoian S, Pirola CJ, et al. Non-alcoholic fatty liver disease and risk of cardiovascular disease. Metabolism. 2016;65:1136–50.PubMedCrossRef Lonardo A, Sookoian S, Pirola CJ, et al. Non-alcoholic fatty liver disease and risk of cardiovascular disease. Metabolism. 2016;65:1136–50.PubMedCrossRef
19.
Zurück zum Zitat Younossi ZM, Blissett D, Blissett R, et al. The economic and clinical burden of nonalcoholic fatty liver disease in the United States and Europe. Hepatology. 2016;64:1577–86.PubMedCrossRef Younossi ZM, Blissett D, Blissett R, et al. The economic and clinical burden of nonalcoholic fatty liver disease in the United States and Europe. Hepatology. 2016;64:1577–86.PubMedCrossRef
20.
Zurück zum Zitat Day CP. Pathogenesis of steatohepatitis. Best Pract Res Clin Gastroenterol. 2002;16:663–78.PubMedCrossRef Day CP. Pathogenesis of steatohepatitis. Best Pract Res Clin Gastroenterol. 2002;16:663–78.PubMedCrossRef
21.
Zurück zum Zitat Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology. 2010;52:1836–46.PubMedCrossRef Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology. 2010;52:1836–46.PubMedCrossRef
22.
Zurück zum Zitat Day CP, James OF. Steatohepatitis: a tale of two “hits”? Gastroenterology. 1998;114:842–5.PubMedCrossRef Day CP, James OF. Steatohepatitis: a tale of two “hits”? Gastroenterology. 1998;114:842–5.PubMedCrossRef
23.
Zurück zum Zitat Marinou K, Adiels M, Hodson L, et al. Young women partition fatty acids towards ketone body production rather than VLDL-TAG synthesis, compared with young men. Br J Nutr. 2011;105:857–65.PubMedCrossRef Marinou K, Adiels M, Hodson L, et al. Young women partition fatty acids towards ketone body production rather than VLDL-TAG synthesis, compared with young men. Br J Nutr. 2011;105:857–65.PubMedCrossRef
24.
Zurück zum Zitat Lee YH, Kim SH, Kim SN, et al. Sex-specific metabolic interactions between liver and adipose tissue in MCD diet-induced non-alcoholic fatty liver disease. Oncotarget. 2016;7:46959–46971. Lee YH, Kim SH, Kim SN, et al. Sex-specific metabolic interactions between liver and adipose tissue in MCD diet-induced non-alcoholic fatty liver disease. Oncotarget. 2016;7:46959–46971.
25.
Zurück zum Zitat Ludwig J, Viggiano TR, McGill DB, et al. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin Proc. 1980;55:434–8.PubMed Ludwig J, Viggiano TR, McGill DB, et al. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin Proc. 1980;55:434–8.PubMed
26.
Zurück zum Zitat Lonardo A, Carani C, Carulli N, et al. ‘Endocrine NAFLD’ a hormonocentric perspective of nonalcoholic fatty liver disease pathogenesis. J Hepatol. 2006;44:1196–207.PubMedCrossRef Lonardo A, Carani C, Carulli N, et al. ‘Endocrine NAFLD’ a hormonocentric perspective of nonalcoholic fatty liver disease pathogenesis. J Hepatol. 2006;44:1196–207.PubMedCrossRef
28.
Zurück zum Zitat Fadini GP, de Kreutzenberg S, Albiero M, et al. Gender differences in endothelial progenitor cells and cardiovascular risk profile: the role of female estrogens. Arterioscler Thromb Vasc Biol. 2008;28:997–1004.PubMedCrossRef Fadini GP, de Kreutzenberg S, Albiero M, et al. Gender differences in endothelial progenitor cells and cardiovascular risk profile: the role of female estrogens. Arterioscler Thromb Vasc Biol. 2008;28:997–1004.PubMedCrossRef
29.
Zurück zum Zitat Turola E, Petta S, Vanni E, et al. Ovarian senescence increases liver fibrosis in humans and zebrafish with steatosis. Dis Model Mech. 2015;8:1037–46.PubMedPubMedCentralCrossRef Turola E, Petta S, Vanni E, et al. Ovarian senescence increases liver fibrosis in humans and zebrafish with steatosis. Dis Model Mech. 2015;8:1037–46.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Kamada Y, Kiso S, Yoshida Y, et al. Estrogen deficiency worsens steatohepatitis in mice fed high-fat and high-cholesterol diet. Am J Physiol Gastrointest Liver Physiol. 2011;301:G1031–43.PubMedCrossRef Kamada Y, Kiso S, Yoshida Y, et al. Estrogen deficiency worsens steatohepatitis in mice fed high-fat and high-cholesterol diet. Am J Physiol Gastrointest Liver Physiol. 2011;301:G1031–43.PubMedCrossRef
32.
Zurück zum Zitat Targher G, Rossini M, Lonardo A. Evidence that non-alcoholic fatty liver disease and polycystic ovary syndrome are associated by necessity rather than chance: a novel hepato-ovarian axis? Endocrine. 2016;51:211–21.PubMedCrossRef Targher G, Rossini M, Lonardo A. Evidence that non-alcoholic fatty liver disease and polycystic ovary syndrome are associated by necessity rather than chance: a novel hepato-ovarian axis? Endocrine. 2016;51:211–21.PubMedCrossRef
33.
Zurück zum Zitat Kojima S, Watanabe N, Numata M, et al. Increase in the prevalence of fatty liver in Japan over the past 12 years: analysis of clinical background. J Gastroenterol. 2003;38:954–61.PubMedCrossRef Kojima S, Watanabe N, Numata M, et al. Increase in the prevalence of fatty liver in Japan over the past 12 years: analysis of clinical background. J Gastroenterol. 2003;38:954–61.PubMedCrossRef
34.
Zurück zum Zitat Hamaguchi M, Kojima T, Takeda N, et al. The metabolic syndrome as a predictor of nonalcoholic fatty liver disease. Ann Intern Med. 2005;143:722–8.PubMedCrossRef Hamaguchi M, Kojima T, Takeda N, et al. The metabolic syndrome as a predictor of nonalcoholic fatty liver disease. Ann Intern Med. 2005;143:722–8.PubMedCrossRef
35.
Zurück zum Zitat Suzuki A, Angulo P, Lymp J, et al. Chronological development of elevated aminotransferases in a nonalcoholic population. Hepatology. 2005;41:64–71.PubMedCrossRef Suzuki A, Angulo P, Lymp J, et al. Chronological development of elevated aminotransferases in a nonalcoholic population. Hepatology. 2005;41:64–71.PubMedCrossRef
36.
Zurück zum Zitat Tsuneto A, Hida A, Sera N, et al. Fatty liver incidence and predictive variables. Hypertens Res. 2010;33:638–43.PubMedCrossRef Tsuneto A, Hida A, Sera N, et al. Fatty liver incidence and predictive variables. Hypertens Res. 2010;33:638–43.PubMedCrossRef
37.
Zurück zum Zitat Hamaguchi M, Kojima T, Ohbora A, et al. Aging is a risk factor of nonalcoholic fatty liver disease in premenopausal women. World J Gastroenterol. 2012;18:237–43.PubMedPubMedCentralCrossRef Hamaguchi M, Kojima T, Ohbora A, et al. Aging is a risk factor of nonalcoholic fatty liver disease in premenopausal women. World J Gastroenterol. 2012;18:237–43.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Zhou YJ, Li YY, Nie YQ, et al. Natural course of nonalcoholic fatty liver disease in southern China: a prospective cohort study. J Dig Dis. 2012;13:153–60.PubMedCrossRef Zhou YJ, Li YY, Nie YQ, et al. Natural course of nonalcoholic fatty liver disease in southern China: a prospective cohort study. J Dig Dis. 2012;13:153–60.PubMedCrossRef
39.
Zurück zum Zitat Zelber-Sagi S, Lotan R, Shlomai A, et al. Predictors for incidence and remission of NAFLD in the general population during a seven-year prospective follow-up. J Hepatol. 2012;56:1145–51.PubMedCrossRef Zelber-Sagi S, Lotan R, Shlomai A, et al. Predictors for incidence and remission of NAFLD in the general population during a seven-year prospective follow-up. J Hepatol. 2012;56:1145–51.PubMedCrossRef
40.
Zurück zum Zitat Sung KC, Kim BS, Cho YK, et al. Predicting incident fatty liver using simple cardio-metabolic risk factors at baseline. BMC Gastroenterol. 2012;12:84.PubMedPubMedCentralCrossRef Sung KC, Kim BS, Cho YK, et al. Predicting incident fatty liver using simple cardio-metabolic risk factors at baseline. BMC Gastroenterol. 2012;12:84.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Xu C, Yu C, Ma H, et al. Prevalence and risk factors for the development of nonalcoholic fatty liver disease in a nonobese Chinese population: the Zhejiang Zhenhai Study. Am J Gastroenterol. 2013;108:1299–304.PubMedCrossRef Xu C, Yu C, Ma H, et al. Prevalence and risk factors for the development of nonalcoholic fatty liver disease in a nonobese Chinese population: the Zhejiang Zhenhai Study. Am J Gastroenterol. 2013;108:1299–304.PubMedCrossRef
42.
Zurück zum Zitat Wong VW, Wong GL, Yeung DK, et al. Incidence of non-alcoholic fatty liver disease in Hong Kong: a population study with paired proton-magnetic resonance spectroscopy. J Hepatol. 2015;62:182–9.PubMedCrossRef Wong VW, Wong GL, Yeung DK, et al. Incidence of non-alcoholic fatty liver disease in Hong Kong: a population study with paired proton-magnetic resonance spectroscopy. J Hepatol. 2015;62:182–9.PubMedCrossRef
43.
Zurück zum Zitat Yun KE, Nam GE, Lim J, et al. Waist gain is associated with a higher incidence of nonalcoholic fatty liver disease in Korean adults: a cohort study. PLoS One. 2016;11:e0158710.PubMedPubMedCentralCrossRef Yun KE, Nam GE, Lim J, et al. Waist gain is associated with a higher incidence of nonalcoholic fatty liver disease in Korean adults: a cohort study. PLoS One. 2016;11:e0158710.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Bruno S, Maisonneuve P, Castellana P, et al. Incidence and risk factors for non-alcoholic steatohepatitis: prospective study of 5408 women enrolled in Italian tamoxifen chemoprevention trial. BMJ. 2005;330:932.PubMedPubMedCentralCrossRef Bruno S, Maisonneuve P, Castellana P, et al. Incidence and risk factors for non-alcoholic steatohepatitis: prospective study of 5408 women enrolled in Italian tamoxifen chemoprevention trial. BMJ. 2005;330:932.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat McKenzie J, Fisher BM, Jaap AJ, et al. Effects of HRT on liver enzyme levels in women with type 2 diabetes: a randomized placebo-controlled trial. Clin Endocrinol (Oxf). 2006;65:40–4.CrossRef McKenzie J, Fisher BM, Jaap AJ, et al. Effects of HRT on liver enzyme levels in women with type 2 diabetes: a randomized placebo-controlled trial. Clin Endocrinol (Oxf). 2006;65:40–4.CrossRef
46.
