+ All Categories
Home > Documents > NAFLD as a Sexual Dimorphic Disease: Role of Gender and ......Alessandra Marrazzo. Dante Romagnoli....

NAFLD as a Sexual Dimorphic Disease: Role of Gender and ......Alessandra Marrazzo. Dante Romagnoli....

Date post: 26-Jan-2021
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
36
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 Stefano Ballestri . Fabio Nascimbeni . Enrica Baldelli . Alessandra Marrazzo . Dante Romagnoli . Amedeo Lonardo Received: November 28, 2016 / Published online: May 19, 2017 Ó The Author(s) 2017. This article is an open access publication 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 mortal- ity. 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 pro- gression of liver disease in an experimental overfed zebrafish model. Consistently, estrogen deficiency, by potentiating hepatic inflamma- tory changes, hastens the progression of disease in a dietary model of nonalcoholic steatohep- atitis (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, def- initely 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, ovar- ian 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 meta- bolic syndrome. Estrogen supplementation may at least theoretically protect from NAFLD development and progression, as suggested by 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. S. Ballestri Azienda USL di Modena, Pavullo Hospital, Pavullo nel Frignano, Italy F. Nascimbeni Á D. Romagnoli Á A. Lonardo Ospedale Civile di Baggiovara, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy F. Nascimbeni (&) Á E. Baldelli Á A. Marrazzo Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy e-mail: [email protected] Adv Ther (2017) 34:1291–1326 DOI 10.1007/s12325-017-0556-1
Transcript
  • REVIEW

    NAFLD as a Sexual Dimorphic Disease: Role of Genderand Reproductive Status in the Developmentand Progression of Nonalcoholic Fatty Liver Diseaseand Inherent Cardiovascular Risk

    Stefano Ballestri . Fabio Nascimbeni . Enrica Baldelli .

    Alessandra Marrazzo . Dante Romagnoli . Amedeo Lonardo

    Received: November 28, 2016 / Published online: May 19, 2017� The Author(s) 2017. This article is an open access publication

    ABSTRACT

    Nonalcoholic fatty liver disease (NAFLD) spanssteatosis through nonalcoholic steatohepatis,cirrhosis, and hepatocellular carcinoma (HCC)associated with striking systemic features andexcess cardiovascular and liver-related mortal-ity. The pathogenesis of NAFLD is complex andmultifactorial. Endocrine derangements areclosely linked with dysmetabolic traits. Forexample, in animal and human studies, femalesex is protected from dysmetabolism thanks toyoung individuals’ ability to partition fattyacids towards ketone body production ratherthan very low density lipoprotein

    (VLDL)-triacylglycerol, and to sex-specificbrowning of white adipose tissue. Ovariansenescence facilitates both the development ofmassive hepatic steatosis and the fibrotic pro-gression of liver disease in an experimentaloverfed zebrafish model. Consistently, estrogendeficiency, by potentiating hepatic inflamma-tory changes, hastens the progression of diseasein a dietary model of nonalcoholic steatohep-atitis (NASH) developing in ovariectomizedmice fed a high-fat diet. In humans, NAFLDmore often affects men; and premenopausalwomen are equally protected from developingNAFLD as they are from cardiovascular disease.It would be expected that early menarche, def-initely associated with estrogen activation,would produce protection against the risk ofNAFLD. Nevertheless, it has been suggested thatearly menarche may confer an increased risk ofNAFLD in adulthood, excess adiposity being theprimary culprit of this association. Fertile agemay be associated with more severe hepatocyteinjury and inflammation, but also with adecreased risk of liver fibrosis compared to menand postmenopausal status. Later in life, ovar-ian senescence is strongly associated with severesteatosis and fibrosing NASH, which may occurin postmenopausal women. Estrogen deficiencyis deemed to be responsible for these findingsvia the development of postmenopausal meta-bolic syndrome. Estrogen supplementation mayat least theoretically protect from NAFLDdevelopment and progression, as suggested by

    Enhanced content To view enhanced content for thisarticle go to http://www.medengine.com/Redeem/2128F060377C0C79.

    Stefano Ballestri and Fabio Nascimbeni contributedequally and are joint first authors.

    S. BallestriAzienda USL di Modena, Pavullo Hospital, Pavullonel Frignano, Italy

    F. Nascimbeni � D. Romagnoli � A. LonardoOspedale Civile di Baggiovara, AziendaOspedaliero-Universitaria di Modena, Modena, Italy

    F. Nascimbeni (&) � E. Baldelli � A. MarrazzoDepartment of Biomedical, Metabolic and NeuralSciences, University of Modena and Reggio Emilia,Modena, Italye-mail: [email protected]

    Adv Ther (2017) 34:1291–1326

    DOI 10.1007/s12325-017-0556-1

    http://www.medengine.com/Redeem/2128F060377C0C79http://www.medengine.com/Redeem/2128F060377C0C79http://crossmark.crossref.org/dialog/?doi=10.1007/s12325-017-0556-1&domain=pdfhttp://crossmark.crossref.org/dialog/?doi=10.1007/s12325-017-0556-1&domain=pdf

  • some studies exploring the effect of hormonalreplacement therapy on postmenopausalwomen, but the variable impact of different sexhormones in NAFLD (i.e., the pro-inflammatoryeffect of progesterone) should be carefullyconsidered.

    Keywords: Fibrosis; Hormones; Inflammation;Man; Menarche; Menopausal status; NASH;Physiopathology; Sex; Steatosis; Women

    INTRODUCTION

    Nonalcoholic fatty liver disease (NAFLD)encompasses the whole spectrum of (predomi-nantly) steatogenic liver disorders spanningsteatosis through nonalcoholic steatohepatitis(NASH), cirrhosis, and hepatocellular carci-noma (HCC) [1–3], associated with strikingsystemic features [4, 5] and excess cardiovascu-lar and liver-related mortality [6–9]. Histologi-cally indistinguishable from alcoholic liverdisease, and nevertheless observed in the non-alcoholic individual [10], NAFLD is closelylinked with insulin resistance (IR) [11] and,bidirectionally, with the metabolic syndrome(MetS) of which it may be both a cause and aconsequence [12, 13]. A leading cause ofchronic liver disease worldwide and affectingone out of four individuals in the European andnorth-American general populations [14],NAFLD is highly prevalent in certain groups ofindividuals carrying either the full-blown orindividual traits of the MetS [15]. Moreover,NAFLD carries an excess of health-relatedexpenditures owing to its close connectionswith progressive liver disease and car-dio-metabolic morbi-mortality [9, 16–19].

    The pathogenesis of NAFLD has variablybeen conceptualized as two-hit [20], a one-hit[11], or a multistep process [21], and the last ispresently the most widely accepted pathogenicmodel. In its original definition, a first hit leadsto steatogenesis and a second one to fibrosis[22]. However, metabolic factors, and particu-larly IR, invariably account for most of the ele-mentary 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 sup-ports the notion that female sex is protectedfrom dysmetabolic traits thanks to young indi-viduals’ ability to partition fatty acids towardsketone body production rather than very lowdensity lipoprotein (VLDL)-triacylglycerol [23],and to sex-specific browning of white adiposetissue which contributes in protecting femalemice from experimental NAFLD associated withmethionine choline deficient diet [24].

    In 1980, Ludwig reported that NASH wascommon among elderly women with metaboliccomorbidity [25]. However, we now know thatNAFLD more often affects men [15, 26] and thatpremenopausal women are equally protectedfrom NAFLD development as they are fromcardiovascular disease (CVD) [27, 28]. Recentstudies in the overnourished zebrafish modelhave shown that ovarian senescence, viahypoestrogenemia, facilitates both the devel-opment of massive hepatic steatosis and thefibrotic progression of liver disease [29]. Con-sistently, estrogen deficiency, by potentiatinghepatic inflammatory changes, hastens theprogression of disease in a dietary model ofNASH developing in ovariectomized mice fed acholesterol-rich hyperlipidic diet [30]. Collec-tively, these findings suggest that hormonalchanges, rather than those multiple physiologicderangements associated with aging per se [31],are a major determinant of progressive NAFLDin human menopause.

    Obesity and obesity-related diseases, such astype 2 diabetes (T2D), MetS, and atherosclerosis,are complex conditions driven by genetic andenvironmental factors, in which a sexualdimorphism has been clearly established. Here,we have reviewed current evidence suggestingthat NAFLD is a sexually dimorphic condition,too. We hypothesized that the higher incidenceof disease in men and the worse outcome inpostmenopausal women, i.e., the ‘‘sexualdimorphism’’ of NAFLD, might offer clues use-ful in expanding our understanding of thepathogenesis and providing hints for preven-tion and treatment of NAFLD. Given thepotential research and clinical implications ofsexual dimorphism in NAFLD, we carried out a

    1292 Adv Ther (2017) 34:1291–1326

  • systematic review of the literature by using thefollowing keywords: steatosis OR fatty liverAND gender OR sex OR menopause. On thesegrounds, this narrative review is aimed tohighlight how sex modulates the developmentand progression of NAFLD and to pinpoint whatthe role of menarche and menopause is. Specialemphasis is conferred to CVD risk. The rela-tionship 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 conductedstudies and does not involve any new studies ofhuman or animal subjects performed by any ofthe authors.

    EPIDEMIOLOGY OF NAFLD

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

    Risk of NAFLD

    IncidenceEvidence from longitudinal studies suggeststhat the incidence of NAFLD is higher in themale as compared to the female gender(Table 1) [33–43]. One study investigating theincidence of NAFLD in women as a function ofreproductive status found incidence to behigher in menopausal (7.5%)/postmenopausal(6.1%) women as compared to premenopausal(3.5%) women. However, postmenopausal sta-tus was associated with an increased risk ofincident NAFLD at univariate but not at multi-variate analysis adjusted for age, metabolicsyndrome, and weight gain [37]. The incidenceof NAFLD in women taking hormonal replace-ment therapy (HRT) (5.3%) was higher than inpremenopausal (3.5%) women but lower thanin menopausal women (7.5%). HRT was notassociated with increased risk of incidentNAFLD either at univariate or at multivariateanalysis [37]. Indeed other longitudinal studies

    suggest that estrogens are a protective factor forthe development of NAFLD. An Italian multi-centric study on 5408 healthy women who hadhad hysterectomies, randomly assigned toreceive tamoxifen (an estrogen inhibitor) orplacebo for 5 years, showed that tamoxifen wasassociated with higher risk of development ofNAFLD/NASH especially in overweight/obesewomen [44]. Finally a small double-blind, ran-domized placebo-controlled trial on 50 womenwith T2D showed that HRT containing low-doseestradiol and norethisterone significantlyreduced serum levels of liver function enzymes,potentially owing to a reduced liver fat accu-mulation [45].

