Subscribe to RSS
DOI: 10.1055/a-2516-0261
Influence of Sex in the Development of Liver Diseases
Funding This study was supported by the National Key Research and Development Program of China (2022YFC3400300), the National Natural Science Foundation of China (82170608), the Shaanxi Science Foundation for Distinguished Young Scholars (2021JC-36), the Shaanxi Innovative Research Team for Science and Technology (2022TD-53), and the “Young Talent Support Plan” of Xi'an Jiaotong University.
- Epidemiological Studies
- Sex Hormones/Sex Hormone Receptors
- X or Y Chromosome-Related Genes
- Other Potential Mechanisms
- Clinical Research and Guidelines
- Conclusion
- References
Abstract
The liver is a sexually dimorphic organ. Sex differences in prevalence, progression, prognosis, and treatment prevail in most liver diseases, and the mechanism of how liver diseases act differently among male versus female patients has not been fully elucidated. Biological sex differences in normal physiology and disease arise principally from sex hormones and/or sex chromosomes. Sex hormones contribute to the development and progression of most liver diseases, with estrogen- and androgen-mediated signaling pathways mechanistically involved. In addition, genetic factors in sex chromosomes have recently been found to contribute to the sex disparity of many liver diseases, which might explain, to some extent, the difference in gene expression pattern, immune response, and xenobiotic metabolism between men and women. Although increasing evidence suggests that sex is one of the most important modulators of disease prevalence and outcomes, at present, basic and clinical studies have long been sex unbalanced, with female subjects underestimated. As such, this review focuses on sex disparities of liver diseases and summarizes the current understanding of sex-specific mechanisms, including sex hormones, sex chromosomes, etc. We anticipate that understanding sex-specific pathogenesis will aid in promoting personalized therapies for liver disease among male versus female patients.
#
The liver functions differently between men and women. Sex differences in incidence, presentation, progression, and outcome exist for common liver diseases. However, the biological mechanism behind these sex differences is still largely unknown and needs to be studied. In 2016, the National Institutes of Health proposed sex as a biological variable, and more researchers have paid attention to the mechanism behind sex differences in liver pathophysiology. Here, we introduced the current knowledge on sex-based differences in common liver diseases, including sex-associated epidemiological characteristics, as well as underlying mechanisms and clinical implications of sex disparity in liver diseases. A better understanding of how sex differences affect liver health and disease is vital to achieving individualized health care and informed decision-making for all patients.
Liver disease, accounting for 2 million deaths annually, remains a leading cause of disability and mortality and has developed into a global public health problem.[1] According to epidemiological data, chronic liver disease is the tenth leading cause of death for men, but not for women.[2] Sex is a strong determinant of prevalence and outcomes in many diseases. However, essential biological differences between male and female individuals are rarely considered during clinical studies.[3] The consequences of overlooking sex are often deleterious for both sexes and can cause delayed diagnosis and inappropriate treatment.[4] For these reasons, a better understanding of sex-specific pathogenesis may help to personalize therapies for both men and women through the development of novel therapeutic approaches.
The liver is a sexually dimorphic organ. It plays a central role in many physiological processes, such as metabolism, energy and lipid storage, and xenobiotic detoxification, and accounts for at least 72% of sexually differentiated genes.[5] [6] In addition, most liver diseases predominantly affect one sex over the other. For example, men are two-fold more likely to develop and die from viral- or non-viral-related liver fibrosis, and five to six times more often in developing hepatocellular carcinoma (HCC) than women.[7] [8] [9] [10] [11] In contrast, females are more susceptible to drug- and alcohol-induced liver disease, as well as autoimmune hepatitis (AIH) than males.[12] [13] [14] [15] As such, sex difference has been recommended to be included in practice guidelines of liver disease.[10] In fact, biological sex differences in normal physiology and disease arise principally from sex chromosomes and/or sex hormones.[10] Sex hormones, notably estrogens and androgens, were found to regulate a variety of pathophysiological processes in the liver, including lipid metabolism, inflammation, and fibrogenesis.[16] [17] [18] [19] However, sex hormone supplements have failed in the treatment of most liver diseases.[20] [21]
As such, increasing studies have focused on the genetic alterations of sex chromosomes, and reported that several X- or Y-chromosome-related genes may contribute to sex disparity in liver diseases. For example, we and others have initially demonstrated that Y-chromosome-encoded genes, including the sex-determining region Y gene (SRY), promoted hepatocarcinogenesis, which might account for the male predominance of HCC.[22] [23] [24] [25] [26] In addition, we have further found that SRY, as the male determinant, exacerbated hepatic I/R injury and liver fibrosis.[27] [28]
Extensive studies have been conducted on exploring mechanisms involved in the sex disparity of liver diseases. As such, this review aims to illustrate the current knowledge on sex-based differences in several liver diseases, including sex-associated epidemiological characteristics, as well as underlying mechanisms and clinical implications of sex disparity in liver diseases ([Table 1]).