Zurück zum Zitat Shen L, Fan JG, Shao Y, et al. Prevalence of nonalcoholic fatty liver among administrative officers in Shanghai: an epidemiological survey. World J Gastroenterol. 2003;9:1106–10.PubMedPubMedCentralCrossRef Shen L, Fan JG, Shao Y, et al. Prevalence of nonalcoholic fatty liver among administrative officers in Shanghai: an epidemiological survey. World J Gastroenterol. 2003;9:1106–10.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Fan JG, Zhu J, Li XJ, et al. Prevalence of and risk factors for fatty liver in a general population of Shanghai, China. J Hepatol. 2005;43:508–14.PubMedCrossRef Fan JG, Zhu J, Li XJ, et al. Prevalence of and risk factors for fatty liver in a general population of Shanghai, China. J Hepatol. 2005;43:508–14.PubMedCrossRef
48.
Zurück zum Zitat Chen CH, Huang MH, Yang JC, et al. Prevalence and risk factors of nonalcoholic fatty liver disease in an adult population of Taiwan: metabolic significance of nonalcoholic fatty liver disease in nonobese adults. J Clin Gastroenterol. 2006;40:745–52.PubMedCrossRef Chen CH, Huang MH, Yang JC, et al. Prevalence and risk factors of nonalcoholic fatty liver disease in an adult population of Taiwan: metabolic significance of nonalcoholic fatty liver disease in nonobese adults. J Clin Gastroenterol. 2006;40:745–52.PubMedCrossRef
49.
Zurück zum Zitat Park SH, Jeon WK, Kim SH, et al. Prevalence and risk factors of non-alcoholic fatty liver disease among Korean adults. J Gastroenterol Hepatol. 2006;21:138–43.PubMedCrossRef Park SH, Jeon WK, Kim SH, et al. Prevalence and risk factors of non-alcoholic fatty liver disease among Korean adults. J Gastroenterol Hepatol. 2006;21:138–43.PubMedCrossRef
50.
Zurück zum Zitat Zelber-Sagi S, Nitzan-Kaluski D, Halpern Z, et al. Prevalence of primary non-alcoholic fatty liver disease in a population-based study and its association with biochemical and anthropometric measures. Liver Int. 2006;26:856–63.PubMedCrossRef Zelber-Sagi S, Nitzan-Kaluski D, Halpern Z, et al. Prevalence of primary non-alcoholic fatty liver disease in a population-based study and its association with biochemical and anthropometric measures. Liver Int. 2006;26:856–63.PubMedCrossRef
51.
Zurück zum Zitat Zhou YJ, Li YY, Nie YQ, et al. Prevalence of fatty liver disease and its risk factors in the population of South China. World J Gastroenterol. 2007;13:6419–24.PubMedPubMedCentralCrossRef Zhou YJ, Li YY, Nie YQ, et al. Prevalence of fatty liver disease and its risk factors in the population of South China. World J Gastroenterol. 2007;13:6419–24.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Li H, Wang YJ, Tan K, et al. Prevalence and risk factors of fatty liver disease in Chengdu, Southwest China. Hepatobiliary Pancreat Dis Int. 2009;8:377–82.PubMed Li H, Wang YJ, Tan K, et al. Prevalence and risk factors of fatty liver disease in Chengdu, Southwest China. Hepatobiliary Pancreat Dis Int. 2009;8:377–82.PubMed
53.
Zurück zum Zitat Caballeria L, Pera G, Auladell MA, et al. Prevalence and factors associated with the presence of nonalcoholic fatty liver disease in an adult population in Spain. Eur J Gastroenterol Hepatol. 2010;22:24–32.PubMedCrossRef Caballeria L, Pera G, Auladell MA, et al. Prevalence and factors associated with the presence of nonalcoholic fatty liver disease in an adult population in Spain. Eur J Gastroenterol Hepatol. 2010;22:24–32.PubMedCrossRef
54.
Zurück zum Zitat Hu X, Huang Y, Bao Z, et al. Prevalence and factors associated with nonalcoholic fatty liver disease in Shanghai work-units. BMC Gastroenterol. 2012;12:123.PubMedPubMedCentralCrossRef Hu X, Huang Y, Bao Z, et al. Prevalence and factors associated with nonalcoholic fatty liver disease in Shanghai work-units. BMC Gastroenterol. 2012;12:123.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Wong VW, Chu WC, Wong GL, et al. Prevalence of non-alcoholic fatty liver disease and advanced fibrosis in Hong Kong Chinese: a population study using proton-magnetic resonance spectroscopy and transient elastography. Gut. 2012;61:409–15.PubMedCrossRef Wong VW, Chu WC, Wong GL, et al. Prevalence of non-alcoholic fatty liver disease and advanced fibrosis in Hong Kong Chinese: a population study using proton-magnetic resonance spectroscopy and transient elastography. Gut. 2012;61:409–15.PubMedCrossRef
56.
Zurück zum Zitat Eguchi Y, Hyogo H, Ono M, et al. Prevalence and associated metabolic factors of nonalcoholic fatty liver disease in the general population from 2009 to 2010 in Japan: a multicenter large retrospective study. J Gastroenterol. 2012;47:586–95.PubMedCrossRef Eguchi Y, Hyogo H, Ono M, et al. Prevalence and associated metabolic factors of nonalcoholic fatty liver disease in the general population from 2009 to 2010 in Japan: a multicenter large retrospective study. J Gastroenterol. 2012;47:586–95.PubMedCrossRef
57.
Zurück zum Zitat Younossi ZM, Stepanova M, Negro F, et al. Nonalcoholic fatty liver disease in lean individuals in the United States. Medicine (Baltimore). 2012;91:319–27.CrossRef Younossi ZM, Stepanova M, Negro F, et al. Nonalcoholic fatty liver disease in lean individuals in the United States. Medicine (Baltimore). 2012;91:319–27.CrossRef
58.
Zurück zum Zitat Lazo M, Hernaez R, Eberhardt MS, et al. Prevalence of nonalcoholic fatty liver disease in the United States: the Third National Health and Nutrition Examination Survey, 1988-1994. Am J Epidemiol. 2013;178:38–45.PubMedPubMedCentralCrossRef Lazo M, Hernaez R, Eberhardt MS, et al. Prevalence of nonalcoholic fatty liver disease in the United States: the Third National Health and Nutrition Examination Survey, 1988-1994. Am J Epidemiol. 2013;178:38–45.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Wang Z, Xu M, Peng J, et al. Prevalence and associated metabolic factors of fatty liver disease in the elderly. Exp Gerontol. 2013;48:705–9.PubMedCrossRef Wang Z, Xu M, Peng J, et al. Prevalence and associated metabolic factors of fatty liver disease in the elderly. Exp Gerontol. 2013;48:705–9.PubMedCrossRef
60.
Zurück zum Zitat Yan J, Xie W, Ou WN, et al. Epidemiological survey and risk factor analysis of fatty liver disease of adult residents, Beijing, China. J Gastroenterol Hepatol. 2013;28:1654–9.PubMed Yan J, Xie W, Ou WN, et al. Epidemiological survey and risk factor analysis of fatty liver disease of adult residents, Beijing, China. J Gastroenterol Hepatol. 2013;28:1654–9.PubMed
61.
Zurück zum Zitat Chiloiro M, Caruso MG, Cisternino AM, et al. Ultrasound evaluation and correlates of fatty liver disease: a population study in a Mediterranean area. Metab Syndr Relat Disord. 2013;11:349–58.PubMedCrossRef Chiloiro M, Caruso MG, Cisternino AM, et al. Ultrasound evaluation and correlates of fatty liver disease: a population study in a Mediterranean area. Metab Syndr Relat Disord. 2013;11:349–58.PubMedCrossRef
62.
Zurück zum Zitat Foster T, Anania FA, Li D, et al. The prevalence and clinical correlates of nonalcoholic fatty liver disease (NAFLD) in African Americans: the multiethnic study of atherosclerosis (MESA). Dig Dis Sci. 2013;58:2392–8.PubMedPubMedCentralCrossRef Foster T, Anania FA, Li D, et al. The prevalence and clinical correlates of nonalcoholic fatty liver disease (NAFLD) in African Americans: the multiethnic study of atherosclerosis (MESA). Dig Dis Sci. 2013;58:2392–8.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Yang W, Huang H, Wang Y, et al. High red blood cell distribution width is closely associated with nonalcoholic fatty liver disease. Eur J Gastroenterol Hepatol. 2014;26:174–8.PubMedCrossRef Yang W, Huang H, Wang Y, et al. High red blood cell distribution width is closely associated with nonalcoholic fatty liver disease. Eur J Gastroenterol Hepatol. 2014;26:174–8.PubMedCrossRef
64.
Zurück zum Zitat Saida T, Fukushima W, Ohfuji S, et al. Effect modification of body mass index and body fat percentage on fatty liver disease in a Japanese population. J Gastroenterol Hepatol. 2014;29:128–36.PubMedCrossRef Saida T, Fukushima W, Ohfuji S, et al. Effect modification of body mass index and body fat percentage on fatty liver disease in a Japanese population. J Gastroenterol Hepatol. 2014;29:128–36.PubMedCrossRef
65.
Zurück zum Zitat Wang Z, Xu M, Hu Z, et al. Sex-specific prevalence of fatty liver disease and associated metabolic factors in Wuhan, south central China. Eur J Gastroenterol Hepatol. 2014;26:1015–21.PubMedCrossRef Wang Z, Xu M, Hu Z, et al. Sex-specific prevalence of fatty liver disease and associated metabolic factors in Wuhan, south central China. Eur J Gastroenterol Hepatol. 2014;26:1015–21.PubMedCrossRef
66.
Zurück zum Zitat Schneider AL, Lazo M, Selvin E, et al. Racial differences in nonalcoholic fatty liver disease in the US population. Obesity (Silver Spring). 2014;22:292–9.CrossRef Schneider AL, Lazo M, Selvin E, et al. Racial differences in nonalcoholic fatty liver disease in the US population. Obesity (Silver Spring). 2014;22:292–9.CrossRef
67.
Zurück zum Zitat Xiao SJ, Fu GJ, Lv YL, et al. Prevalence and risk factors of fatty liver disease in young and middle-aged population: one center study in Southwestern China. J Gastroenterol Hepatol. 2014;29:358–64.PubMedCrossRef Xiao SJ, Fu GJ, Lv YL, et al. Prevalence and risk factors of fatty liver disease in young and middle-aged population: one center study in Southwestern China. J Gastroenterol Hepatol. 2014;29:358–64.PubMedCrossRef
68.
Zurück zum Zitat Martinez-Alvarado Mdel R, Juarez-Rojas JG, Medina-Urrutia AX, et al. Association of fatty liver with cardiovascular risk factors and subclinical atherosclerosis in a Mexican population. Rev Invest Clin. 2014;66:407–14.PubMed Martinez-Alvarado Mdel R, Juarez-Rojas JG, Medina-Urrutia AX, et al. Association of fatty liver with cardiovascular risk factors and subclinical atherosclerosis in a Mexican population. Rev Invest Clin. 2014;66:407–14.PubMed
69.