    Overall, data from epidemiological longitu-dinal studies have shown a key role of weightgain, presence of MetS and its single traits asindependent predictors of the development ofNAFLD [33–43]. Notably, some studies havefound that male sex was associated with inci-dent NAFLD independently of age and meta-bolic factors [33, 35, 41]. Moreover, two Asianstudies have reported a specific role of ageaccording to gender and reproductive status inmodulating the risk of incident NAFLD. Onestudy showed that age was an independentpredictor for developing NAFLD only in females[34] and the other one reported that ageincreased the risk for incident NAFLD in pre-menopausal but not in postmenopausal women[37].

    PrevalenceDespite preliminary studies reporting a higherrisk of NAFLD in females, a large body of evi-dence now definitely supports the notion thatthe prevalence of NAFLD is higher in men thanin women and that gender-specific differencesexist in relation to age (Table 1)[33, 37, 41, 46–77]. Conflicting with the abovenotion, two large sample community Iranianstudies reported a higher prevalence of NAFLDin women than in men [70, 74]. However, thesefindings might, at least in part, be accounted forby women having markedly higher rates ofprevalence of obesity compared to men in thesestudies. Consistent with the view reportedabove, a higher prevalence of fatty liver has

    Adv Ther (2017) 34:1291–1326 1293

  • Table1

    Genderim

    pactson

    incident

    andprevalentNAFL

    D.E

    videncefrom

    longitudinalandcross-sectionalstudies

    Stud

    yMetho

    dFind

    ings

    Con

    clusion

    Stud

    ypo

    pulation

    Diagnosis

    ofFL

    Incidence(lon

    gitudinal

    stud

    ies)

    Independ

    ent

    predictors

    Kojim

    a

    etal.[33]

    35,519

    Japanese

    subjects(health

    check-up),65.2%M,m

    eanage

    45.8years,F-up

    12years

    US

    14.3%

    Msex(O

    R1.7),

    metabolicfactors

    (BMIOR6.3)

    Msexandmetabolicfactorsare

    independ

    entrisk

    factorsforthe

    incidenceof

    NAFL

    D

    Ham

    aguchi

    etal.[34]

    3147

    healthyJapanese

    (1694M),aged

    21–8

    0years,

    meanF-up

    414days

    US

    10%;in

    M(14%

    )[F(5%)

    Age

    (onlyin

    F),w

    eight

    gain,and

    MetS

    NAFL

    Dismoreincident

    inmen

    than

    inwom

    en

    Suzuki

    etal.

    [35]

    1537

    Japanese

    subjects

    (1352M),meanage35

    years,

    meanF-up

    60months

    AST

    ,ALT

    31/1000person-years

    Msexoverallandin

    subjects\40

    years

    (HR4.6);metabolic

    factors

    Msexandmetabolicfactorsare

    independ

    entrisk

    factorsforthe

    incidenceof

    NAFL

    D

    Tsuneto

    etal.[36]

    1635

    Nagasakiatom

    icbomb

    survivors(606

    M),meanage

    63years,F-up

    11.6years

    US

    19.9/1000person-years(22.3in

    M[

    18.6in

    F);M

    (38%

    )[F

    (25%

    )before

    50years

    Obesity,h

    ypertension,

    andhigh-TG

    (gender

    NS)

    Genderisnotan

    independ

    entrisk

    factor

    forincident

    NAFL

    D

    Ham

    aguchi

    etal.[37]

    1603

    Japanese

    wom

    en,aged

    21–8

    0years,meanF-up

    414days

    US

    5%in

    F;in

    postmenopausal

    (6.1–7

    .5%)andun

    derHRT

    (5.3%)[

    prem

    enopausal

    (3.5%)

    Age

    (onlyin

    prem

    enopausal),

    weightgain,and

    MetS

    Thereisagradient

    intheincidenceof

    NAFL

    Din

    wom

    en:

    postmenopausal[

    HRT[

    prem

    enopausal

    Zhouet

    al.

    [38]

    507Chinese

    NAFL

    D-free

    participants,m

    edianF-up

    4years

    US

    9.1%

    ;in

    M7.3%

    [F9.7%

    Age,m

    etabolicfactors

    (genderNS)

    Genderisnotan

    independ

    entrisk

    factor

    forincident

    NAFL

    D

    Zelber-Sagi

    etal.[39]

    147Israelisubjects,m

    eanage

    51.2years,F-up

    7years

    US

    19.0%

    (2.7%/year)

    Weightgain

    and

    HOMA

    (genderNS)

    Genderisnotan

    independ

    entrisk

    factor

    forincident

    NAFL

    D

    Sung

    etal.

    [40]

    2589

    Koreansubjects,m

    ean

    F-up

    4.37

    years

    US

    34.7/1000person-years;in

    M

    (23.4%

    )[F(9.7%)

    MetStraits(genderNS)

    Genderwas

    notan

    independ

    entrisk

    factor

    forincident

    NAFL

    D

    Xuet

    al.

    [41]

    5562

    non-obeseChinese

    subjects,m

    eanage43

    years,

    F-up

    5years

    US

    8.9%

    Msex,youn

    gerage,and

    metabolicfactors

    Msexisarisk

    factor

    fortheincidence

    ofNAFL

    Dat

    multivariateanalysis

    1294 Adv Ther (2017) 34:1291–1326

  • Table1

    continued

    Stud

    yMetho

    dFind

    ings

    Con

    clusion

    Stud

    ypo

    pulation

    Diagnosis

    ofFL

    Incidence(lon

    gitudinal

    stud

    ies)

    Independ

    ent

    predictors

    Wonget

    al.

    [42]

    565HongKongsubjects,m

    ean

    age48

    years,medianF-up

    47months

    1 HNMR

    3.4%

    /year

    MetS(M

    sexpredictor

    onlyat

    univariate

    analysis)

    Msexisarisk

    factor

    fortheincidence

    ofNAFL

    Dat

    univariate

    analysis

    Yun

    etal.

    [43]

    37,130

    Koreansubjects,m

    ean

    age(M

    39.4,F

    38.6years),

    46%

    M,F

    -up2years

    US

    M(44.5/1000

    person-years)[

    F

    (20.4/1000

    person-years)

    WC

    gain

    inboth

    sexes

    Visceralobesitystronglypredicts

    NAFL

    Din

    either

    gend

    er

    Stud

    ypo

    pulation

    Diagnosisof

    FLPrevalence

    (cross-section

    alstud

    ies)

    Independ

    ent

    predictors

    Kojim

    aet

    al.[33]

    39,151

    Japanese

    subjects(health

    check-up),meanage45.8years,

    61%

    M

    US

    M(26%

    )[F(12.7%

    )NA

    NAFL

    Dismore

    prevalentin

    M

    gend

    er

    Shen

    etal.[46]

    4009

    Chinese

    administrative

    officersnon-drinkers,aged

    20–8

    1years,64%

    M

    US

    M(13.3%

    )[F

    (2.7%)before

    50years;similar

    after50

    years

    Msex,age[

    50years,

    andmetabolictraits

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforthe

    prevalence

    of

    NAFL

    D

    Fanet

    al.[47]

    3175

    Chinese

    subjectsfrom

    Shanghai,m

    eanage52years,

    38.4%

    M

    US

    M(19.2%

    )[F

    (11.3%

    )before

    50years;F[

    M

    after50

    years

    Msex(O

    R2.7)

    and

    metabolicfactors

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforthe

    prevalence

    of

    NAFL

    D

    Adv Ther (2017) 34:1291–1326 1295

  • Table1

    continued

    Stud

    ypo

    pulation

    Diagnosisof

    FLPrevalence

    (cross-section

    alstud

    ies)

    Independ

    ent

    predictors

    Chenet

    al.[48]

    3245

    adultsin

    aTaiwan

    rural

    village,agedC18

    years,45.3%

    M

    US

    M(19.7%

    )[F

    (10.7%

    )

    Msexandmetabolic

    factors;

    ageC65

    years

    negative

    predictor

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforthe

    prevalence

    of

    NAFL

    D

    Park

    etal.[49]

    6648

    Koreansubjects,

    aged

    C20

    years,53%

    M

    US

    M(22.6%

    )[F

    (6.8%)before

    50years;similar

    after50

    years

    Menopausestatus

    and

    increasing

    agein

    F

    Menopauseisastrong

    risk

    factor

    for

    prevalentNAFL

    D

    Zelber-Sagi

    etal.[50]

    352Israelisubjects,m

    eanage

    51years,53.4%

    M

    US

    M(38%

    )[F(21%

    )M

    sex(after

    adjustmentfor

    obesity)

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforprevalent

    NAFL

    D

    Zhouet

    al.[51]

    3543

    inhabitantsof

    6urbanand

    ruralareasin

    China,

    aged[7years,37%

    M

    US

    M(13.8%

    )[F

    (7.1%)before

    50years;the

    opposite

    after

    50years

    Msexandmetabolic

    factors

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforprevalent

    NAFL

    D

    Liet

    al.[52]

    9094

    Chinese

    subjects(m

    edical

    check-up),aged[18

    years,52%

    M

    US

    M(18.9%

    )[F

    (5.7%);increased

    withagein

    both

    sexes\

    50years

    Msex,increasing

    age,

    BMI,andother

    MetSfeatures

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforprevalent

    NAFL

    D

    1296 Adv Ther (2017) 34:1291–1326

  • Table1

    continued

    Stud

    ypo

    pulation

    Diagnosisof

    FLPrevalence

    (cross-section

    alstud

    ies)

    Independ

    ent

    predictors

    Caballeriaet

    al.[53]

    766Spanishindividuals,aged

    17–8

    3years,33.4%

    M

    US

    M(42.2%

    )[F

    (20.3%

    )

    Msex,increasing

    age,

    andmetabolic

    factors

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforprevalent

    NAFL

    D

    Huet

    al.[54]

    7152

    employeesof

    Shanghai

    work-un

    its,aged

    18–6

    5years,

    60.5%

    M

    US

    M[

    Fin

    thesame

    agegroup,

    peaking

    at50–6

    5years;in

    F[

    50years

    doubled

    HighTG

    levelwas

    thestrongest

    predictorin

    M

    whileobesitythe

    strongestonein

    F

    Riskfactorsfor

    prevalentNAFL

    D

    aregend

    er-specific

    Wonget

    al.[55]

    922individualsfrom

    HongKong

    population,aged19–7

    2years,

    58%

    F

    1 HMRS

    M(36.8%

    )[F

    (22.7%

    );in

    M

    peakingat

    40years;

    inFincreasing

    after

    menopause

    Metabolicfactors(M

    sexandolderage

    NS)

    Genderisnotan

    independ

    entrisk

    factor

    forprevalent

    NAFL

    D

    Ham

    aguchi

    etal.[37]

    4401

    Japanese

    subjects(health

    check-up),aged

    21-80years,

    51.6%

    men

    US

    M(24%

    )and

    postmenopausalF

    (15%

    )[remenopausalF

    (6%);increased

    withagein

    Fonly

    Postmenopause

    and

    HRT

    NS(age

    and

    MetSadjusted);

    agingin

    prem

    enopausalF

    only(weightgain

    andMetSadjusted)

    MenopauseandHRT

    arenot

    independ

    entrisk

    factorsforprevalent

    NAFL

    D

    Eguchiet

    al.[56]

    8352

    Japanese

    subjects(health

    check-ups),aged21–8

    6years,

    51.8%

    M

    US

    M[

    Fat

    allages

    Metabolicfactors

    (age[50

    yearsin

    F

    only)

    Genderisnotan

    independ

    entrisk

    factor

    forprevalent

    NAFL

    D

    Adv Ther (2017) 34:1291–1326 1297

  • Table1

    continued

    Stud

    ypo

    pulation

    Diagnosisof

    FLPrevalence

    (cross-section

    alstud

    ies)

    Independ

    ent

    predictors

    Younossiet

    al.[57]

    11,613

    American

    participants

    (from

    NHANESIII)

    US

    19%

    Fsexandyoun

    gerage

    forlean

    NAFL

    D;

    theopposite

    for

    NAFL

    Dwith

    BMIC25

    Fsexisarisk

    factor

    forlean

    NAFL

    D

    Lazoet

    al.[58]

    12,454

    American

    subjects(from

    NHANESIII),aged

    20–7

    4years

    US

    M(20.2%

    )[F

    (15.8%

    )

    Prevalence

    inM

    (20.2%

    )[F

    (15.8%

    )(adjusted

    forage,race,B

    MI,

    T2D

    )

    Mgend

    eris

    associated

    withan

    increasedNAFL

    D

    prevalence

    Wanget

    al.[59]

    4226

    Chinese

    subjects[60

    years

    vs3145

    controls\60

    yearsfrom

    thesamecohort

    US

    M(32%

    )[F(9%)

    before

    60years;

    similarafter

    60years

    Msex(onlyin

    those\

    60years)

    andmetabolic

    factors

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforprevalent

    NAFL

    D

    Xuet

    al.[41]

    6905

    non-obese(BMI\

    25)

    Chinese

    subjects,6

    3%M

    US

    7.3%

    Msex,youn

    gerage,

    andmetabolic

    factors

    Msexandmetabolic

    factorsare

    independ

    entrisk

    factorsforprevalent

    NAFL

    D

    Yan

    etal.[60]

    3762

    Chinese

    residents,aged

    20–9

    2years,61.9%

    M

    US

    M(45%

    )[F(30%

    );

    higher

    before

    50yearsbutsimilar

    after

    Metabolicfactors(M

    sexpredictedonly

    atun

    ivariate

    analysis)

    Msexisarisk

    factor

    forprevalent

    NAFL

    Dat

    univariate

    analysis

    only

    1298 Adv Ther (2017) 34:1291–1326

  • Table1

    continued

    Stud

    ypo

    pulation

    Diagnosisof

    FLPrevalence

    (cross-section

    alstud

    ies)

    Independ

    ent

    predictors

    Chiloiroet

    al.[61]

    2974

    SouthItaliansubjects,aged

    30–8

    9years,56.5%

    M

    US

    M(37%

    )[F(26%

    )MetSanditsfeatures,

    BMI,visceral,and

    subcutaneous

    fatin

    both

    MandF

    Riskfactorsfor

    prevalentNAFL

    D

    arenot

    gend

    er-specific

    Foster

    etal.[62]

    3056

    American

    multiethn

    ic

    subjects,m

    eanage63

    years,55%

    F

    CT

    NA

    Fsexprotective

    inall

    andAAs;notin

    Caucasiansand

    Hispanics.Y

    ounger

    agein

    all,

    Caucasiansand

    Hispanics;notin

    AAs

    Fsexisspared

    from

    prevalentNAFL

    D

    Yanget

    al.[63]

    2256

    Chinese

    subjects(health

    check-up),meanage62.04years,

    87.9%

    M

    US

    M(28.4%

    )[F

    (20.3%

    )

    Younger

    age,RDW

    andmetabolic

    factors(genderNS)

    Genderisnotan

    independ

    entrisk

    factor

    forprevalent

    NAFL

    D

    Saidaet

    al.[64]

    3110

    Japanese

    subjects(health

    check-up),aged

    C30

    years,

    59.5%

    M

    US

    M(45%

    )[F(23%

    )Weightgain

    C10

    kg

    sincetheageof

    20yearsregardless

    ofsex

    Genderisnotan

    independ

    entrisk

    factor

    forprevalent

    NAFL

    D

    Wanget

    al.[65]

    25,032

    Chinese

    subjects(health

    check-up),aged

    18–9

    4years,

    62%

    M

    US

    M(32%

    )[F(13%

    );

    similarin

    both

    sexes[

    60years

    Metabolicparameters

    inboth

    sexes;

    increasing

    age

    (C60

    years)onlyin

    F

    Riskfactorsfor

    prevalentNAFL

    D

    arenot

    gend

    er-specific

    Adv Ther (2017) 34:1291–1326 1299

  • Table1

    continued

    Stud

    ypo

    pulation

    Diagnosisof

    FLPrevalence

    (cross-section

    alstud

    ies)

    Independ

    ent

    predictors

    Schn

    eideret

    al.[66]

    4037

    non-Hispanicwhite,2

    746

    non-Hispanicblack,

    and2892

    Mexican–A

    merican

    adultsin

    theNHANESIII

    US

    M(15%

    )[F(10%

    )

    among

    non-Hispanic

    whitesonly

    (age-adjusted)

    Genderdifference

    still

    significant

    after

    adjustmentfor

    BMIandWC

    Mgend

    eris

    associated

    withan

    increasedNAFL

    D

    prevalence

    independ

    entof

    visceralobesity

    Xiaoet

    al.[67]

    18,676

    Chinesesubjects,m

    eanage

    40.6years,55.4%

    M

    US

    M(33%

    )[F(8%)

    Msexandmetabolic

    factors

    Msexisan

    independ

    entrisk

    factor

    forprevalent

    NAFL

    D

    Martı́nez-Alvaradoet

    al.[68]

    846Mexican

    subjects,m

    eanage

    53years,49.3%

    M

    CT

    M(36.5%

    )[F

    (28.4%

    )

    Metabolicfactors

    (MetSin

    Fonly)

    Genderisnotan

    independ

    entrisk

    factor

    forprevalent

    NAFL

    D

    Liu

    etal.[69]

    11,200

    Chinese

    gallstone-free

    subjects,4

    6.7%

    M

    US

    M(31%

    )[F(13%

    )NA

    NAFL

    Dismore

    prevalentin

    M

    gend

    er

    Amirkalaliet

    al.[70]

    5023

    Iranianparticipants,m

    ean

    age45.35years,56.7%

    M

    US

    F(46%

    )[M

    (42%

    );

    peakingin

    M

    40–6

    0years,in

    F[60

    years

    Age,M

    etS,

    andits

    factors(higherORs

    inF)

    Genderisnotan

    independ

    entrisk

    factor

    forprevalent

    NAFL

    D

    Nishiojiet

    al.[71]

    3271

    Japanese

    subjects(health

    check-up),44.2%

    M

    US

    M[

    Fforallages,

    except

    in

    obeseC70

    years

    Metabolicfactorsin

    MandF

    Riskfactorsfor

    prevalentNAFL

    D

    arenot

    gend

    er-specific

    Huang

    etal.[72]

    8626

    Chinese

    subjectsC40

    years

    US

    M(28.7%

    )*

    F

    (28.1%

    )

    NA

    Nogend

    erspecific

    difference

    in

    prevalentNAFL

    D

    1300 Adv Ther (2017) 34:1291–1326

  • been also observed in male obese children andadolescents than in female ones [78].

    Men commonly display an increasingprevalence of NAFLD during adulthood fromyoung to middle-age, describing an ‘‘invertedU-shaped curve’’ which starts declining after theage of 50–60 years [47, 56]. In women of fertileage, the prevalence of NAFLD is lower than inmen owing to the putative protective effect ofestrogens which, however, wanes after themenopause. Accordingly, the prevalence ofNAFLD 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 fifthdecade, postmenopausal women compared tomen of the same age have a similar[46, 49, 60, 65] or even higher [47, 51] preva-lence of NAFLD. In agreement with these find-ings, a multicenter study from northern Italyfound that men with NAFLD were approxi-mately 10 years younger than women with thiscondition [79], a finding compatible with thehypothesis that premenopausal women are‘‘protected’’ from developing NAFLD. Con-versely, NAFLD is more prevalent in post-menopausal [37, 80–82] and PCOS-affectedwomen than premenopausal ones [80]. Consis-tently, a large cross-sectional population-basedsurvey in northeast Germany on 808 womenaged 40–59 years showed that menopause statuswas independently associated with hepaticsteatosis after adjustment for metabolic factors[83]. Interestingly, women with NAFLD exhibita significantly lower concentration of serumestradiol, which is the principal active estrogen,than NAFLD-free (premenopausal, post-menopausal, and PCOS) controls [80]. A lowerprevalence of NAFLD, as well as of MetS, hasbeen reported in postmenopausal womenreceiving HRT compared to those not receivingit, which suggests that HRT probably protectsfrom NAFLD. Moreover, in this study post-menopausal women with NAFLD who receivedHRT had lower frequency of insulin resistanceas well as MetS and showed reduced serumlevels of liver enzymes and ferritin [84].

    Most studies using multivariate analysis haveshown that male sex is associated with anincreased prevalence of NAFLD independentlyof age and metabolic conditionsT

    able1

    continued

    Stud

    ypo

    pulation

    Diagnosisof

    FLPrevalence

    (cross-section

    alstud

    ies)

    Independ

    ent

    predictors

    Fattahiet

    al.[73]

    2980

    Iraniansubjects,

    aged

    C18

    years,31.3%

    M

    US

    M(32.9%

    )[F

    (27.4%

    )

    NA

    NAFL

    Dismore

    prevalentin

    M

    gend

    er

    Motam

    edet

    al.[74]

    5052

    Iraniansubjects,

    aged

    C18

    years,56.6%

    M

    US

    F(44.2%

    )[M

    (40.1%

    )

    Age,m

    etabolic

    factors;Fsex

    protective

    Fsexisspared

    from

    prevalentNAFL

    D

    AAsAfrican-Americans,BMIbody

    massindex,Ffemale/s,FL

    fattyliver,F

    -upfollow-up,

    1 HMRSproton

    magneticresonancespectroscopy,H

    OMAhomeostasis

    modelassessment,HRThorm

    onereplacem

    enttherapy,M

    male/s,MetSmetabolicsynd

    rome,NAnotassessed,N

    AFL

    Dnonalcoholicfattyliverdisease,NHANES

    IIINationalHealth

    andNutrition

    ExaminationSurvey

    III,NSnotsignificant,O

    Rodds

    ratio,RDW

    redbloodcelldistribution

    width,T

    2Dtype

    2diabetes,T

    Gtriglycerides,USultrasound

    ,WCwaistcircum

    ference

    Adv Ther (2017) 34:1291–1326 1301

  • [41, 46, 48, 50, 51, 53, 58, 62, 63, 66, 67, 74].Some studies have shown an independent,positive association between NAFLD andincreasing age in both sexes [46, 50–53, 70, 74]or in females only [65], while others reported aninverse association [41, 48, 62, 63]. At variance,other studies have shown that sex differences inthe prevalence of NAFLD were mostly accoun-ted for by metabolic factors [36, 37, 55, 56, 60,63, 64]. Finally, some studies failed to investi-gate the role of gender given that a separatemultivariate analysis according to sex was notperformed [34, 49, 54, 56, 61, 65, 68, 70, 71].