Zurück zum Zitat Liu J, Lin H, Zhang C, et al. Non-alcoholic fatty liver disease associated with gallstones in females rather than males: a longitudinal cohort study in Chinese urban population. BMC Gastroenterol. 2014;14:213.PubMedPubMedCentralCrossRef Liu J, Lin H, Zhang C, et al. Non-alcoholic fatty liver disease associated with gallstones in females rather than males: a longitudinal cohort study in Chinese urban population. BMC Gastroenterol. 2014;14:213.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Amirkalali B, Poustchi H, Keyvani H, et al. Prevalence of non-alcoholic fatty liver disease and its predictors in North of Iran. Iran J Public Health. 2014;43:1275–83.PubMedPubMedCentral Amirkalali B, Poustchi H, Keyvani H, et al. Prevalence of non-alcoholic fatty liver disease and its predictors in North of Iran. Iran J Public Health. 2014;43:1275–83.PubMedPubMedCentral
71.
Zurück zum Zitat Nishioji K, Sumida Y, Kamaguchi M, et al. Prevalence of and risk factors for non-alcoholic fatty liver disease in a non-obese Japanese population, 2011–2012. J Gastroenterol. 2015;50:95–108.PubMedCrossRef Nishioji K, Sumida Y, Kamaguchi M, et al. Prevalence of and risk factors for non-alcoholic fatty liver disease in a non-obese Japanese population, 2011–2012. J Gastroenterol. 2015;50:95–108.PubMedCrossRef
72.
Zurück zum Zitat Huang X, Xu M, Chen Y, et al. Validation of the fatty liver index for nonalcoholic fatty liver disease in middle-aged and elderly Chinese. Medicine (Baltimore). 2015;94:e1682.CrossRef Huang X, Xu M, Chen Y, et al. Validation of the fatty liver index for nonalcoholic fatty liver disease in middle-aged and elderly Chinese. Medicine (Baltimore). 2015;94:e1682.CrossRef
73.
Zurück zum Zitat Fattahi MR, Niknam R, Safarpour A, et al. The prevalence of metabolic syndrome in non-alcoholic fatty liver disease; a population-based study. Middle East J Dig Dis. 2016;8:131–7.PubMedPubMedCentralCrossRef Fattahi MR, Niknam R, Safarpour A, et al. The prevalence of metabolic syndrome in non-alcoholic fatty liver disease; a population-based study. Middle East J Dig Dis. 2016;8:131–7.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Motamed N, Sohrabi M, Ajdarkosh H, et al. Fatty liver index vs waist circumference for predicting non-alcoholic fatty liver disease. World J Gastroenterol. 2016;22:3023–30.PubMedPubMedCentralCrossRef Motamed N, Sohrabi M, Ajdarkosh H, et al. Fatty liver index vs waist circumference for predicting non-alcoholic fatty liver disease. World J Gastroenterol. 2016;22:3023–30.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Lonardo A, Lombardini S, Scaglioni F, et al. Fatty liver, carotid disease and gallstones: a study of age-related associations. World J Gastroenterol. 2006;12:5826–33.PubMedPubMedCentralCrossRef Lonardo A, Lombardini S, Scaglioni F, et al. Fatty liver, carotid disease and gallstones: a study of age-related associations. World J Gastroenterol. 2006;12:5826–33.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Bertolotti M, Lonardo A, Mussi C, et al. Nonalcoholic fatty liver disease and aging: epidemiology to management. World J Gastroenterol. 2014;20:14185–204.PubMedPubMedCentralCrossRef Bertolotti M, Lonardo A, Mussi C, et al. Nonalcoholic fatty liver disease and aging: epidemiology to management. World J Gastroenterol. 2014;20:14185–204.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Zhu JZ, Dai YN, Wang YM, et al. Prevalence of nonalcoholic fatty liver disease and economy. Dig Dis Sci. 2015;60:3194–202.PubMedCrossRef Zhu JZ, Dai YN, Wang YM, et al. Prevalence of nonalcoholic fatty liver disease and economy. Dig Dis Sci. 2015;60:3194–202.PubMedCrossRef
78.
Zurück zum Zitat Denzer C, Thiere D, Muche R, et al. Gender-specific prevalences of fatty liver in obese children and adolescents: roles of body fat distribution, sex steroids, and insulin resistance. J Clin Endocrinol Metab. 2009;94:3872–81.PubMedCrossRef Denzer C, Thiere D, Muche R, et al. Gender-specific prevalences of fatty liver in obese children and adolescents: roles of body fat distribution, sex steroids, and insulin resistance. J Clin Endocrinol Metab. 2009;94:3872–81.PubMedCrossRef
79.
Zurück zum Zitat Carulli L, Lonardo A, Lombardini S, et al. Gender, fatty liver and GGT. Hepatology. 2006;44:278–9.PubMedCrossRef Carulli L, Lonardo A, Lombardini S, et al. Gender, fatty liver and GGT. Hepatology. 2006;44:278–9.PubMedCrossRef
80.
Zurück zum Zitat Gutierrez-Grobe Y, Ponciano-Rodriguez G, Ramos MH, et al. Prevalence of non alcoholic fatty liver disease in premenopausal, posmenopausal and polycystic ovary syndrome women. The role of estrogens. Ann Hepatol. 2010;9:402–9.PubMed Gutierrez-Grobe Y, Ponciano-Rodriguez G, Ramos MH, et al. Prevalence of non alcoholic fatty liver disease in premenopausal, posmenopausal and polycystic ovary syndrome women. The role of estrogens. Ann Hepatol. 2010;9:402–9.PubMed
81.
Zurück zum Zitat Chung GE, Yim JY, Kim D, et al. The influence of metabolic factors for nonalcoholic fatty liver disease in women. Biomed Res Int. 2015;2015:131528.PubMedPubMedCentral Chung GE, Yim JY, Kim D, et al. The influence of metabolic factors for nonalcoholic fatty liver disease in women. Biomed Res Int. 2015;2015:131528.PubMedPubMedCentral
82.
Zurück zum Zitat Wang Z, Xu M, Hu Z, et al. Prevalence of nonalcoholic fatty liver disease and its metabolic risk factors in women of different ages and body mass index. Menopause. 2015;22:667–73.PubMedCrossRef Wang Z, Xu M, Hu Z, et al. Prevalence of nonalcoholic fatty liver disease and its metabolic risk factors in women of different ages and body mass index. Menopause. 2015;22:667–73.PubMedCrossRef
83.
84.
Zurück zum Zitat Florentino GS, Cotrim HP, Vilar CP, et al. Nonalcoholic fatty liver disease in menopausal women. Arq Gastroenterol. 2013;50:180–5.PubMedCrossRef Florentino GS, Cotrim HP, Vilar CP, et al. Nonalcoholic fatty liver disease in menopausal women. Arq Gastroenterol. 2013;50:180–5.PubMedCrossRef
85.
Zurück zum Zitat Ryu S, Suh BS, Chang Y, et al. Menopausal stages and non-alcoholic fatty liver disease in middle-aged women. Eur J Obstet Gynecol Reprod Biol. 2015;190:65–70.PubMedCrossRef Ryu S, Suh BS, Chang Y, et al. Menopausal stages and non-alcoholic fatty liver disease in middle-aged women. Eur J Obstet Gynecol Reprod Biol. 2015;190:65–70.PubMedCrossRef
86.
Zurück zum Zitat Rodrigues MH, Bruno AS, Nahas-Neto J, et al. Evaluation of clinical and inflammatory markers of nonalcoholic fatty liver disease in postmenopausal women with metabolic syndrome. Metab Syndr Relat Disord. 2014;12:330–8.PubMedCrossRef Rodrigues MH, Bruno AS, Nahas-Neto J, et al. Evaluation of clinical and inflammatory markers of nonalcoholic fatty liver disease in postmenopausal women with metabolic syndrome. Metab Syndr Relat Disord. 2014;12:330–8.PubMedCrossRef
87.
Zurück zum Zitat Loomis AK, Kabadi S, Preiss D, et al. Body mass index and risk of nonalcoholic fatty liver disease: two electronic health record prospective studies. J Clin Endocrinol Metab. 2016;101:945–52.PubMedCrossRef Loomis AK, Kabadi S, Preiss D, et al. Body mass index and risk of nonalcoholic fatty liver disease: two electronic health record prospective studies. J Clin Endocrinol Metab. 2016;101:945–52.PubMedCrossRef
89.
Zurück zum Zitat Argo CK, Northup PG, Al-Osaimi AM, et al. Systematic review of risk factors for fibrosis progression in non-alcoholic steatohepatitis. J Hepatol. 2009;51:371–9.PubMedCrossRef Argo CK, Northup PG, Al-Osaimi AM, et al. Systematic review of risk factors for fibrosis progression in non-alcoholic steatohepatitis. J Hepatol. 2009;51:371–9.PubMedCrossRef
90.
Zurück zum Zitat Singh DK, Sakhuja P, Malhotra V, et al. Independent predictors of steatohepatitis and fibrosis in Asian Indian patients with non-alcoholic steatohepatitis. Dig Dis Sci. 2008;53:1967–76.PubMedCrossRef Singh DK, Sakhuja P, Malhotra V, et al. Independent predictors of steatohepatitis and fibrosis in Asian Indian patients with non-alcoholic steatohepatitis. Dig Dis Sci. 2008;53:1967–76.PubMedCrossRef
91.
Zurück zum Zitat Hossain N, Afendy A, Stepanova M, et al. Independent predictors of fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2009;7:1224-9, 1229 e1-2. Hossain N, Afendy A, Stepanova M, et al. Independent predictors of fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2009;7:1224-9, 1229 e1-2.
92.
Zurück zum Zitat Al-hamoudi W, El-Sabbah M, Ali S, et al. Epidemiological, clinical, and biochemical characteristics of Saudi patients with nonalcoholic fatty liver disease: a hospital-based study. Ann Saudi Med. 2012;32:288–92.PubMedCrossRef Al-hamoudi W, El-Sabbah M, Ali S, et al. Epidemiological, clinical, and biochemical characteristics of Saudi patients with nonalcoholic fatty liver disease: a hospital-based study. Ann Saudi Med. 2012;32:288–92.PubMedCrossRef
94.
Zurück zum Zitat Tapper EB, Krajewski K, Lai M, et al. Simple non-invasive biomarkers of advanced fibrosis in the evaluation of non-alcoholic fatty liver disease. Gastroenterol Rep (Oxf). 2014;2:276–80.CrossRef Tapper EB, Krajewski K, Lai M, et al. Simple non-invasive biomarkers of advanced fibrosis in the evaluation of non-alcoholic fatty liver disease. Gastroenterol Rep (Oxf). 2014;2:276–80.CrossRef
95.