    In addition to and independent of the role ofMetS and its components, menopausal status[49, 83, 85] and increasing age [49, 56, 82] haveall been consistently identified as strong riskfactors for NAFLD in women. Only in a fewstudies was the association between menopau-sal status and NAFLD no longer significant afteradjustment for age and metabolic factors[37, 81], indicating that menopause predisposesto developing NAFLD via incident dysmetabolictraits which typically appear in the post-menopausal age.

    A recent cross-sectional study conducted in apopulation of postmenopausal women con-cluded that MetS, abdominal obesity, and IR arerisk factors for the development of NAFLD whilehigher adiponectin levels protect from devel-oping it [86]. These findings indicate thatNAFLD modifiers in this specific population ofwomen closely mirror those found in the gen-eral population.

    In lean subjects [body mass index (BMI) lessthan 25 kg/m2)] NAFLD has been associatedwith 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 electronichealth file database reported that the risk of‘‘recorded’’ NAFLD/NASH diagnosis increasedlinearly with BMI, was higher in males thanfemales and in those with T2D [87]. However, theproportion of true NASH cases among the ‘‘recor-ded’’ diagnosis of NAFLD/NASH was unclear.

    A longitudinal study in subjects with biop-sy-proven NAFLD found that sex was not anindependent risk factor for the progression offibrosis [88]. In agreement, a systematic reviewhas shown that age and hepatic necroinflam-mation are the only independent predictors ofthe development of advanced fibrosis in NASHpatients, while other parameters such as MetSfeatures and sex are not [89].

    However, conflicting with the above studies,data from cross-sectional studies (Table 2)[29, 57, 65, 90–97], which are based, in themajority of cases, on a histological diagnosis ofNASH, tend to suggest that the risk of NASH andadvanced fibrosis is indeed higher in femalesthan males independent of metabolic factors[65, 90, 93, 94], and only a few studies conflictwith the above findings [91, 92, 95].

    As is easily foreseeable, obese and post-menopausal women, compared to pre-menopausal and non-obese women, suffer froma remarkably higher prevalence of NAFLD andNASH 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 hepatocyteinjury was significantly increased both in pre-menopausal women and in those taking syn-thetic hormones such as oral contraceptives andHRT [97]. Given the supposed effects of estro-gens on metabolic health and liver injury, thefindings of this study appear counterintuitive. Itis noteworthy that the study by Yang et al. hasseveral limitations, including some importantsources of potential bias, such as the restrictedenrollment at tertiary academic centers, meno-pausal category and synthetic hormone usewere self-reported, and information on cumu-lative estrogen and/or progesterone exposureand serum hormonal levels were lacking. Ofnote, the authors highlight that, despiteincreased liver injury and inflammation, pre-menopausal women were at decreased risk ofliver fibrosis compared with men and post-menopausal women. Moreover, sensitivityanalyses separately assessing the impact of pro-gesterone use and estrogen use clearly suggestedthat only the former was associated with liverdamage. Collectively these findings provide

    1302 Adv Ther (2017) 34:1291–1326

  • Table2

    Impact

    ofgend

    eron

    NASH

    /fibrosisprevalence

    Stud

    yMetho

    dCross-section

    alstud

    ies

    Find

    ings

    Con

    clusion

    Stud

    ypo

    pulation

    Diagnosis

    ofNASH

    Prevalence

    Independ

    entpredictors

    Singhet

    al.

    [90]

    71consecutive

    Asian-Ind

    ianNASH

    patients,aged9–

    57years,

    76.1%

    M

    Biopsy

    NA

    Fsexforfibrosisstage

    Fsexisan

    independ

    ent

    risk

    factor

    forfibrosis

    Hossain

    etal.[91]

    432American

    NAFL

    D

    patients,m

    eanage

    43.6years,22.9%

    M

    Biopsy

    26.8%

    NASH

    and

    17.4%

    moderate-to

    severe

    fibrosis

    MsexforNASH

    andmoderate-to-severefibrosis

    inaddition

    toethn

    icity,ALT,A

    ST,m

    etabolic

    factors

    Mgend

    erisastrong

    independ

    entrisk

    factor

    forboth

    NASH

    and

    fibrosis

    Al-h

    amoudi

    etal.[92]

    1312

    SaudiArabian

    inpatients,m

    eanage

    44.7years,51.0%

    M

    US

    16.6%

    NASH

    (by

    ALT[60

    U/L)

    Msex,youn

    gageandlowtotalCH

    predicted

    high

    ALT

    inNAFL

    D

    Mgend

    erisastrong

    independ

    entrisk

    factor

    for

    hypertransam

    inasem

    ic

    NAFL

    D

    Bam

    bha

    etal.[93]

    1026

    adults(N

    ASH

    CRN

    Database),m

    eanage

    48.8years,37%

    M

    Biopsy

    61%

    NASH

    and29%

    advanced

    fibrosis

    FsexforNASH

    andadvanced

    fibrosis;increasing

    ageforadvanced

    fibrosis;plus

    metabolicfactors

    Fsexisan

    independ

    ent

    risk

    factor

    forNASH

    andfibrosis

    Younossi

    etal.[57]

    11,613

    American

    participants(from

    NHANESIII)

    US

    12%

    NASH

    (bygrade

    2–3US-FL

    ?high

    ALT,A

    ST,and

    /or

    T2D

    /IR)

    Younger

    ageandmetabolicfactors(genderNS)

    Genderisnotan

    independ

    entrisk

    factor

    forNASH

    Tapperet

    al.

    [94]

    358NAFL

    Dpatients,

    62.9%

    M

    Biopsy

    NASH

    andadvanced

    fibrosis[

    inFvs

    M

    (45%

    vs30%;23%

    vs

    14%)

    Fsex,BMI,andALTforNASH

    ;age,AST

    ,and

    APR

    Iforadvanced

    fibrosis

    Fsexisan

    independ

    ent

    risk

    factor

    forNASH

    andfibrosis

    Adv Ther (2017) 34:1291–1326 1303

  • Table2

    continued

    Stud

    yMetho

    dCross-section

    alstud

    ies

    Find

    ings

    Con

    clusion

    Stud

    ypo

    pulation

    Diagnosis

    ofNASH

    Prevalence

    Independ

    entpredictors

    Wanget

    al.

    [65]

    25,032

    Chinese

    subjects

    (health

    check-up),aged

    18–9

    4years,62%

    M

    US

    NASH

    withadvanced

    fibrosis(byBAAT

    score,AST

    /ALT)[

    Fvs

    M

    NA

    Prevalence

    ofNASH

    with

    advanced

    fibrosisis

    higher

    inFsex

    Yanget

    al.

    [95]

    541American

    patientswith

    NASH

    ,meanage48

    years,35.1%

    M

    Biopsy

    100%

    NASH

    ;22%

    advanced

    fibrosis

    Msex,postmenopausalFstatus

    (premenopausal

    Freference;borderlin

    eP),p

    re-T2D

    /T2D

    for

    fibrosis

    Msexandpostmenopausal

    status

    areindepend

    ent

    risk

    factorsforfibrosing

    NASH

    Turolaet

    al.

    [29]

    244females

    and244

    age-matched

    males

    with

    NAFL

    D

    Biopsy

    F2–F

    4fibrosis

    Menopause,m

    etabolicfactors,andNASH

    for

    F2–F

    4fibrosisin

    FwithNAFL

    D

    Menopause

    isstrongly

    associated

    withfibrosing

    NASH

    Klairet

    al.

    [96]

    488postmenopausal

    NAFL

    Dsubjects

    Biopsy

    Advancedfibrosis

    38.4%

    inprem

    ature

    menopause

    Fvs

    32.7%

    inotherF

    Prem

    aturemenopause

    andtimefrom

    menopause

    foradvanced

    fibrosis(adjustedforage,race,

    metabolicfactors)

    The

    longer

    theduration

    of

    postmenopausalstatus

    thehigher

    therisk

    of

    NASH

    Yanget

    al.

    [97]

    1112

    American

    NAFL

    D

    patients(160

    prem

    enopausaland489

    postmenopausalF;

    463M)

    Biopsy

    NASH

    [in

    pre/postmenopausal

    F(62%

    )vs

    M(50%

    )

    Lobular

    inflammationrisk

    increasedin

    (1)

    prem

    enopausalF[

    MandpostmenopausalF,

    (2)oralcontraceptives

    andHRT

    users

    (adjustedforcovariatesof

    livermetabolicstress)

    Fertile

    ageandestrogen

    use

    may

    predispose

    tomore

    necroinfl

    ammatory

    NASH

    variants

    ALTalanineam

    inotransferase,A

    STaspartateam

    inotransferase,A

    PRIAST

    -to-plateletratioindex,BMIbody

    massindex,Ffemale/s,FL

    fattyliver,F-upfollow-up,

    HRT

    horm

    onereplacem

    enttherapy,

    Mmale/s,MetSmetabolic

    synd

    rome,

    NA

    notassessed,NAFL

    Dnonalcoholic

    fattyliver

    disease,

    NASH

    nonalcoholic

    steatohepatitis,NHANESIIINationalHealth

    andNutrition

    ExaminationSurvey

    III,NSnotsignificant,T2D

    type

    2diabetes,USultrasound

    ,WC

    waist

    circum

    ference,yearsyears

    1304 Adv Ther (2017) 34:1291–1326

  • novel hints regarding a potential multifacetedimpact of sex hormones in NAFLD.