Zurück zum Zitat Yang JD, Abdelmalek MF, Pang H, et al. Gender and menopause impact severity of fibrosis among patients with nonalcoholic steatohepatitis. Hepatology. 2014;59:1406–14.PubMedPubMedCentralCrossRef Yang JD, Abdelmalek MF, Pang H, et al. Gender and menopause impact severity of fibrosis among patients with nonalcoholic steatohepatitis. Hepatology. 2014;59:1406–14.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Klair JS, Yang JD, Abdelmalek MF, et al. A longer duration of estrogen deficiency increases fibrosis risk among postmenopausal women with nonalcoholic fatty liver disease. Hepatology. 2016;64:85–91.PubMedCrossRef Klair JS, Yang JD, Abdelmalek MF, et al. A longer duration of estrogen deficiency increases fibrosis risk among postmenopausal women with nonalcoholic fatty liver disease. Hepatology. 2016;64:85–91.PubMedCrossRef
97.
Zurück zum Zitat Yang JD, Abdelmalek MF, Guy CD, et al. Patient sex, reproductive status, and synthetic hormone use associate with histologic severity of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2017;15:127–131.e2. Yang JD, Abdelmalek MF, Guy CD, et al. Patient sex, reproductive status, and synthetic hormone use associate with histologic severity of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2017;15:127–131.e2.
98.
Zurück zum Zitat Yoneda M, Thomas E, Sumida Y, et al. The influence of menopause on the development of hepatic fibrosis in nonobese women with nonalcoholic fatty liver disease. Hepatology. 2014;60:1792.PubMedCrossRef Yoneda M, Thomas E, Sumida Y, et al. The influence of menopause on the development of hepatic fibrosis in nonobese women with nonalcoholic fatty liver disease. Hepatology. 2014;60:1792.PubMedCrossRef
99.
Zurück zum Zitat Bedogni G, Miglioli L, Masutti F, et al. Incidence and natural course of fatty liver in the general population: the Dionysos study. Hepatology. 2007;46:1387–91.PubMedCrossRef Bedogni G, Miglioli L, Masutti F, et al. Incidence and natural course of fatty liver in the general population: the Dionysos study. Hepatology. 2007;46:1387–91.PubMedCrossRef
100.
Zurück zum Zitat Ong JP, Pitts A, Younossi ZM. Increased overall mortality and liver-related mortality in non-alcoholic fatty liver disease. J Hepatol. 2008;49:608–12.PubMedCrossRef Ong JP, Pitts A, Younossi ZM. Increased overall mortality and liver-related mortality in non-alcoholic fatty liver disease. J Hepatol. 2008;49:608–12.PubMedCrossRef
101.
Zurück zum Zitat Haring R, Wallaschofski H, Nauck M, et al. Ultrasonographic hepatic steatosis increases prediction of mortality risk from elevated serum gamma-glutamyl transpeptidase levels. Hepatology. 2009;50:1403–11.PubMedCrossRef Haring R, Wallaschofski H, Nauck M, et al. Ultrasonographic hepatic steatosis increases prediction of mortality risk from elevated serum gamma-glutamyl transpeptidase levels. Hepatology. 2009;50:1403–11.PubMedCrossRef
102.
Zurück zum Zitat Villa E. Role of estrogen in liver cancer. Womens Health (Lond). 2008;4:41–50.CrossRef Villa E. Role of estrogen in liver cancer. Womens Health (Lond). 2008;4:41–50.CrossRef
103.
Zurück zum Zitat Reddy SK, Steel JL, Chen HW, et al. Outcomes of curative treatment for hepatocellular cancer in nonalcoholic steatohepatitis versus hepatitis C and alcoholic liver disease. Hepatology. 2012;55:1809–19.PubMedCrossRef Reddy SK, Steel JL, Chen HW, et al. Outcomes of curative treatment for hepatocellular cancer in nonalcoholic steatohepatitis versus hepatitis C and alcoholic liver disease. Hepatology. 2012;55:1809–19.PubMedCrossRef
104.
Zurück zum Zitat Yasui K, Hashimoto E, Komorizono Y, et al. Characteristics of patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. Clin Gastroenterol Hepatol. 2011;9:428–33 (quiz e50).PubMedCrossRef Yasui K, Hashimoto E, Komorizono Y, et al. Characteristics of patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. Clin Gastroenterol Hepatol. 2011;9:428–33 (quiz e50).PubMedCrossRef
105.
Zurück zum Zitat Yang D, Hanna DL, Usher J, et al. Impact of sex on the survival of patients with hepatocellular carcinoma: a surveillance, epidemiology, and end results analysis. Cancer. 2014;120:3707–16.PubMedCrossRef Yang D, Hanna DL, Usher J, et al. Impact of sex on the survival of patients with hepatocellular carcinoma: a surveillance, epidemiology, and end results analysis. Cancer. 2014;120:3707–16.PubMedCrossRef
106.
Zurück zum Zitat Ballestri S, Lonardo A, Bonapace S, et al. Risk of cardiovascular, cardiac and arrhythmic complications in patients with non-alcoholic fatty liver disease. World J Gastroenterol. 2014;20:1724–45.PubMedPubMedCentralCrossRef Ballestri S, Lonardo A, Bonapace S, et al. Risk of cardiovascular, cardiac and arrhythmic complications in patients with non-alcoholic fatty liver disease. World J Gastroenterol. 2014;20:1724–45.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Mantovani A, Ballestri S, Lonardo A, et al. cardiovascular disease and myocardial abnormalities in nonalcoholic fatty liver disease. Dig Dis Sci. 2016;61:1246–67.PubMedCrossRef Mantovani A, Ballestri S, Lonardo A, et al. cardiovascular disease and myocardial abnormalities in nonalcoholic fatty liver disease. Dig Dis Sci. 2016;61:1246–67.PubMedCrossRef
108.
Zurück zum Zitat Wells GL. Cardiovascular risk factors: does sex matter? Curr Vasc Pharmacol. 2016;14:452–7.PubMedCrossRef Wells GL. Cardiovascular risk factors: does sex matter? Curr Vasc Pharmacol. 2016;14:452–7.PubMedCrossRef
109.
Zurück zum Zitat Stepanova M, Younossi ZM. Independent association between nonalcoholic fatty liver disease and cardiovascular disease in the US population. Clin Gastroenterol Hepatol. 2012;10:646–50.PubMedCrossRef Stepanova M, Younossi ZM. Independent association between nonalcoholic fatty liver disease and cardiovascular disease in the US population. Clin Gastroenterol Hepatol. 2012;10:646–50.PubMedCrossRef
110.
Zurück zum Zitat Treeprasertsuk S, Leverage S, Adams LA, et al. The Framingham risk score and heart disease in nonalcoholic fatty liver disease. Liver Int. 2012;32:945–50.PubMedPubMedCentralCrossRef Treeprasertsuk S, Leverage S, Adams LA, et al. The Framingham risk score and heart disease in nonalcoholic fatty liver disease. Liver Int. 2012;32:945–50.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Kim D, Choi SY, Park EH, et al. Nonalcoholic fatty liver disease is associated with coronary artery calcification. Hepatology. 2012;56:605–13.PubMedCrossRef Kim D, Choi SY, Park EH, et al. Nonalcoholic fatty liver disease is associated with coronary artery calcification. Hepatology. 2012;56:605–13.PubMedCrossRef
112.
Zurück zum Zitat Mirbagheri SA, Rashidi A, Abdi S, et al. Liver: an alarm for the heart? Liver Int. 2007;27:891–4.PubMedCrossRef Mirbagheri SA, Rashidi A, Abdi S, et al. Liver: an alarm for the heart? Liver Int. 2007;27:891–4.PubMedCrossRef
113.
Zurück zum Zitat Acikel M, Sunay S, Koplay M, et al. Evaluation of ultrasonographic fatty liver and severity of coronary atherosclerosis, and obesity in patients undergoing coronary angiography. Anadolu Kardiyol Derg. 2009;9:273–9.PubMed Acikel M, Sunay S, Koplay M, et al. Evaluation of ultrasonographic fatty liver and severity of coronary atherosclerosis, and obesity in patients undergoing coronary angiography. Anadolu Kardiyol Derg. 2009;9:273–9.PubMed
114.
Zurück zum Zitat Sun L, Lu SZ. Association between non-alcoholic fatty liver disease and coronary artery disease severity. Chin Med J (Engl). 2011;124:867–72. Sun L, Lu SZ. Association between non-alcoholic fatty liver disease and coronary artery disease severity. Chin Med J (Engl). 2011;124:867–72.
115.
Zurück zum Zitat Wong VW, Wong GL, Yip GW, et al. Coronary artery disease and cardiovascular outcomes in patients with non-alcoholic fatty liver disease. Gut. 2011;60:1721–7.PubMedCrossRef Wong VW, Wong GL, Yip GW, et al. Coronary artery disease and cardiovascular outcomes in patients with non-alcoholic fatty liver disease. Gut. 2011;60:1721–7.PubMedCrossRef
116.
Zurück zum Zitat Fraser A, Harris R, Sattar N, et al. Gamma-glutamyltransferase is associated with incident vascular events independently of alcohol intake: analysis of the British Women’s Heart and Health Study and Meta-Analysis. Arterioscler Thromb Vasc Biol. 2007;27:2729–35.PubMedCrossRef Fraser A, Harris R, Sattar N, et al. Gamma-glutamyltransferase is associated with incident vascular events independently of alcohol intake: analysis of the British Women’s Heart and Health Study and Meta-Analysis. Arterioscler Thromb Vasc Biol. 2007;27:2729–35.PubMedCrossRef
117.
Zurück zum Zitat Ioannou GN, Weiss NS, Boyko EJ, et al. Elevated serum alanine aminotransferase activity and calculated risk of coronary heart disease in the United States. Hepatology. 2006;43:1145–51.PubMedCrossRef Ioannou GN, Weiss NS, Boyko EJ, et al. Elevated serum alanine aminotransferase activity and calculated risk of coronary heart disease in the United States. Hepatology. 2006;43:1145–51.PubMedCrossRef
118.
Zurück zum Zitat Schindhelm RK, Dekker JM, Nijpels G, et al. Alanine aminotransferase predicts coronary heart disease events: a 10-year follow-up of the Hoorn Study. Atherosclerosis. 2007;191:391–6.PubMedCrossRef Schindhelm RK, Dekker JM, Nijpels G, et al. Alanine aminotransferase predicts coronary heart disease events: a 10-year follow-up of the Hoorn Study. Atherosclerosis. 2007;191:391–6.PubMedCrossRef
119.
Zurück zum Zitat Feitosa MF, Reiner AP, Wojczynski MK, et al. Sex-influenced association of nonalcoholic fatty liver disease with coronary heart disease. Atherosclerosis. 2013;227:420–4.PubMedPubMedCentralCrossRef Feitosa MF, Reiner AP, Wojczynski MK, et al. Sex-influenced association of nonalcoholic fatty liver disease with coronary heart disease. Atherosclerosis. 2013;227:420–4.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Buday B, Pach PF, Literati-Nagy B, et al. Sex influenced association of directly measured insulin sensitivity and serum transaminase levels: why alanine aminotransferase only predicts cardiovascular risk in men? Cardiovasc Diabetol. 2015;14:55.PubMedPubMedCentralCrossRef Buday B, Pach PF, Literati-Nagy B, et al. Sex influenced association of directly measured insulin sensitivity and serum transaminase levels: why alanine aminotransferase only predicts cardiovascular risk in men? Cardiovasc Diabetol. 2015;14:55.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Kim HJ, Lim CW, Lee JH, et al. Gender-based differences in the relationship between fatty liver disease and atherosclerosis. Cardiovasc J Afr. 2016;27:281–6.PubMedPubMedCentralCrossRef Kim HJ, Lim CW, Lee JH, et al. Gender-based differences in the relationship between fatty liver disease and atherosclerosis. Cardiovasc J Afr. 2016;27:281–6.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Jepsen P, Vilstrup H, Mellemkjaer L, et al. Prognosis of patients with a diagnosis of fatty liver—a registry-based cohort study. Hepatogastroenterology. 2003;50:2101–4.PubMed Jepsen P, Vilstrup H, Mellemkjaer L, et al. Prognosis of patients with a diagnosis of fatty liver—a registry-based cohort study. Hepatogastroenterology. 2003;50:2101–4.PubMed
123.
Zurück zum Zitat Hamaguchi M, Kojima T, Takeda N, et al. Nonalcoholic fatty liver disease is a novel predictor of cardiovascular disease. World J Gastroenterol. 2007;13:1579–84.PubMedPubMedCentralCrossRef Hamaguchi M, Kojima T, Takeda N, et al. Nonalcoholic fatty liver disease is a novel predictor of cardiovascular disease. World J Gastroenterol. 2007;13:1579–84.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Kim MK, Ahn CW, Nam JS, et al. Association between nonalcoholic fatty liver disease and coronary artery calcification in postmenopausal women. Menopause. 2015;22:1323–7.PubMedPubMedCentralCrossRef Kim MK, Ahn CW, Nam JS, et al. Association between nonalcoholic fatty liver disease and coronary artery calcification in postmenopausal women. Menopause. 2015;22:1323–7.PubMedPubMedCentralCrossRef
125.
126.
Zurück zum Zitat Lonardo A, Trande P. Are there any sex differences in fatty liver? A study of glucose metabolism and body fat distribution. J Gastroenterol Hepatol. 2000;15:775–82.PubMedCrossRef Lonardo A, Trande P. Are there any sex differences in fatty liver? A study of glucose metabolism and body fat distribution. J Gastroenterol Hepatol. 2000;15:775–82.PubMedCrossRef
127.
Zurück zum Zitat Suzuki A, Abdelmalek MF, Unalp-Arida A, et al. Regional anthropometric measures and hepatic fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2010;8:1062–9.PubMedPubMedCentralCrossRef Suzuki A, Abdelmalek MF, Unalp-Arida A, et al. Regional anthropometric measures and hepatic fibrosis in patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2010;8:1062–9.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Fischer LM, da Costa KA, Kwock L, et al. Dietary choline requirements of women: effects of estrogen and genetic variation. Am J Clin Nutr. 2010;92:1113–9.PubMedPubMedCentralCrossRef Fischer LM, da Costa KA, Kwock L, et al. Dietary choline requirements of women: effects of estrogen and genetic variation. Am J Clin Nutr. 2010;92:1113–9.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Bruno Ade S, Rodrigues MH, Alvares MC, et al. Non-alcoholic fatty liver disease and its associated risk factors in Brazilian postmenopausal women. Climacteric. 2014;17:465–71.PubMedCrossRef Bruno Ade S, Rodrigues MH, Alvares MC, et al. Non-alcoholic fatty liver disease and its associated risk factors in Brazilian postmenopausal women. Climacteric. 2014;17:465–71.PubMedCrossRef
130.
Zurück zum Zitat Lu J, Zhang J, Du R, et al. Age at menarche is associated with the prevalence of NAFLD later in life. J Diabetes. 2017;9:53–60. Lu J, Zhang J, Du R, et al. Age at menarche is associated with the prevalence of NAFLD later in life. J Diabetes. 2017;9:53–60.
131.
Zurück zum Zitat Mueller NT, Pereira MA, Demerath EW, et al. Earlier menarche is associated with fatty liver and abdominal ectopic fat in midlife, independent of young adult BMI: the CARDIA study. Obesity (Silver Spring). 2015;23:468–74.CrossRef Mueller NT, Pereira MA, Demerath EW, et al. Earlier menarche is associated with fatty liver and abdominal ectopic fat in midlife, independent of young adult BMI: the CARDIA study. Obesity (Silver Spring). 2015;23:468–74.CrossRef
132.
Zurück zum Zitat Laitinen J, Power C, Jarvelin MR. Family social class, maternal body mass index, childhood body mass index, and age at menarche as predictors of adult obesity. Am J Clin Nutr. 2001;74:287–94.PubMed Laitinen J, Power C, Jarvelin MR. Family social class, maternal body mass index, childhood body mass index, and age at menarche as predictors of adult obesity. Am J Clin Nutr. 2001;74:287–94.PubMed
133.
Zurück zum Zitat Ryu S, Chang Y, Choi Y, et al. Age at menarche and non-alcoholic fatty liver disease. J Hepatol. 2015;62:1164–70.PubMedCrossRef Ryu S, Chang Y, Choi Y, et al. Age at menarche and non-alcoholic fatty liver disease. J Hepatol. 2015;62:1164–70.PubMedCrossRef
134.
Zurück zum Zitat Cao X, Zhou J, Yuan H, et al. Duration of reproductive lifespan and age at menarche in relation to metabolic syndrome in postmenopausal Chinese women. J Obstet Gynaecol Res. 2016;42:1581–7.PubMedCrossRef Cao X, Zhou J, Yuan H, et al. Duration of reproductive lifespan and age at menarche in relation to metabolic syndrome in postmenopausal Chinese women. J Obstet Gynaecol Res. 2016;42:1581–7.PubMedCrossRef
135.
Zurück zum Zitat Kavanagh K, Espeland MA, Sutton-Tyrrell K, et al. Liver fat and SHBG affect insulin resistance in midlife women: the Study of Women’s Health Across the Nation (SWAN). Obesity (Silver Spring). 2013;21:1031–8.CrossRef Kavanagh K, Espeland MA, Sutton-Tyrrell K, et al. Liver fat and SHBG affect insulin resistance in midlife women: the Study of Women’s Health Across the Nation (SWAN). Obesity (Silver Spring). 2013;21:1031–8.CrossRef
136.
Zurück zum Zitat Matsuo K, Gualtieri MR, Cahoon SS, et al. Surgical menopause and increased risk of nonalcoholic fatty liver disease in endometrial cancer. Menopause. 2016;23:189–96.PubMedCrossRef Matsuo K, Gualtieri MR, Cahoon SS, et al. Surgical menopause and increased risk of nonalcoholic fatty liver disease in endometrial cancer. Menopause. 2016;23:189–96.PubMedCrossRef
137.
Zurück zum Zitat Codes L, Asselah T, Cazals-Hatem D, et al. Liver fibrosis in women with chronic hepatitis C: evidence for the negative role of the menopause and steatosis and the potential benefit of hormone replacement therapy. Gut. 2007;56:390–5.PubMedCrossRef Codes L, Asselah T, Cazals-Hatem D, et al. Liver fibrosis in women with chronic hepatitis C: evidence for the negative role of the menopause and steatosis and the potential benefit of hormone replacement therapy. Gut. 2007;56:390–5.PubMedCrossRef
138.
Zurück zum Zitat Lu G, Shimizu I, Cui X, et al. Antioxidant and antiapoptotic activities of idoxifene and estradiol in hepatic fibrosis in rats. Life Sci. 2004;74:897–907.PubMedCrossRef Lu G, Shimizu I, Cui X, et al. Antioxidant and antiapoptotic activities of idoxifene and estradiol in hepatic fibrosis in rats. Life Sci. 2004;74:897–907.PubMedCrossRef
139.
Zurück zum Zitat Lavoie JM, Pighon A. NAFLD, estrogens, and physical exercise: the animal model. J Nutr Metab. 2012;2012:914938.PubMedCrossRef Lavoie JM, Pighon A. NAFLD, estrogens, and physical exercise: the animal model. J Nutr Metab. 2012;2012:914938.PubMedCrossRef
141.
Zurück zum Zitat Tian GX, Sun Y, Pang CJ, et al. Oestradiol is a protective factor for non-alcoholic fatty liver disease in healthy men. Obes Rev. 2012;13:381–7.PubMedCrossRef Tian GX, Sun Y, Pang CJ, et al. Oestradiol is a protective factor for non-alcoholic fatty liver disease in healthy men. Obes Rev. 2012;13:381–7.PubMedCrossRef
142.
Zurück zum Zitat Barton M. Cholesterol and atherosclerosis: modulation by oestrogen. Curr Opin Lipidol. 2013;24:214–20.PubMedCrossRef Barton M. Cholesterol and atherosclerosis: modulation by oestrogen. Curr Opin Lipidol. 2013;24:214–20.PubMedCrossRef
143.
Zurück zum Zitat Feingold K, Brinton EA, Grunfeld C. The effect of endocrine disorders on lipids and lipoproteins. In: De Groot LJ, Chrousos G, Dungan K, et al, editors. South Dartmouth: Endotext; 2000. Feingold K, Brinton EA, Grunfeld C. The effect of endocrine disorders on lipids and lipoproteins. In: De Groot LJ, Chrousos G, Dungan K, et al, editors. South Dartmouth: Endotext; 2000.
146.
Zurück zum Zitat Aquila S, De Amicis F. Steroid receptors and their ligands: effects on male gamete functions. Exp Cell Res. 2014;328:303–13.PubMedCrossRef Aquila S, De Amicis F. Steroid receptors and their ligands: effects on male gamete functions. Exp Cell Res. 2014;328:303–13.PubMedCrossRef
147.
Zurück zum Zitat Taraborrelli S. Physiology, production and action of progesterone. Acta Obstet Gynecol Scand. 2015;94(Suppl 161):8–16.PubMedCrossRef Taraborrelli S. Physiology, production and action of progesterone. Acta Obstet Gynecol Scand. 2015;94(Suppl 161):8–16.PubMedCrossRef
148.
Zurück zum Zitat Aldhoon-Hainerova I, Zamrazilova H, Hill M, et al. Insulin sensitivity and its relation to hormones in adolescent boys and girls. Metabolism. 2017;67:90–8.PubMedCrossRef Aldhoon-Hainerova I, Zamrazilova H, Hill M, et al. Insulin sensitivity and its relation to hormones in adolescent boys and girls. Metabolism. 2017;67:90–8.PubMedCrossRef
149.
Zurück zum Zitat Morita A, Ishigaki Y. Gender-difference in diabetes mellitus. Nihon Rinsho. 2015;73:606–10.PubMed Morita A, Ishigaki Y. Gender-difference in diabetes mellitus. Nihon Rinsho. 2015;73:606–10.PubMed
150.
Zurück zum Zitat Corradi PF, Corradi RB, Greene LW. Physiology of the hypothalamic pituitary gonadal axis in the male. Urol Clin N Am. 2016;43:151–62.CrossRef Corradi PF, Corradi RB, Greene LW. Physiology of the hypothalamic pituitary gonadal axis in the male. Urol Clin N Am. 2016;43:151–62.CrossRef
151.