    Consistent with the hypothesis that estro-gens do exert a beneficial effect on NAFLD,menopause has been independently associatedwith significant fibrosis both in women withNAFLD and in an experimental zebrafishsteatosis model [29]. A recent study has shownthat men display a higher risk of advancedfibrosis compared to premenopausal women,while after menopause both sexes show a simi-lar severity of liver fibrosis, suggesting thatestrogens protect from the development offibrosis [95]. A subsequent study limited tonon-obese women with biopsy-proven NAFLDconfirmed that, even in non-obese NAFLDpatients, postmenopausal women still had moresevere fibrosis, when compared to pre-menopausal subjects [98]. Accordingly, a studyconducted in 488 postmenopausal women withbiopsy-proven NAFLD has shown that, inde-pendently of age and metabolic factors, thelonger the estrogen deficiency in post-menopausal status is (i.e., premature meno-pause and time from menopause), the higherthe risk of fibrosis is [96].

    The role of gender in influencing liver-re-lated mortality in NAFLD is still uncertain,although some studies have reported that menare at an increased risk [99–101].

    Risk of Hepatocellular Carcinoma

    A universal feature of HCC is the striking maleprevalence, with a male/female ratio averaging2:1 to 7:1; the latter proportion is more oftenfound in HBV-related HCC [102]. Nevertheless,compared to viral etiologies of HCC,NAFLD-related HCC was found to be associatedwith the lowest male/female ratios in one study[103]. NAFLD-related HCC may also occur innon-cirrhotic livers, but this seems to be morelikely in men [104]. The sexual dimorphism inHCC is also maintained regarding prognosis,with women showing better survival rates[102, 105]. However, menopause seems toattenuate these advantages [102, 105].

    In summary, the incidence of NAFLD ishigher in men than in women, and some

    longitudinal studies indicate that male gender isan independent predictor of NAFLD develop-ment. The prevalence of NAFLD is globallyhigher in men than women, but after meno-pause women display a similar or even a higherprevalence compared to men, a finding sup-porting a protective effect of estrogens. Incross-sectional studies, male gender and meno-pausal status have often been associated withthe risk of NAFLD, independent of age andmetabolic factors. The prevalence of NASH andadvanced fibrosis has been found to be higherin postmenopausal women than in men; how-ever, longitudinal studies have failed to supporta role for gender in influencing the progressionof liver fibrosis. Finally, HCC is definitely morecommon in men than in women in cases due toviral etiology; NAFLD may probably lower themale/female ratio in the risk of developing HCCand it is possible that gender affects the prog-nosis of HCC.

    NAFLD AND CVD

    NAFLD is increasingly recognized as a multi-system disease [5]. A growing body of evidencesuggests that NAFLD (assessed by liver enzymes,imaging, or biopsy) is associated with increasedincidence and prevalence of subclinical andclinical CVD, mainly coronary heart disease(CHD), independently of age, gender, andmetabolic factors, as recently reviewed in detailelsewhere [106, 107]. In the general population,male sex is more prone to incident CHD underthe age of 50 years compared to women but,after menopause, the incidence in women dra-matically increases to approach that of men[108].

    Gender-Specific Risk of CVD in Studieson NAFLD and CVD

    Several longitudinal studies investigating theassociation between CVD and NAFLD havereported multivariate analysis models, whichhave been adjusted for multiple confounders,including sex; as a result of this, the influence ofgender in the risk of CVD cannot be evaluatedin such studies (reviewed in [106, 107]). A large

    Adv Ther (2017) 34:1291–1326 1305

  • population-based American study (NHANES III)found that NAFLD assessed with ultrasound,together with male sex, age, race, and metabolicfactors independently predicted incident CVD[109]. Another study addressing CHD as a pre-specified outcome found that patients withNAFLD had a higher 10-year risk for CHD, ascalculated by the Framingham risk score, thanthe matched control population. This estimatedrisk was higher in men than in women andmore CVD events were reported among menthan women at the end of 10 year follow-up,although this finding was not statistically sig-nificant [110].

    Many cross-sectional studies in which thediagnosis of NAFLD was based on liver imagingstudies (either ultrasound or CT scanning)showed an independent association of male sexwith coronary artery calcifications [111] as wellas significant CHD [112–115].

    Gender Differences in the AssociationBetween NAFLD and CVD

    Studies in which the diagnosis of NAFLD wasbased on surrogate indices, such as otherwiseunexplained raised liver enzymes, found thatsex modulates the association of NAFLD withincident CVD/mortality. For example, mostpopulation-based cohort and meta-analyticstudies reported an independent associationbetween raised GGT and incident CVD in bothsexes [116]; conversely, a population-basedcohort study from Germany found thatincreased GGT was associated with higher riskof all-cause and CVD mortality only in men butnot in women, and this association was strongerin men who also had ultrasound scanningfindings compatible with steatosis [101].Increased ALT has been variably associated withincident CVD either in both sexes [117, 118] orin men only [119]. A recent study found thatALT levels independently predicted insulinsensitivity only in women, suggesting that thisgender-specific association might explainthe sex difference in the predictive role ofincreased ALT for CVD [120]. Finally, a recentlarge Korean cross-sectional study reported thatmen had more prevalent ultrasonographic fatty

    liver disease, carotid plaques, and increasedcarotid intima-media thickness (IMT) valuesthan women, but ultrasonographic fatty liverdisease independently predicted subclinicalcarotid atherosclerosis (IMT and plaques) inwomen only [121].

    No gender difference has been reported inthe association between NAFLD assessed withliver ultrasound and incident CVD/mortality. Astudy based on a national Danish registryshowed that patients with a hospital dischargediagnosis of NAFLD had a higher all-causemortality, including liver and CVD related,which was similar among sexes [122]. A studyconducted in a community-based Japanesecohort of 1637 apparently healthy subjectsfound that the diagnosis of NAFLD based onultrasound findings was a predictor of CVD inboth men and women [123].

    The only study carried out in post-menopausal women found a significant corre-lation between NAFLD (based on CT scanningfindings) and prevalence of coronary arterycalcification (CAC); however, NAFLD was notindependently associated with CAC in thesepostmenopausal women [124].

    In summary, male patients carry anincreased risk of CVD; moreover, NAFLD seemsto be associated with CVD independently ofmetabolic factors in both sexes. Few data areavailable in postmenopausal women and stud-ies should specifically be conducted to ascertainwhether NAFLD is a specific/independent car-diovascular risk factor in this population ofpatients.

    ROLE OF GENDER, REPRODUCTIVESTATUS, AND AGEIN THE HETEROGENEITY OF NAFLDPATHOBIOLOGY

    Although NAFLD may be found in either genderfrom infancy to old adulthood, gender andreproductive status modulate the susceptibilityto development and progression of disease[125]. Indeed age, sex, and fertility exert avariable impact on those general pathogenicmechanisms which are involved in NAFLD.

    1306 Adv Ther (2017) 34:1291–1326

  • Here we will specifically examine how body fatdistribution, obesity and local hypercorticolism,steatogenesis and lipidomics, oxidative stressand antioxidant mechanisms, endotoxins,immune response, and fibrogenesis are affectedby gender and sex hormones as a result ofreproductive status and age.

    Factors Associated with NAFLD areDifferent in Men and in Women

    A pioneering study supporting the notion thatgender-dimorphic risk factors are associatedwith NAFLD was published in 2000. This study,by evaluating 199 individuals, found that ele-vated BMI was an independent predictor of fattyliver in either sex. Glucose area under the curveand a central-type body fat distribution pre-dicted fatty liver only in women [126]. Simi-larly, insulin sensitivity has also been reportedto be strongly associated with gender-dimor-phic risk factors, i.e., fasting insulin and leptinlevels (but none of the liver enzymes) in menversus BMI and ALT in women [120]. Of note,Suzuki et al. demonstrated that the associationsbetween anthropometric measures (regionaladiposity) and degree of fibrosis clearly differbetween premenopausal women and post-menopausal women/men [127].

    NAFLD Epidemiologyand Physiopathology are Modulatedby Age at Menarche and PostmenopausalStatus

    In women, a complex interaction includinggenetic polymophisms, dietary habits, endoge-nous sex hormones, age at menarche, meno-pausal status, dysmetabolic traits, and HRTmodulates the risk of developing NAFLD, NASH,and fibrosis [84, 128–130].

    Early menarche has been associated withhigher alanine aminotransferase, C-reactiveprotein, triglyceride levels, BMI, waist circum-ference, adult diabetes, cardiovascular morbid-ity and mortality, advanced liver disease, andHCC [131]. A recent Chinese study conductedin postmenopausal women has identified earlymenarche as a potential risk factor for NAFLD

    later in life; consistently, late menarche protectsfrom NAFLD [130]. These associations weresignificantly attenuated after adjustment forcurrent BMI or HOMA-IR, suggesting that obe-sity and insulin resistance may partly mediatethe association between age at menarche andNAFLD [130]. The biological mechanism link-ing early menarche with increased risk ofNAFLD is far from being clearly elucidated. Ithas been suggested that early maturation maydetermine a longer duration of positive energybalance and a greater accumulation of body fat[132]. Consistently, a large cross-sectional studyamong middle-aged Korean women confirmedthat the inverse association between age atmenarche and NAFLD was partially mediated byadiposity [133]. Again, a recent study from theAmerican CARDIA cohort showed that earlymenarche was associated with NAFLD and vis-ceral and subcutaneous abdominal ectopic fatdepots in middle adulthood; these associationswere attenuated after adjustment for weightgain between young and middle adulthood[131]. Finally, a Chinese study suggested thatthe presence of central obesity and MetS, butnot NAFLD, after menopause was predicted bylonger duration of menstruation and earlymenarche [134].

    Compared to the fertile age, menopauseincreases the risk of NAFLD and liver fibrosis[135] via long-standing estrogen deficiencyassociated with ovarian senescence and dys-metabolic features such as T2D, hypertriglyc-eridemia, and central obesity [29, 81].Consistently, HRT protects from NAFLD devel-opment [84], and oophorectomy in youngwomen with endometrial cancer independentlyincreases the risk of NAFLD together with thedevelopment of T2D and hypercholesterolemia[136]. These findings are in agreement with astudy suggesting that, in HCV infection,increasing severity of fibrosis is associated witha higher BMI, advanced steatosis, and themenopause and that, conversely, menopausalwomen receiving HRT exhibit a lower stage offibrosis [137]. Collectively, these data supportthe notion that estrogens have antifibrogenicproperties in humans. This antifibrotic activitymay occur by triggering anti-inflammatory,antioxidant, and antiapoptotic molecular

    Adv Ther (2017) 34:1291–1326 1307

  • pathways [138], which are mimicked by exercisetraining [139].

    Of concern, however, young women intheir reproductive age and those exposed tofemale synthetic hormones (oral contraceptiveor HRT) are not completely spared the risk ofdeveloping NAFLD and, indeed, they tend tohave more severe hepatocyte injury andinflammation. Notably, despite the possibilityof enhanced hepatocellular damage, pre-menopausal women have been consistentlyreported at decreased risk of liver fibrosiscompared to men and postmenopausalwomen. Moreover, sex hormones exert com-plex and variable effects on human NAFLD;indeed, the detrimental pro-inflammatoryimpact may be conveyed by progesterone, butnot estrogen [97].

    Table 3 summarizes the physiological role ofestrogen, progesterone, and androgens accord-ing to gender, age, reproductive status, andobesity [140–157].

    Obesity and Local Hypercortisolism

    Obesity, which is closely linked with NAFLD,mimics hypercortisolism. It is of interest thatdespite the prevalence of obesity being higheramong women than men, the former aresomewhat protected from the associated car-diometabolic consequences; however, thiswanes after menopause, suggesting a role forestrogens. Mouse models suggest that sexuallydimorphic expression and activity of glucocor-ticoid metabolizing enzymes may have a role inthe differential metabolic responses to obesityin males and females [158].

    Biochemical, genetic, and therapeutic stud-ies have provided robust evidence for 11b-hy-droxysteroid dehydrogenase type 1 (11b-HSD1)being key in the pathogenesis of NAFLD[159–161]. It is of interest, therefore, that11b-HSD1 gene expression is regulated in a tis-sue-specific and sexually dimorphic manner. Inparticular, intact rats exhibit hepatic 11b-HSD1mRNA levels 18-fold lower in the female thanthe male [162].

    Body Fat Distribution

    Regional adiposity displays a typical sexualdimorphism in humans. Women have a largercapacity to store fat in the subcutaneous com-partment [163]. Men generally have twice asmuch visceral fat compared to women for anygiven fat mass value [164]. This is relevant giventhat, compared to subcutaneous adipose tissuedepots, visceral adipose tissue depots in generaldisplay greater secretory capacity and a morepro-inflammatory profile [165]. Moreover, vis-ceral adipose depots release free fatty acids(FFAs) directly into the portal blood and thuspotentially overload the liver [166]. Not sur-prisingly, those phenotypes which occurwhenever, due to hormonal effect, peripheraladiposity is relatively restricted and visceraladiposity is expanded (i.e., male obesity andpostmenopausal women) have all been associ-ated with NASH, and fibrosing NASH[127, 167–169].

    Steatogenesis and Lipidomics are Affectedby Sex Hormones

    Steatosis will invariably occur as a result of animbalance among enhanced steatogenesis anddecreased capacity of oxidation of fatty acids[170]. Moreover, qualitative changes in thehepatic lipidomics may concur with lipotoxic-ity. Data suggest that all these mechanisms areunder the control of sex hormones. For exampleovariectomy in rats is associated with increasedintrahepatic steatogenesis which occursthrough a decreased synthesis of peroxisomeproliferator-activated receptor (PPAR), and anincreased transcription of genes encoding sterolregulatory element-binding protein 1[(SREBP-1), a nuclear transcription factor andmaster regulator of the endogenous synthesis ofcholesterol, fatty acids, triglycerides, and phos-pholipids] and stearoyl coenzyme A desaturase1 [(SCD1), the rate-limiting enzyme for gener-ating monounsaturated fatty acids (MUFA)from saturated fatty acids protects from hepa-tocyte lipotoxicity]; all these effects of

    1308 Adv Ther (2017) 34:1291–1326

  • Table3

    Physiologicalroleof

    horm

    ones

    Men

    Wom

    enBothsexes

    Obesity/type2diabetes

    Estradiol

    Estradiol

    inmen

    isessentialfor

    modulatinglibido,

    erectile

    function,and

    spermatogenesis

    [140]

    Astudycond

    uctedin

    healthymen

    suggeststhat

    estradiolprotects

    from

    NAFL

    D[141]

    End

    ogenousestrogensaremaster

    regulatorsof

    lipid

    metabolism

    andinhibitinflammation,

    vascular

    cellgrow

    th,and

    plaque

    progressionin

    prem

    enopausal

    wom

    en[142]

    The

    lossof

    estrogenswhich

    occurs

    postmenopausally

    isassociated

    withamodestincrease

    inLDL

    cholesterolw

    itheitherno

    change

    orasm

    alldecrease

    inHDL

    cholesterol.Estrogen

    administrationdecreasesLDL

    cholesteroland

    Lp(a)levelswhile

    increasing

    triglyceridesandHDL

    cholesterollevels,

    butthese

    effectsaredepend

    enton

    thedose

    androuteof

    administration

    [143]

    Estrogens

    improveinflammation

    relatedto

    metabolicdysfun

    ction

    (‘‘metaflam

    mation’’).

    Furtherto

    adirect

    downregulationof

    inflammatorypathways,this

    effect

    isalso

    mediatedby

    metabolicam

    elioration

    [144]

    Obesity

    isassociated

    with

    hyperestrogenism

    which,inturn,

    increasestherisk

    ofbreastcancer

    inmen

    [145]

    Progesterone

    Progesterone

    hasim

    portanteffects

    inregulating

    malefertility

    by

    affectingtheenergetic

    homeostasisof

    sperm

    [146]

    Progesterone

    hasamajor

    rolein

    theovarianandmenstrualcycle;

    moreoveritexertsan

    immun

    o

    regulatory

    function;regulates

    thecontractionof

    human

    intestinalsm

    ooth

    musclecells

    andthemotility

    ofvarious

    human

    celltypes[147]

    Progesterone

    isan

    independ

    ent

    predictorof

    insulin

    resistance

    in

    girls[148]

    Progesterone

    hasbeen

    potentially

    implicated

    asatherapeutic

    adjunctin

    manyclinical

    cond

    itions

    such

    astraumatic

    braininjury,A

    lzheim

    er’sdisease,

    anddiabeticneuropathy

    [147]

    Increasing

    levelsof

    progesterone

    have

    been

    associated

    withthe

    developm

    entof

    system

    icinsulin

    resistance

    [149]

    Little

    isknow

    nregardingtherole,if

    any,of

    serum

    progesterone

    in

    NAFL

    D

    Adv Ther (2017) 34:1291–1326 1309

  • Table3

    continued

    Men

    Wom

    enBothsexes

    Obesity/type2diabetes

    And

    rogens

    The

    synthesisof

    testosterone

    iskey

    tomalefertility.A

    negative

    feedback

    finelyregulatesthe

    secretionof

    horm

    ones

    atthe

    levelsof

    hypothalam

    ic-pituitary–gonadal

    axis.C

    ongenitalor

    acquired

    disturbancesof

    thisaxiswill

    lead

    tohypogonadism

    andthus

    impairmalefertility

    [150].

    Testosteronehasno

    significant

    correlationwithNAFL

    Din

    a

    studyfrom

    China

    [141].

    How

    ever,itisan

    independ

    ent

    predictorof

    insulin

    resistance

    in

    boys

    [148]

    And

    rogens

    have

    important

    biologicalrolesin

    youn

    gwom

    en,

    influencing

    bone

    andmuscle

    mass,vascular

    health,cognition,

    mood,

    well-b

    eing,and

    libido

    [151]

    How

    ever,testosteronedeficiency

    inyoun

    gwom

    enmay

    pass

    underdiagnosed

    becauseof

    generally

    nonspecific

    symptom

    s

    andinaccuracy

    oftestosterone

    measurement[152].The

    prim

    ary

    indication

    fortheprescription

    of

    testosterone

    forwom

    enislossof

    libido[153]

    Sarcopenia,n

    amelythedeclinein

    musclemassandstrength

    which

    occurswithageing,h

    asbeen

    associated

    withadeficiencyin

    both

    17b-estradioland

    testosterone,twosexhorm

    ones

    which

    acto

    nsatellitecells.T

    hese

    remainquiescentthroughout

    life

    andareactivatedin

    response

    to

    stressfulevents,enablin

    gthem

    to

    guiderepairandregeneration

    of

    theskeletalmuscle[154]

    Obese

    men

    tend

    tobe

    hypogonadic

    asaresultof

    thefunctional

    suppressionof

    the

    hypothalam

    ic–p

    ituitary–testicular

    axis[155].How

    ever,w

    eightloss

    obtained

    witheither

    alow-calorie

    diet

    orbariatricsurgeryis

    associated

    withthenorm

    alization

    ofsexhorm

    onelevelsexhibiting

    a

    significant

    increase

    inboun

    dand

    unboun

    dtestosterone

    and

    gonadotropin

    levelsandadecrease

    inestradiol[156].A

    studyfrom

    Japanreported

    that

    testosterone

    levelswereinverselyassociated

    withdiabetes

    amongmen

    butnot

    amongwom

    en[157]

    1310 Adv Ther (2017) 34:1291–1326

  • ovariectomy are prevented by 177b-estradiolreplacement, indicating a role for estrogens inthe prevention of hepatic fat accumulation[171]. Consistently, hypoestrogenemia is asso-ciated with hepatic steatosis through changes ingene expression of molecules related to fat oxi-dation and lipogenesis; resistance and endur-ance training prevent this both in rats andhuman models [172–175]. Moreover, a normalactivity of stearoyl-CoA desaturase (SCD), therate-limiting enzyme for generating monoun-saturated fatty acids from saturated fatty acids,protects from hepatocyte lipotoxicity. Indeed,in mouse models of NAFLD, either geneticmanipulation or dietary changes that inhibitthe activity of SCD promote fibrosing NASH viahepatocyte lipotoxicity, despite inhibiting obe-sity and improving insulin resistance [176, 177].Interestingly, ovarian hormones are alsoinvolved in MUFA biosynthesis, via SCD1 [178].

    Excess 16:0 fatty acids associated with denovo lipogenesis from high carbohydrate dietinhibits the synthesis of highly unsaturatedfatty acids; this may potentially account for theimproved metabolic profile which results fromsupplementing a hypercaloric diet with pre-formed eicosapentaenoic acid (EPA) anddocosahexaenoic acid (DHA) [179]. For exam-ple, findings from a genetically engineeredNASH mouse model fed a Western diet haveshown that dietary DHA was superior to EPA inattenuating Western diet-induced hyperlipi-demia and hepatic injury and at reversing thosedeleterious metabolic and histological effectsinduced in the liver by a Western diet [180].Interestingly, human (women using the con-traceptive pill or HRT and transsexual subjects)and experimental data in rats consistentlyindicate that sex hormones act to modifyplasma and tissue n-3 PUFA content, possibly byaltering the expression of desaturase and elon-gase enzymes in the liver [181]. Collectively,these findings suggest that enzyme activitieswhich are key in the development of a poten-tially hepatotoxic lipidomic signature are criti-cally controlled by sex hormones. Finally, it isof clinical importance that the therapeuticactivity of PPAR-a agonists in obesity and fattyacid oxidation is modulated by sex and estro-gens [182].