Zurück zum Zitat Davis SR, Wahlin-Jacobsen S. Testosterone in women—the clinical significance. Lancet Diabetes Endocrinol. 2015;3:980–92.PubMedCrossRef Davis SR, Wahlin-Jacobsen S. Testosterone in women—the clinical significance. Lancet Diabetes Endocrinol. 2015;3:980–92.PubMedCrossRef
152.
Zurück zum Zitat Kalantaridou SN, Calis KA. Testosterone therapy in premenopausal women. Semin Reprod Med. 2006;24:106–14.PubMedCrossRef Kalantaridou SN, Calis KA. Testosterone therapy in premenopausal women. Semin Reprod Med. 2006;24:106–14.PubMedCrossRef
153.
Zurück zum Zitat Davis SR, Worsley R, Miller KK, et al. Androgens and female sexual function and dysfunction-findings from the fourth international consultation of sexual medicine. J Sex Med. 2016;13:168–78.PubMedCrossRef Davis SR, Worsley R, Miller KK, et al. Androgens and female sexual function and dysfunction-findings from the fourth international consultation of sexual medicine. J Sex Med. 2016;13:168–78.PubMedCrossRef
154.
Zurück zum Zitat La Colla A, Pronsato L, Milanesi L, et al. 17β-Estradiol and testosterone in sarcopenia: role of satellite cells. Ageing Res Rev. 2015;24:166–77.PubMedCrossRef La Colla A, Pronsato L, Milanesi L, et al. 17β-Estradiol and testosterone in sarcopenia: role of satellite cells. Ageing Res Rev. 2015;24:166–77.PubMedCrossRef
155.
Zurück zum Zitat Grossmann M. Testosterone and glucose metabolism in men: current concepts and controversies. J Endocrinol. 2014;220:R37–55.PubMedCrossRef Grossmann M. Testosterone and glucose metabolism in men: current concepts and controversies. J Endocrinol. 2014;220:R37–55.PubMedCrossRef
156.
Zurück zum Zitat Corona G, Rastrelli G, Monami M, et al. Body weight loss reverts obesity-associated hypogonadotropic hypogonadism: a systematic review and meta-analysis. Eur J Endocrinol. 2013;168:829–43.PubMedCrossRef Corona G, Rastrelli G, Monami M, et al. Body weight loss reverts obesity-associated hypogonadotropic hypogonadism: a systematic review and meta-analysis. Eur J Endocrinol. 2013;168:829–43.PubMedCrossRef
157.
Zurück zum Zitat Goto A, Morita A, Goto M, et al. Associations of sex hormone-binding globulin and testosterone with diabetes among men and women (the Saku Diabetes study): a case control study. Cardiovasc Diabetol. 2012;11:130.PubMedPubMedCentralCrossRef Goto A, Morita A, Goto M, et al. Associations of sex hormone-binding globulin and testosterone with diabetes among men and women (the Saku Diabetes study): a case control study. Cardiovasc Diabetol. 2012;11:130.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Dakin RS, Walker BR, Seckl JR, et al. Estrogens protect male mice from obesity complications and influence glucocorticoid metabolism. Int J Obes (Lond). 2015;39:1539–47. Dakin RS, Walker BR, Seckl JR, et al. Estrogens protect male mice from obesity complications and influence glucocorticoid metabolism. Int J Obes (Lond). 2015;39:1539–47.
159.
Zurück zum Zitat Candia R, Riquelme A, Baudrand R, et al. Overexpression of 11beta-hydroxysteroid dehydrogenase type 1 in visceral adipose tissue and portal hypercortisolism in non-alcoholic fatty liver disease. Liver Int. 2012;32:392–9.PubMed Candia R, Riquelme A, Baudrand R, et al. Overexpression of 11beta-hydroxysteroid dehydrogenase type 1 in visceral adipose tissue and portal hypercortisolism in non-alcoholic fatty liver disease. Liver Int. 2012;32:392–9.PubMed
160.
Zurück zum Zitat Moon SS, Lee YS, Kim JG, et al. Association of 11beta-hydroxysteroid dehydrogenase type 1 gene polymorphisms with serum alanine aminotransferase activity. Diabetes Res Clin Pract. 2013;99:343–50.PubMedCrossRef Moon SS, Lee YS, Kim JG, et al. Association of 11beta-hydroxysteroid dehydrogenase type 1 gene polymorphisms with serum alanine aminotransferase activity. Diabetes Res Clin Pract. 2013;99:343–50.PubMedCrossRef
161.
Zurück zum Zitat Stefan N, Ramsauer M, Jordan P, et al. Inhibition of 11beta-HSD1 with RO5093151 for non-alcoholic fatty liver disease: a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol. 2014;2:406–16.PubMedCrossRef Stefan N, Ramsauer M, Jordan P, et al. Inhibition of 11beta-HSD1 with RO5093151 for non-alcoholic fatty liver disease: a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol. 2014;2:406–16.PubMedCrossRef
162.
Zurück zum Zitat Albiston AL, Smith RE, Krozowski ZS. Sex- and tissue- specific regulation of 11 beta-hydroxysteroid dehydrogenase mRNA. Mol Cell Endocrinol. 1995;109:183–8.PubMedCrossRef Albiston AL, Smith RE, Krozowski ZS. Sex- and tissue- specific regulation of 11 beta-hydroxysteroid dehydrogenase mRNA. Mol Cell Endocrinol. 1995;109:183–8.PubMedCrossRef
163.
Zurück zum Zitat Tchernof A, Belanger C, Morisset AS, et al. Regional differences in adipose tissue metabolism in women: minor effect of obesity and body fat distribution. Diabetes. 2006;55:1353–60.PubMedCrossRef Tchernof A, Belanger C, Morisset AS, et al. Regional differences in adipose tissue metabolism in women: minor effect of obesity and body fat distribution. Diabetes. 2006;55:1353–60.PubMedCrossRef
164.
Zurück zum Zitat Jensen MD, Cardin S, Edgerton D, et al. Splanchnic free fatty acid kinetics. Am J Physiol Endocrinol Metab. 2003;284:E1140–8.PubMedCrossRef Jensen MD, Cardin S, Edgerton D, et al. Splanchnic free fatty acid kinetics. Am J Physiol Endocrinol Metab. 2003;284:E1140–8.PubMedCrossRef
165.
Zurück zum Zitat Hocking SL, Wu LE, Guilhaus M, et al. Intrinsic depot-specific differences in the secretome of adipose tissue, preadipocytes, and adipose tissue-derived microvascular endothelial cells. Diabetes. 2010;59:3008–16.PubMedPubMedCentralCrossRef Hocking SL, Wu LE, Guilhaus M, et al. Intrinsic depot-specific differences in the secretome of adipose tissue, preadipocytes, and adipose tissue-derived microvascular endothelial cells. Diabetes. 2010;59:3008–16.PubMedPubMedCentralCrossRef
167.
Zurück zum Zitat Cheung O, Kapoor A, Puri P, et al. The impact of fat distribution on the severity of nonalcoholic fatty liver disease and metabolic syndrome. Hepatology. 2007;46:1091–100.PubMedCrossRef Cheung O, Kapoor A, Puri P, et al. The impact of fat distribution on the severity of nonalcoholic fatty liver disease and metabolic syndrome. Hepatology. 2007;46:1091–100.PubMedCrossRef
168.
Zurück zum Zitat Garaulet M, Perez-Llamas F, Baraza JC, et al. Body fat distribution in pre-and post-menopausal women: metabolic and anthropometric variables. J Nutr Health Aging. 2002;6:123–6.PubMed Garaulet M, Perez-Llamas F, Baraza JC, et al. Body fat distribution in pre-and post-menopausal women: metabolic and anthropometric variables. J Nutr Health Aging. 2002;6:123–6.PubMed
169.
Zurück zum Zitat Kvist H, Chowdhury B, Grangard U, et al. Total and visceral adipose-tissue volumes derived from measurements with computed tomography in adult men and women: predictive equations. Am J Clin Nutr. 1988;48:1351–61.PubMed Kvist H, Chowdhury B, Grangard U, et al. Total and visceral adipose-tissue volumes derived from measurements with computed tomography in adult men and women: predictive equations. Am J Clin Nutr. 1988;48:1351–61.PubMed
170.
Zurück zum Zitat Italian Association for the Study of the Liver, Lonardo A, Nascimbeni F, et al. AISF position paper on nonalcoholic fatty liver disease (NAFLD): updates and future directions. Dig Liver Dis. 2017;49:471–483. Italian Association for the Study of the Liver, Lonardo A, Nascimbeni F, et al. AISF position paper on nonalcoholic fatty liver disease (NAFLD): updates and future directions. Dig Liver Dis. 2017;49:471–483.
171.
Zurück zum Zitat Paquette A, Wang D, Jankowski M, et al. Effects of ovariectomy on PPAR alpha, SREBP-1c, and SCD-1 gene expression in the rat liver. Menopause. 2008;15:1169–75.PubMedCrossRef Paquette A, Wang D, Jankowski M, et al. Effects of ovariectomy on PPAR alpha, SREBP-1c, and SCD-1 gene expression in the rat liver. Menopause. 2008;15:1169–75.PubMedCrossRef
172.
Zurück zum Zitat Spangenburg EE, Wohlers LM, Valencia AP. Metabolic dysfunction under reduced estrogen levels: looking to exercise for prevention. Exerc Sport Sci Rev. 2012;40:195–203.PubMedCrossRef Spangenburg EE, Wohlers LM, Valencia AP. Metabolic dysfunction under reduced estrogen levels: looking to exercise for prevention. Exerc Sport Sci Rev. 2012;40:195–203.PubMedCrossRef
173.
Zurück zum Zitat Domingos MM, Rodrigues MF, Stotzer US, et al. Resistance training restores the gene expression of molecules related to fat oxidation and lipogenesis in the liver of ovariectomized rats. Eur J Appl Physiol. 2012;112:1437–44.PubMedCrossRef Domingos MM, Rodrigues MF, Stotzer US, et al. Resistance training restores the gene expression of molecules related to fat oxidation and lipogenesis in the liver of ovariectomized rats. Eur J Appl Physiol. 2012;112:1437–44.PubMedCrossRef
174.
Zurück zum Zitat Lundholm L, Zang H, Hirschberg AL, et al. Key lipogenic gene expression can be decreased by estrogen in human adipose tissue. Fertil Steril. 2008;90:44–8.PubMedCrossRef Lundholm L, Zang H, Hirschberg AL, et al. Key lipogenic gene expression can be decreased by estrogen in human adipose tissue. Fertil Steril. 2008;90:44–8.PubMedCrossRef
175.
Zurück zum Zitat Pighon A, Gutkowska J, Jankowski M, et al. Exercise training in ovariectomized rats stimulates estrogenic-like effects on expression of genes involved in lipid accumulation and subclinical inflammation in liver. Metabolism. 2011;60:629–39.PubMedCrossRef Pighon A, Gutkowska J, Jankowski M, et al. Exercise training in ovariectomized rats stimulates estrogenic-like effects on expression of genes involved in lipid accumulation and subclinical inflammation in liver. Metabolism. 2011;60:629–39.PubMedCrossRef
176.