    Oxidative Stress and AntioxidantMechanisms

    Reactive oxygen species (ROS) result from theoxidation of fatty acids within mitochondriaand peroxisomes. The antioxidant defenseswhich physiologically constrain oxidative stressare hindered by nutritional deficiencies orchanges in the intestinal microbiome that limitavailability of choline [183]; by aging and thecontent of cysteine in diet, which, in turn, willaffect the intrahepatic synthesis of glutathione[184]; and by sex and menopause which bothaffect the normal metabolism of choline [185].

    Endotoxin

    Quali-quantitative changes in intestinal micro-biota (dysbiosis), which are often associatedwith dietary indiscretions, have consistentlybeen associated with increased gut permeabilitywhich could lead to increased translocation ofbacterial products from the intestinal lumeninto the portal circulation, thereby triggeringchronic inflammation [170]. It this context, it isrelevant that in an experimental model of liverfailure due to endotoxemia after hepatectomy,sexually mature female rats are more exposedthan males to endotoxin-induced liver injuryand that ovariectomy abrogates this sexualdimorphism [186]. Whether this also applies tohuman NAFLD, however, remains to be proven.

    Interindividual Variation in ImmuneResponses is Another Key Liver DiseaseModifier in NAFLD

    Whether metabolic inflammation is a gen-der-dimorphic phenomenon has not beenexplored in a systematic and organized manner.Obesity, however, will typically exhibit changesin the innate and adaptive immune mecha-nisms [187]. However, how mechanisticallysuch obesity-related dysregulation of immunedefenses will impact on the pathogenesis ofNASH is not fully elucidated. Experimental dataobtained in the ob/ob mouse model suggest arole for natural killer T cells (NKT cells) whichmay at least partly account for the

    Adv Ther (2017) 34:1291–1326 1311

  • interindividual variation in immune responsesbeing a key modifier in the development andprogression of NAFLD [125]. In this respect, itshould be highlighted that sex is a majordeterminant in the immune response. Sup-porting this notion, one study in young chil-dren reported that immune responses tovaccines were consistently higher or equivalentin girls compared with boys [188].

    Liver Fibrosis

    Liver fibrosis is the end-stage result of variousliver injuries. In recent times, importance hasbeen given to the Hedgehog signaling pathwayon the grounds that activation of this pathwaywill stimulate the proliferation and differentia-tion of hepatic stellate cells (HSCs) as myofi-broblasts (MF-HSCs), and, conversely,inhibiting Hedgehog activity in myofibroblastsderived from HSCs causes them to revert to amore quiescent (namely less fibrogenic) phe-notype [125]. Accordingly, Hedgehog ligandsand other factors that control fate decisions inHSCs are critical determinants of the develop-ment of cirrhosis due to various causes as well ofthe course of NASH [125]. For example, hepaticexpression of Hedgehog ligands and Hedgehogpathway activity progressively increase fromsimple steatosis, to NASH, and reach highestlevels in NASH-cirrhosis [189]. Of major signif-icance to the pathogenesis of NAFLD, theHedgehog pathway is strongly regulated bylipids [190], and conversely, Hedgehog signal-ing is a master regulator of body fat distributionand glucose metabolism [191, 192]. Thesefindings suggest that interindividual variationin Hedgehog signaling might contribute tovariability in both hepatic and extrahepaticoutcomes of the metabolic syndrome [125].

    Experimental overexpression of Hedgehogligand in hepatocytes is able to induce a fibro-genic liver response and to promote hepato-carcinogenesis in a transgenic mouse model[193]. At least in part by modulating interme-diary metabolism [194, 195], Hedgehog alsointeracts with pathways which regulate growthand differentiation [194, 196–198], such asWnt/b-catenin signaling, which are of potential

    significance for the development of hepatocel-lular carcinoma [199].

    Treatment with compounds, e.g., vitamin E,that suppress Hedgehog ligand production andreduce the hepatic accumulation of Hedge-hog-responsive myofibroblasts has proven ben-eficial in human NASH [200].

    Further studies are necessary to establishwhether and to what extent sex hormonesaffect Hedgehog signaling and how manipula-tion of cellular energy homeostasis might beexploited to prevent and manage fibrosingNASH, cirrhosis, and HCC in individuals withNAFLD.

    CAN GENDER DIMORPHISMOF NAFLD BE EXPLOITEDFOR THERAPEUTIC PURPOSES?

    In principle, sex differences found in NAFLDmay be accounted for by the effects of sexchromosomes; sex hormones; and by differ-ences in dietary and lifestyle habits [201, 202]. Abetter understanding of the physiopathologicalpeculiarities of NAFLD in women may con-tribute to developing tailored therapeuticinterventions [203]. Accordingly, the roles ofestrogen and HRT, the metabolism of choline,and the effect of weight reduction and exercis-ing in women are discussed in detail hereafter.

    Estrogens

    As detailed in Table 3, estrogens exert severalbeneficial metabolic effects. Experimental stud-ies suggest that estrogens promote the accu-mulation of peripheral gluteofemoralsubcutaneous adipose tissue and, within theliver, promote FFA beta-oxidation and preventthe accumulation of triglycerides; moreover,they regulate energy homeostasis, enhanceinsulin sensitivity, and exert a protective roleon the function of pancreatic beta-cells [201].Finally, estrogens seem to have antisteatotic,antioxidant, and antifibrogenic properties inthe liver [204, 205]. Estrogens may protect fromliver steatosis and fibrosis in female mice viaupregulation of miRNA-125b and miRNA-29,

    1312 Adv Ther (2017) 34:1291–1326

  • respectively [206, 207]. A recent experimentalstudy showed that the estrogen/estrogenreceptor alpha signaling plays essential roles inROS detoxification and in the reduction ofoxidative damage in the liver via partneringwith hepatic PPARG coactivator 1 alpha, thushalting the transition from simple steatosis tosteatohepatitis [205]. Estrogens are also knownto inhibit stellate cell activation and fibrogene-sis in experimental models [204]. Of note, astudy showed that the natural estrogen17b-estradiol prevents deoxycholic acid-in-duced hepatocellular damage in several celllines. This hepatoprotective effect of estrogenwas sustained by mechanisms which wereunlikely to be mediated by nuclear estrogenreceptor alpha [208]. Interestingly, it hasrecently been demonstrated that a non-nuclearestrogen receptor plays a key role in reducinghepatic steatosis in female mice [209]. Similarly,estrogen deficiency leads to fat redistributiontoward visceral fat accumulation and its inher-ently unfavorable metabolic derangements, andis thus able to induce the development andprogression of NAFLD/NASH both in mice andhumans. Of note, both aromatase knockoutmice, which are unable to synthesize endoge-nous estrogen, and estrogen receptor alphaknockout mice develop hepatic steatosis[210–212]. Patients with breast cancer treatedwith tamoxifen, a potent estrogen antagonist,develop progressive liver steatosis and NASH[213]. Surgical and physiological menopausalstatus have been invariably associated withexcess risk of NAFLD progression and liverfibrosis, and duration of estrogen deficiency hasbeen directly related to increased fibrosis risk inpostmenopausal women with NAFLD[95, 96, 98, 136].

    Hormonal Replacement Therapy

    Despite the above premises, the role of HRT inreverting the metabolic alterations associatedwith low levels of estrogens is a matter of dis-pute. In animal models, both estrogen andraloxifene, a second-generation selective estro-gen receptor modulator, have improvedinflammation and ballooning so halting the

    progression of liver fibrosis in diet-inducedNASH in female ovariectomized mice [30, 214].However, the theoretical benefits of HRT in liverdisease remain to be proven in humans. Aseminal randomized placebo-controlled studysuggested that HRT containing low-dose estra-diol and norethisterone was able to reduce liverenzyme concentrations in women with T2D,potentially through the reduction of accumu-lation of fat in the liver [45]. Accordingly, anorth American study using data of theNHANES III survey showed that post-menopausal women who received HRT had asignificantly lower risk of NAFLD than post-menopausal women who did not (OR 0.69, 95%CI 0.48–0.99) [215]. However, whether HRTreduces the risk of NASH and/or fibrosis amongpostmenopausal women remains uncertain.Two studies showed no, or borderline, benefi-cial effects of HRT on fibrosis among post-menopausal women with NAFLD [95, 96].Worryingly, a recent study reported that syn-thetic hormone use was associated with moresevere hepatocellular injury and lobularinflammation. Further analysis showed thatprogesterone, but not estrogen use, was associ-ated with worse liver histological lesions, sug-gesting a multifaceted impact of sex hormoneson NASH features [97].

    Choline

    Several lines of evidence support a key role ofcholine as an essential nutrient and cellularcomponent. Choline is the major source ofmethyl groups in the diet and is a precursor ofphosphatidylcholine, which is an importantcomponent of cell membranes and VLDL,required for the export of lipids from the liverinto the bloodstream. Thus, depletion of cholineinhibits hepatic triglyceride export and inducesfatty liver in both experimental models andhumans [185, 203, 216, 217]. Of note, post-menopausal women are more prone than pre-menopausal women and men to develop NAFLDas a consequence of choline deficiency[128, 185, 217], and decreased dietary cholineintake has been significantly associated withincreased fibrosis in postmenopausal women

    Adv Ther (2017) 34:1291–1326 1313

  • with NAFLD [218]. Indeed, menopausal statusdetermines a differential susceptibility to cholinedeficiency arising from the estrogen-inducibleexpression and activity of phos-phatidylethanolamine-N-methyltransferase(PEMT), a key enzyme in de novo cholinebiosynthetic pathway. Following menopause,the endogenous supply of choline decreases as aresult of estrogen deficiency; consequently, thedietary requirement of choline is increased[185, 203, 216, 217]. Consistently, choline sup-plementation has been reported to improve orrevert liver steatosis in patients receivinglong-term total parenteral nutrition [219]. How-ever, no interventional studies have addressedwhether choline supplementation is able to pre-vent NAFLD or reduce NAFLD progression; more-over, future studies investigating the role ofpreventive or therapeutic choline supplementa-tion in postmenopausal women are eagerlyawaited.

    Weight Reduction and Exercise

    Lifestyle changes, i.e., physical activity and bal-anced diet, are the milestone of intervention forNAFLD treatment. In general, weight lossinduced by lifestyle changes improves IR andfeatures of the MetS, including NAFLD. Of note, amodest weight loss of as little as 2–3 kg is associ-ated with NAFLD reversal [39], and weight loss ofgreater than 7–10% significantly improves his-tological features of NASH, including fibrosis[220]. However, significant weight reductionachieved through dietary restrictions results innegative effects on lean muscle and bone mass inpostmenopausal women. Therefore, whileweight reduction in postmenopausal womenwith metabolic abnormalities and risk factors forNAFLD should be strongly recommended, spe-cial attention should be paid to how weight loss isobtained in older women to prevent the wors-ening of loss of lean mass [139, 203]. An inte-grated approach consisting of dietary changesalong with regular exercise is mandatory to pre-vent the untoward effect of losing lean mass.Both resistance training and aerobic exercise areeffective in preventing muscle and bone lossduring weight loss [203], and equally reduce liver

    fat content [221, 222]. Postmenopausal womenwith higher levels of physical activity seem tohave lower total body and visceral fat and to beless likely to gain fat mass during menopause[139, 223]. However, only few studies havespecifically assessed the role of physical activityin NAFLD prevention/reversal in post-menopausal women. Data have shown thatexercise training effectively reduces liverenzymes in overweight postmenopausal women,probably as a reflection of liver fat content [224],and aerobic physical activity reduces cardiovas-cular risk factors in postmenopausal women withNAFLD [225]. Further studies are needed todefine the ideal structure and duration of exer-cise-based interventions in postmenopausalwomen with NAFLD or at risk of developing it.

    In summary, there are no codified therapeuticinterventions approved for tackling NAFLD inwomen. Given the theoretical beneficial meta-bolic and hepatic effects of estrogens, it isintriguing to speculate that HRT might possiblyplay a role in the prevention and treatment ofmetabolic alterations associated with meno-pause, including NAFLD. However, studies inhumans are lacking and the potential metabolicbenefits should be weighed, in clinical practice,against the detrimental cardiovascular and neo-plastic risk associated with long-term HRT[226, 227]. Moreover, the multifaceted and vari-able impact of different sex hormones in NAFLDshould be carefully considered. Dietary changesassociated with either aerobic or resistancephysical exercise should be considered as themilestone of treatment of NAFLD. Future studieswill have to evaluate the effects on NAFLD pre-vention/treatment of supplementing choline toa balanced diet in postmenopausal women.Finally, the physiopathological peculiarities ofNAFLD in women and the gender-based differ-ences in the kinetics and dynamics of drugs andxenobiotics should be taken into account whendeveloping new treatment strategies andproposing interventional trials for NAFLD.

    CONCLUSION

    Consistent with the notion that NAFLD andNASH are strongly linked with hormonal

    1314 Adv Ther (2017) 34:1291–1326

  • influences [228], this narrative review article wasdriven by the hypothesis that what we called the‘‘sexualdimorphism’’ ofNAFLD might be useful inidentifying clues of pathogenic significance andproviding hints for prevention and treatment ofNAFLD. On these grounds, a systematic researchof the literature was conducted. What we foundwas that not only are men at an increased risk ofdeveloping NAFLD (Fig. 1) but also significantage-related changes in NAFLD epidemiology inwomen may potentially bear physiopathological,clinical, and therapeutic significance. NAFLDepidemiology and physiopathology are modu-lated by age at menarche and postmenopausalstatus (Fig. 2). It would be expected that earlymenarche, definitely associated with estrogen

    activation, would produce protection against therisk of NAFLD. Nevertheless, it has been suggestedthat early menarche may confer an increased riskof NAFLD in adulthood, excess adiposity beingthe primary culprit of this association. Fertile agemay be associated with more severe hepatocyteinjury and inflammation, but also with adecreased risk of liver fibrosis compared to menandpostmenopausal status.Ovariansenescence isstrongly associated with severe steatosis andfibrosing NASH which may occur in post-menopausal women. Estrogen deficiency isdeemedtobe responsible for these findings via thedevelopment of postmenopausal metabolic syn-drome. Estrogen supplementation may at leasttheoretically protect from NAFLD development

    NAFLD RISK HIGHLOW

    PUTATIVE MECHANISMSNEMNEMOW

    GLUTEOFEMORAL Distribu�on of adipose �ssue VISCERAL

    KETONE BODY Fa�y acid par��oning VLDL-TAG

    + Browning of WAT -

    Fig. 1 Physiopathological grounds accounting for male sexas a strong predictor of NAFLD. Male gender andmenopausal status have been associated with the risk ofNAFLD independently of age and metabolic factors incross-sectional studies. On the basis of human studies andextrapolation of notions from animal studies, it can bespeculated that female sex is protected from dysmetabolismthanks to young individuals’ ability to partition fatty acidstowards ketone body production rather than VLDL-TAG

    [23], and to sex-specific browning of white adipose tissuewhich contributes in protecting female mice from exper-imental NAFLD associated with methionine cholinedeficient diet [24]. However, after menopause womendisplay a similar or even higher prevalence of NAFLDcompared to men, supporting a protective effect ofestrogens. Finally, risk factors associated with NAFLDdevelopment are different in men compared to women.TAG triacylglycerols, WAT white adipose tissue

    Adv Ther (2017) 34:1291–1326 1315

  • and progression, as suggested by some studiesexploring the effect of HRT on postmenopausalwomen, but the variable impact of different sexhormones in NAFLD (i.e., the pro-inflammatoryeffect of progesterone) should be carefully takeninto account when proposing treatment withsynthetic hormones.

    Taken collectively, these data may generateinnovative hypotheses to be tested in appro-priate clinical and experimental studies onNAFLD physiopathology and treatment.

    ACKNOWLEDGEMENTS

    No funding or sponsorship was received for thisstudy or publication of this article.

    All named authors meet the InternationalCommittee of Medical Journal Editors (ICMJE)criteria for authorship for this manuscript, takeresponsibility for the integrity of the work as awhole, and have given final approval for theversion to be published.

    Stefano Ballestri, Fabio Nascimbeni, andAmedeo Lonardo conceived the idea of thisarticle and wrote its first draft. All the authorshave read, edited, and approved the variousversions of this article.

    Disclosures. Stefano Ballestri and EnricaBaldelli have nothing to disclose. FabioNascimbeni is involved as a researcher in the‘‘Phase 3, Double Blind, Randomized,Long-Term, Placebo-controlled, Multicenter

    STEATOFIBROSIS

    Fig. 2 Hormonal changes are a major determinant ofprogressive NAFLD in human menopause. NAFLDepidemiology and physiopathology are modulated by ageat menarche and postmenopausal status. For example, earlymenarche may confer an increased risk of NAFLD inadulthood partly mediated by excess adiposity

    [130, 131, 133]. Moreover, ovarian senescence, via estrogendeficiency, may eventually be conducive to both massiveliver steatosis and its fibrotic evolution via dysmetabolictraits including T2D, hypertriglyceridemia, and visceralobesity which are often found postmenopausally[29, 81, 135]

    1316 Adv Ther (2017) 34:1291–1326

  • study evaluating the Safety and Efficacy ofObethicolic Acid in Subjects with NASH’’ (Eu-draCT 2015-002560-16). Alessandra Marrazzo isinvolved as a researcher in the ‘‘Phase 3, DoubleBlind, Randomized, Long-Term, Placebo-con-trolled, Multicenter study evaluating the Safetyand Efficacy of Obethicolic Acid in Subjects withNASH’’ (EudraCT 2015-002560-16). AmedeoLonardo is involved as a researcher in the ‘‘Phase3, Double Blind, Randomized, Long-Term,Placebo-controlled, Multicenter study evaluatingthe Safety and Efficacy of ObethicolicAcid in Subjects with NASH’’ (EudraCT2015-002560-16). Dante Romagnoli serves as aconsultant for AbbVie.

    Compliance with Ethics Guidelines. Thisarticle is based on previously conducted studiesand does not involve any new studies of humanor animal subjects performed by any of theauthors.

    Data Availability. All data generated oranalyzed during this study are included in thispublished article.

    Open Access. This article is distributedunder the terms of the Creative CommonsAttribution-NonCommercial 4.0 InternationalLicense (http://creativecommons.org/licenses/by-nc/4.0/), which permits any noncommer-cial use, distribution, and reproduction in anymedium, provided you give appropriate creditto the original author(s) and the source, providea link to the Creative Commons license, andindicate if changes were made.

    REFERENCES

    1. Brunt EM. Nonalcoholic fatty liver disease: pros andcons of histologic systems of evaluation. Int J MolSci 2016;17.

    2. Ballestri S, Nascimbeni F, Romagnoli D, et al. Therole of nuclear receptors in the pathophysiology,natural course, and drug treatment of NAFLD inhumans. Adv Ther. 2016;33:291–319.

    3. Piscaglia F, Svegliati-Baroni G, Barchetti A, et al.Clinical patterns of hepatocellular carcinoma in

    nonalcoholic fatty liver disease: a multicenterprospective study. Hepatology. 2016;63:827–38.

    4. Petta S, Valenti L, Bugianesi E, et al. A ‘‘systemsmedicine’’ approach to the study of non-alcoholicfatty liver disease. Dig Liver Dis. 2016;48:333–42.

    5. Byrne CD, Targher G. NAFLD: a multisystem dis-ease. J Hepatol. 2015;62:S47–64.

    6. Lonardo A, Sookoian S, Chonchol M, et al. Cardio-vascular and systemic risk in nonalcoholic fattyliver disease—atherosclerosis as a major player inthe natural course of NAFLD. Curr Pharm Des.2013;19:5177–92.

    7. Loria P, Marchesini G, Nascimbeni F, et al. Cardio-vascular risk, lipidemic phenotype and steatosis. Acomparative analysis of cirrhotic and non-cirrhoticliver disease due to varying etiology. Atherosclero-sis. 2014;232:99–109.

    8. Lonardo A, Ballestri S, Targher G, et al. Diagnosisand management of cardiovascular risk in nonal-coholic fatty liver disease. Expert Rev GastroenterolHepatol. 2015;9:629–50.

    9. Targher G, Byrne CD, Lonardo A, et al. Non-alco-holic fatty liver disease and risk of incident cardio-vascular disease: a meta-analysis. J Hepatol.2016;65:589–600.

    10. Nascimbeni F, Pais R, Bellentani S, et al. FromNAFLD in clinical practice to answers from guide-lines. J Hepatol. 2013;59:859–71.

    11. Lonardo A, Bellentani S, Ratziu V, et al. Insulinresistance in nonalcoholic steatohepatitis: neces-sary but not sufficient - death of a dogma fromanalysis of therapeutic studies? Expert Rev Gas-troenterol Hepatol. 2011;5:279–89.

    12. Ballestri S, Nascimbeni F, Romagnoli D, et al. Theindependent predictors of non-alcoholic steato-hepatitis and its individual histological features.Insulin resistance, serum uric acid, metabolic syn-drome, alanine aminotransferase and serum totalcholesterol are a clue to pathogenesis and candidatetargets for treatment. Hepatol Res.2016;46:1074–87.

    13. Ballestri S, Zona S, Targher G, et al. Nonalcoholicfatty liver disease is associated with an almost two-fold increased risk of incident type 2 diabetes andmetabolic syndrome. Evidence from a systematicreview and meta-analysis. J Gastroenterol Hepatol.2016;31:936–44.

    14. Younossi ZM, Koenig AB, Abdelatif D, et al. Globalepidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence,and outcomes. Hepatology. 2016;64:73–84.

    Adv Ther (2017) 34


Recommended