Zurück zum Zitat Li ZZ, Berk M, McIntyre TM, et al. Hepatic lipid partitioning and liver damage in nonalcoholic fatty liver disease: role of stearoyl-CoA desaturase. J Biol Chem. 2009;284:5637–44.PubMedPubMedCentralCrossRef Li ZZ, Berk M, McIntyre TM, et al. Hepatic lipid partitioning and liver damage in nonalcoholic fatty liver disease: role of stearoyl-CoA desaturase. J Biol Chem. 2009;284:5637–44.PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Liu X, Burhans MS, Flowers MT, et al. Hepatic oleate regulates liver stress response partially through PGC-1alpha during high-carbohydrate feeding. J Hepatol. 2016;65:103–12.PubMedPubMedCentralCrossRef Liu X, Burhans MS, Flowers MT, et al. Hepatic oleate regulates liver stress response partially through PGC-1alpha during high-carbohydrate feeding. J Hepatol. 2016;65:103–12.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Marks KA, Kitson AP, Shaw B, et al. Stearoyl-CoA desaturase 1, elongase 6 and their fatty acid products and precursors are altered in ovariectomized rats with 17beta-estradiol and progesterone treatment. Prostaglandins Leukot Essent Fatty Acids. 2013;89:89–96.PubMedCrossRef Marks KA, Kitson AP, Shaw B, et al. Stearoyl-CoA desaturase 1, elongase 6 and their fatty acid products and precursors are altered in ovariectomized rats with 17beta-estradiol and progesterone treatment. Prostaglandins Leukot Essent Fatty Acids. 2013;89:89–96.PubMedCrossRef
179.
Zurück zum Zitat Park HG, Kothapalli KS, Park WJ, et al. Palmitic acid (16:0) competes with omega-6 linoleic and omega-3 a-linolenic acids for FADS2 mediated delta6-desaturation. Biochim Biophys Acta. 2016;1861:91–7.PubMedCrossRef Park HG, Kothapalli KS, Park WJ, et al. Palmitic acid (16:0) competes with omega-6 linoleic and omega-3 a-linolenic acids for FADS2 mediated delta6-desaturation. Biochim Biophys Acta. 2016;1861:91–7.PubMedCrossRef
180.
Zurück zum Zitat Jump DB, Depner CM, Tripathy S, et al. Potential for dietary omega-3 fatty acids to prevent nonalcoholic fatty liver disease and reduce the risk of primary liver cancer. Adv Nutr. 2015;6:694–702.PubMedPubMedCentralCrossRef Jump DB, Depner CM, Tripathy S, et al. Potential for dietary omega-3 fatty acids to prevent nonalcoholic fatty liver disease and reduce the risk of primary liver cancer. Adv Nutr. 2015;6:694–702.PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Childs CE, Romeu-Nadal M, Burdge GC, et al. Gender differences in the n-3 fatty acid content of tissues. Proc Nutr Soc. 2008;67:19–27.PubMedCrossRef Childs CE, Romeu-Nadal M, Burdge GC, et al. Gender differences in the n-3 fatty acid content of tissues. Proc Nutr Soc. 2008;67:19–27.PubMedCrossRef
183.
Zurück zum Zitat Romano KA, Vivas EI, Amador-Noguez D, et al. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio. 2015;6:e02481.PubMedPubMedCentralCrossRef Romano KA, Vivas EI, Amador-Noguez D, et al. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio. 2015;6:e02481.PubMedPubMedCentralCrossRef
184.
Zurück zum Zitat McCarty MF, DiNicolantonio JJ. An increased need for dietary cysteine in support of glutathione synthesis may underlie the increased risk for mortality associated with low protein intake in the elderly. Age (Dordr). 2015;37:96.CrossRef McCarty MF, DiNicolantonio JJ. An increased need for dietary cysteine in support of glutathione synthesis may underlie the increased risk for mortality associated with low protein intake in the elderly. Age (Dordr). 2015;37:96.CrossRef
185.
Zurück zum Zitat Fischer LM, daCosta KA, Kwock L, et al. Sex and menopausal status influence human dietary requirements for the nutrient choline. Am J Clin Nutr. 2007;85:1275–85.PubMedPubMedCentral Fischer LM, daCosta KA, Kwock L, et al. Sex and menopausal status influence human dietary requirements for the nutrient choline. Am J Clin Nutr. 2007;85:1275–85.PubMedPubMedCentral
186.
Zurück zum Zitat Inaba K, Suzuki S, Ihara H, et al. Sexual dimorphism in endotoxin susceptibility after partial hepatectomy in rats. J Hepatol. 2005;42:719–27.PubMedCrossRef Inaba K, Suzuki S, Ihara H, et al. Sexual dimorphism in endotoxin susceptibility after partial hepatectomy in rats. J Hepatol. 2005;42:719–27.PubMedCrossRef
187.
Zurück zum Zitat Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177–85.PubMedCrossRef Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177–85.PubMedCrossRef
188.
Zurück zum Zitat Voysey M, Barker CI, Snape MD, et al. Sex-dependent immune responses to infant vaccination: an individual participant data meta-analysis of antibody and memory B cells. Vaccine. 2016;34:1657–64.PubMedCrossRef Voysey M, Barker CI, Snape MD, et al. Sex-dependent immune responses to infant vaccination: an individual participant data meta-analysis of antibody and memory B cells. Vaccine. 2016;34:1657–64.PubMedCrossRef
189.
Zurück zum Zitat Syn WK, Jung Y, Omenetti A, et al. Hedgehog-mediated epithelial-to-mesenchymal transition and fibrogenic repair in nonalcoholic fatty liver disease. Gastroenterology. 2009;137(1478–1488):e8. Syn WK, Jung Y, Omenetti A, et al. Hedgehog-mediated epithelial-to-mesenchymal transition and fibrogenic repair in nonalcoholic fatty liver disease. Gastroenterology. 2009;137(1478–1488):e8.
190.
Zurück zum Zitat Myers BR, Sever N, Chong YC, et al. Hedgehog pathway modulation by multiple lipid binding sites on the smoothened effector of signal response. Dev Cell. 2013;26:346–57.PubMedPubMedCentralCrossRef Myers BR, Sever N, Chong YC, et al. Hedgehog pathway modulation by multiple lipid binding sites on the smoothened effector of signal response. Dev Cell. 2013;26:346–57.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Pospisilik JA, Schramek D, Schnidar H, et al. Drosophila genome-wide obesity screen reveals hedgehog as a determinant of brown versus white adipose cell fate. Cell. 2010;140:148–60.PubMedCrossRef Pospisilik JA, Schramek D, Schnidar H, et al. Drosophila genome-wide obesity screen reveals hedgehog as a determinant of brown versus white adipose cell fate. Cell. 2010;140:148–60.PubMedCrossRef
192.
Zurück zum Zitat Teperino R, Amann S, Bayer M, et al. Hedgehog partial agonism drives Warburg-like metabolism in muscle and brown fat. Cell. 2012;151:414–26.PubMedCrossRef Teperino R, Amann S, Bayer M, et al. Hedgehog partial agonism drives Warburg-like metabolism in muscle and brown fat. Cell. 2012;151:414–26.PubMedCrossRef
193.
Zurück zum Zitat Chung SI, Moon H, Ju HL, et al. Hepatic expression of Sonic Hedgehog induces liver fibrosis and promotes hepatocarcinogenesis in a transgenic mouse model. J Hepatol. 2016;64:618–27.PubMedCrossRef Chung SI, Moon H, Ju HL, et al. Hepatic expression of Sonic Hedgehog induces liver fibrosis and promotes hepatocarcinogenesis in a transgenic mouse model. J Hepatol. 2016;64:618–27.PubMedCrossRef
194.
Zurück zum Zitat Wehner D, Weidinger G. Signaling networks organizing regenerative growth of the zebrafish fin. Trends Genet. 2015;31:336–43.PubMedCrossRef Wehner D, Weidinger G. Signaling networks organizing regenerative growth of the zebrafish fin. Trends Genet. 2015;31:336–43.PubMedCrossRef
196.
Zurück zum Zitat Borggrefe T, Lauth M, Zwijsen A, et al. The Notch intracellular domain integrates signals from Wnt, Hedgehog, TGFbeta/BMP and hypoxia pathways. Biochim Biophys Acta. 2016;1863:303–13.PubMedCrossRef Borggrefe T, Lauth M, Zwijsen A, et al. The Notch intracellular domain integrates signals from Wnt, Hedgehog, TGFbeta/BMP and hypoxia pathways. Biochim Biophys Acta. 2016;1863:303–13.PubMedCrossRef
197.
Zurück zum Zitat Shi Y, Chen J, Karner CM, et al. Hedgehog signaling activates a positive feedback mechanism involving insulin-like growth factors to induce osteoblast differentiation. Proc Natl Acad Sci USA. 2015;112:4678–83.PubMedPubMedCentralCrossRef Shi Y, Chen J, Karner CM, et al. Hedgehog signaling activates a positive feedback mechanism involving insulin-like growth factors to induce osteoblast differentiation. Proc Natl Acad Sci USA. 2015;112:4678–83.PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat Varelas X. The Hippo pathway effectors TAZ and YAP in development, homeostasis and disease. Development. 2014;141:1614–26.PubMedCrossRef Varelas X. The Hippo pathway effectors TAZ and YAP in development, homeostasis and disease. Development. 2014;141:1614–26.PubMedCrossRef
200.
Zurück zum Zitat Guy CD, Suzuki A, Zdanowicz M, et al. Hedgehog pathway activation parallels histologic severity of injury and fibrosis in human nonalcoholic fatty liver disease. Hepatology. 2012;55:1711–21.PubMedPubMedCentralCrossRef Guy CD, Suzuki A, Zdanowicz M, et al. Hedgehog pathway activation parallels histologic severity of injury and fibrosis in human nonalcoholic fatty liver disease. Hepatology. 2012;55:1711–21.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Varlamov O, Bethea CL, Roberts CT Jr. Sex-specific differences in lipid and glucose metabolism. Front Endocrinol (Lausanne). 2014;5:241. Varlamov O, Bethea CL, Roberts CT Jr. Sex-specific differences in lipid and glucose metabolism. Front Endocrinol (Lausanne). 2014;5:241.
203.
Zurück zum Zitat Suzuki A, Abdelmalek MF. Nonalcoholic fatty liver disease in women. Womens Health (Lond). 2009;5:191–203.CrossRef Suzuki A, Abdelmalek MF. Nonalcoholic fatty liver disease in women. Womens Health (Lond). 2009;5:191–203.CrossRef
204.
Zurück zum Zitat Yasuda M, Shimizu I, Shiba M, et al. Suppressive effects of estradiol on dimethylnitrosamine-induced fibrosis of the liver in rats. Hepatology. 1999;29:719–27.PubMedCrossRef Yasuda M, Shimizu I, Shiba M, et al. Suppressive effects of estradiol on dimethylnitrosamine-induced fibrosis of the liver in rats. Hepatology. 1999;29:719–27.PubMedCrossRef
205.
Zurück zum Zitat Besse-Patin A, Leveille M, Oropeza D, et al. Estrogen signals through peroxisome proliferator-activated receptor-gamma coactivator 1alpha to reduce oxidative damage associated with diet-induced fatty liver disease. Gastroenterology 2017;152:243–256. Besse-Patin A, Leveille M, Oropeza D, et al. Estrogen signals through peroxisome proliferator-activated receptor-gamma coactivator 1alpha to reduce oxidative damage associated with diet-induced fatty liver disease. Gastroenterology 2017;152:243–256.
206.
Zurück zum Zitat Zhang Y, Wu L, Wang Y, et al. Protective role of estrogen-induced miRNA-29 expression in carbon tetrachloride-induced mouse liver injury. J Biol Chem. 2012;287:14851–62.PubMedPubMedCentralCrossRef Zhang Y, Wu L, Wang Y, et al. Protective role of estrogen-induced miRNA-29 expression in carbon tetrachloride-induced mouse liver injury. J Biol Chem. 2012;287:14851–62.PubMedPubMedCentralCrossRef
207.
Zurück zum Zitat Zhang ZC, Liu Y, Xiao LL, et al. Upregulation of miR-125b by estrogen protects against non-alcoholic fatty liver in female mice. J Hepatol. 2015;63:1466–75.PubMedCrossRef Zhang ZC, Liu Y, Xiao LL, et al. Upregulation of miR-125b by estrogen protects against non-alcoholic fatty liver in female mice. J Hepatol. 2015;63:1466–75.PubMedCrossRef
208.
Zurück zum Zitat Ricchi M, Bertolotti M, Anzivino C, et al. 17 Beta-estradiol prevents cytotoxicity from hydrophobic bile acids in HepG2 and WRL-68 cell cultures. J Gastroenterol Hepatol. 2006;21:894–901.PubMedCrossRef Ricchi M, Bertolotti M, Anzivino C, et al. 17 Beta-estradiol prevents cytotoxicity from hydrophobic bile acids in HepG2 and WRL-68 cell cultures. J Gastroenterol Hepatol. 2006;21:894–901.PubMedCrossRef
209.
Zurück zum Zitat Chambliss KL, Barrera J, Umetani M, et al. Nonnuclear estrogen receptor activation improves hepatic steatosis in female mice. Endocrinology. 2016;157:3731–41.PubMedCrossRef Chambliss KL, Barrera J, Umetani M, et al. Nonnuclear estrogen receptor activation improves hepatic steatosis in female mice. Endocrinology. 2016;157:3731–41.PubMedCrossRef
210.
Zurück zum Zitat Nemoto Y, Toda K, Ono M, et al. Altered expression of fatty acid-metabolizing enzymes in aromatase-deficient mice. J Clin Invest. 2000;105:1819–25.PubMedPubMedCentralCrossRef Nemoto Y, Toda K, Ono M, et al. Altered expression of fatty acid-metabolizing enzymes in aromatase-deficient mice. J Clin Invest. 2000;105:1819–25.PubMedPubMedCentralCrossRef
211.
Zurück zum Zitat Heine PA, Taylor JA, Iwamoto GA, et al. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc Natl Acad Sci USA. 2000;97:12729–34.PubMedPubMedCentralCrossRef Heine PA, Taylor JA, Iwamoto GA, et al. Increased adipose tissue in male and female estrogen receptor-alpha knockout mice. Proc Natl Acad Sci USA. 2000;97:12729–34.PubMedPubMedCentralCrossRef
212.
Zurück zum Zitat Lemieux C, Phaneuf D, Labrie F, et al. Estrogen receptor alpha-mediated adiposity-lowering and hypocholesterolemic actions of the selective estrogen receptor modulator acolbifene. Int J Obes (Lond). 2005;29:1236–44.CrossRef Lemieux C, Phaneuf D, Labrie F, et al. Estrogen receptor alpha-mediated adiposity-lowering and hypocholesterolemic actions of the selective estrogen receptor modulator acolbifene. Int J Obes (Lond). 2005;29:1236–44.CrossRef
213.
Zurück zum Zitat Saphner T, Triest-Robertson S, Li H, et al. The association of nonalcoholic steatohepatitis and tamoxifen in patients with breast cancer. Cancer. 2009;115:3189–95.PubMedCrossRef Saphner T, Triest-Robertson S, Li H, et al. The association of nonalcoholic steatohepatitis and tamoxifen in patients with breast cancer. Cancer. 2009;115:3189–95.PubMedCrossRef
214.
Zurück zum Zitat Luo F, Ishigami M, Achiwa K, et al. Raloxifene ameliorates liver fibrosis of nonalcoholic steatohepatitis induced by choline-deficient high-fat diet in ovariectomized mice. Dig Dis Sci. 2015;60:2730–9.PubMedCrossRef Luo F, Ishigami M, Achiwa K, et al. Raloxifene ameliorates liver fibrosis of nonalcoholic steatohepatitis induced by choline-deficient high-fat diet in ovariectomized mice. Dig Dis Sci. 2015;60:2730–9.PubMedCrossRef
215.
Zurück zum Zitat Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver disease. Gastroenterology. 2002;122:1649–57.PubMedCrossRef Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver disease. Gastroenterology. 2002;122:1649–57.PubMedCrossRef
216.
Zurück zum Zitat Vetelainen R, van Vliet A, van Gulik TM. Essential pathogenic and metabolic differences in steatosis induced by choline or methione-choline deficient diets in a rat model. J Gastroenterol Hepatol. 2007;22:1526–33.PubMedCrossRef Vetelainen R, van Vliet A, van Gulik TM. Essential pathogenic and metabolic differences in steatosis induced by choline or methione-choline deficient diets in a rat model. J Gastroenterol Hepatol. 2007;22:1526–33.PubMedCrossRef
217.
Zurück zum Zitat Yu D, Shu XO, Xiang YB, et al. Higher dietary choline intake is associated with lower risk of nonalcoholic fatty liver in normal-weight Chinese women. J Nutr. 2014;144:2034–40.PubMedPubMedCentralCrossRef Yu D, Shu XO, Xiang YB, et al. Higher dietary choline intake is associated with lower risk of nonalcoholic fatty liver in normal-weight Chinese women. J Nutr. 2014;144:2034–40.PubMedPubMedCentralCrossRef
218.
Zurück zum Zitat Guerrerio AL, Colvin RM, Schwartz AK, et al. Choline intake in a large cohort of patients with nonalcoholic fatty liver disease. Am J Clin Nutr. 2012;95:892–900.PubMedPubMedCentralCrossRef Guerrerio AL, Colvin RM, Schwartz AK, et al. Choline intake in a large cohort of patients with nonalcoholic fatty liver disease. Am J Clin Nutr. 2012;95:892–900.PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Buchman AL, Dubin MD, Moukarzel AA, et al. Choline deficiency: a cause of hepatic steatosis during parenteral nutrition that can be reversed with intravenous choline supplementation. Hepatology. 1995;22:1399–403.PubMed Buchman AL, Dubin MD, Moukarzel AA, et al. Choline deficiency: a cause of hepatic steatosis during parenteral nutrition that can be reversed with intravenous choline supplementation. Hepatology. 1995;22:1399–403.PubMed
220.
Zurück zum Zitat Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology. 2015;149(367–78):e5 (quiz e14-5). Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology. 2015;149(367–78):e5 (quiz e14-5).
221.
Zurück zum Zitat Orci LA, Gariani K, Oldani G, et al. Exercise-based interventions for nonalcoholic fatty liver disease: a meta-analysis and meta-regression. Clin Gastroenterol Hepatol. 2016;14:1398–411.PubMedCrossRef Orci LA, Gariani K, Oldani G, et al. Exercise-based interventions for nonalcoholic fatty liver disease: a meta-analysis and meta-regression. Clin Gastroenterol Hepatol. 2016;14:1398–411.PubMedCrossRef
222.
Zurück zum Zitat Hashida R, Kawaguchi T, Bekki M, et al. Aerobic vs. resistance exercise in non-alcoholic fatty liver disease: A systematic review. J Hepatol. 2017;66:142–152. Hashida R, Kawaguchi T, Bekki M, et al. Aerobic vs. resistance exercise in non-alcoholic fatty liver disease: A systematic review. J Hepatol. 2017;66:142–152.
223.
Zurück zum Zitat Astrup A. Physical activity and weight gain and fat distribution changes with menopause: current evidence and research issues. Med Sci Sports Exerc. 1999;31:S564–7.PubMedCrossRef Astrup A. Physical activity and weight gain and fat distribution changes with menopause: current evidence and research issues. Med Sci Sports Exerc. 1999;31:S564–7.PubMedCrossRef
224.
Zurück zum Zitat Barsalani R, Riesco E, Lavoie JM, et al. Effect of exercise training and isoflavones on hepatic steatosis in overweight postmenopausal women. Climacteric. 2013;16:88–95.PubMedCrossRef Barsalani R, Riesco E, Lavoie JM, et al. Effect of exercise training and isoflavones on hepatic steatosis in overweight postmenopausal women. Climacteric. 2013;16:88–95.PubMedCrossRef
225.
Zurück zum Zitat Rezende RE, Duarte SM, Stefano JT, et al. Randomized clinical trial: benefits of aerobic physical activity for 24 weeks in postmenopausal women with nonalcoholic fatty liver disease. Menopause. 2016;23:876–83.PubMedCrossRef Rezende RE, Duarte SM, Stefano JT, et al. Randomized clinical trial: benefits of aerobic physical activity for 24 weeks in postmenopausal women with nonalcoholic fatty liver disease. Menopause. 2016;23:876–83.PubMedCrossRef
226.
Zurück zum Zitat Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321–33.PubMedCrossRef Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288:321–33.PubMedCrossRef
227.
Zurück zum Zitat Beral V, Million Women Study Collaborators. Breast cancer and hormone-replacement therapy in the Million Women Study. Lancet. 2003;362:419–27.PubMedCrossRef Beral V, Million Women Study Collaborators. Breast cancer and hormone-replacement therapy in the Million Women Study. Lancet. 2003;362:419–27.PubMedCrossRef
228.
Zurück zum Zitat Loria P, Carulli L, Bertolotti M, et al. Endocrine and liver interaction: the role of endocrine pathways in NASH. Nat Rev Gastroenterol Hepatol. 2009;6:236–47.PubMedCrossRef Loria P, Carulli L, Bertolotti M, et al. Endocrine and liver interaction: the role of endocrine pathways in NASH. Nat Rev Gastroenterol Hepatol. 2009;6:236–47.PubMedCrossRef
Metadaten
Titel
NAFLD as a Sexual Dimorphic Disease: Role of Gender and Reproductive Status in the Development and Progression of Nonalcoholic Fatty Liver Disease and Inherent Cardiovascular Risk
verfasst von
Stefano Ballestri
Fabio Nascimbeni
Enrica Baldelli
Alessandra Marrazzo
Dante Romagnoli
Amedeo Lonardo
Publikationsdatum
19.05.2017
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 6/2017
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-017-0556-1

Weitere Artikel der Ausgabe 6/2017

Advances in Therapy 6/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.