Diseases |
Epidemiology |
Sex hormones |
X or Y chromosome-related genes |
---|---|---|---|
DILI |
Estrogen: a protective role 1. inhibits gastric emptying[89] 2. inhibits expression of CYP1A2[93] 3. reduces inflammation and apoptosis[94] [95] Androgen: an aggravating factor 1. associates with proinflammatory monocytes recruitment[78] |
– |
|
ALD |
F: drinks less, but more likely to develop ALD[13] [46] [47] [48] [49] |
Estrogen: an aggravating factor 1. enhances intestinal permeability, portal endotoxin levels, and sensitivity to endotoxin[98] [99] [100] [101] |
Associates with alcohol habit formation[175] |
MAFLD |
Estrogen: a protective role 1. ERα: prevents hepatic lipid synthesis and deposition, mitigates oxidative damage, inhibits M1 macrophage activation[17] [109] [110] [111] 2. ERRα: mediates VLDL-TG secretion[112] 3. GPER: increases HDL level[113] [114] [115] Androgen 1. F: increased risk of MAFLD[119]; |
X chromosome 1. associates with increased adiposity[176] Y chromosome 1. SOX9: induces MASH-associated fibrosis[178] |
|
AILD |
F: higher levels of CD4+ T cells and antibody in PBC patients[125] |
X chromosome 1. DNA methylation, instability, and escape from inactivation in patients with PBC[124] [125] |
|
LF |
M: stronger progression and worse outcome[28] |
Estrogen: a protective role 1. inhibits ECM deposition and HSCs transformation[126] [127] [128] [129] [130] [131] [132] 2. ERβ: antifibrotic effect[135] [136] [137] [138] 3. ERα: anti-inflammatory effect[139] Androgen: an aggravating factor |
Y chromosome: 1. SRY: promotes fibrosis[28] |
HCC |
Estrogen: a protective role 1. ERα: inhibits HCC metastasis,[146] [152] promotes HCC apoptosis[153] [154] 2. ERβ: inhibits the malignant behavior of HCC cell[156] Androgen: an aggravating factor 1. enhances HBV induced HCC[141] 2. miR-216a → TSLC1 gene[147] 3. AR → CCRK → Wnt/β-catenin[148] |
X chromosome 1. DAX1 [184] 2.GPC3 rs2267531[185] Y chromosome 1.SRY: an oncogene[23] [24] [52] 3. RBMY, TSPY: new candidate oncogenes[26] [190] [191] [192] |
|
HIRI |
M: more susceptible to HIRI[27] |
Estrogen: a protective role 1. enhances the activity of hepatic eNOS[163] 2. regulates MPT[164] Androgen: an aggravating factor |
Y chromosome 1. SRY: an aggravating factor[27] 2. SOX9[193] |
Liver repair and regeneration |
Estrogen 1. ERα: promotes hepatocyte proliferation[171] 2. ERβ: mediates differentiation and metabolic function of hepatocytes[171] 3. GPER: cellular proliferation and cell cycle progression[172] 4. later onset of hepatocyte proliferation[81] Androgen 1. earlier onset hepatocyte proliferation[81] |
Y chromosome 1. SOX9: promotes hepatocyte regeneration and hepatocyte-to-BEC differentiation[194] [195] |
Abbreviations: ADH, alcohol dehydrogenase; AIH, autoimmune hepatitis; AILD, autoimmune liver diseases; ALD, alcohol-associated liver disease; BEC, hepatocyte-to-biliary epithelial cell; CYP, cytochrome P450 protein; DAX1, X chromosome congenital critical region gene 1; DILI, drug-induced liver injury; ECM, extracellular matrix; eNOS, endothelial nitric oxide synthase; ER, estrogen receptor; ERRα, estrogen-related receptor α; F, female; GPC3, glypican-3; GPER, G-protein-coupled estrogen receptor; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HDL, high-density lipoprotein; HIRI, hepatic ischemia/reperfusion injury; HSC, hepatic stellate cell; LF, liver fibrosis; M, male; MAFLD, metabolic dysfunction-associated fatty liver disease; MASH, metabolic dysfunction-associated steatohepatitis; MPT, mitochondrial permeability transition; PBC, primary biliary cholangitis; PSC, primary sclerosing cholangitis; RBMY, RNA-binding motif gene on Y chromosome; SOX4, SRY-Box transcription factor 4; SOX9, SRY-Box transcription factor 9; SRY, sex determining region Y gene; TSLC1, tumor suppressor in lung cancer-1; TSPY, testis-specific protein Y-encoded; VLDL-TG, triglyceride-rich very low-density lipoprotein.
Epidemiological Studies
Drug-Induced Liver Injury
Drug-induced liver injury (DILI) is an adverse reaction to a wide spectrum of medications, and it is the most common cause of acute liver failure and drug withdrawal from the market.[29] [30] [31] DILI is increasingly appreciated to be one of the most challenging diseases for clinicians. Some clinical trials demonstrated that women generally have a greater risk of adverse drug reactions,[32] [33] but other studies did not support this gender disparity in DILI cases.[34] [35] [36] Moreover, many potential confounding factors that can influence sex-dependent differences in drug toxicity, such as concomitant alcohol use, concurrent medications, and underlying fatty liver disease, need to be considered in human studies. While whether gender is a risk factor for susceptibility to DILI is still controversial, it is clear that female patients experience more severe clinical manifestations, have a higher frequency of jaundice, and are more likely to progress to acute liver failure than men.[34] [36] [37] [38] Acetaminophen (APAP) overdose is a leading cause of DILI; however, in contrast to clinical studies, a greater susceptibility of male mice to APAP hepatotoxicity has been well-documented.[39] [40] [41] The possible reason may be that female patients account for most cases of APAP overdose and have a higher prevalence of co-ingestions with sedating agents.[42] [43] [44] However, the reason for the disparity of phenotype in humans and mice needs to be further explored.
#
Alcohol-Associated Liver Disease
Alcohol-associated liver disease (ALD) is a spectrum of clinical disorders caused by excessive and chronic alcohol consumption that comprises alcohol-associated hepatitis (AH), alcohol-associated steatohepatitis, fibrosis/cirrhosis, and HCC.[45] Recently, there has been increasing evidence that the risk factors, incidence, severity, and clinical outcomes of ALD are influenced by sex. Alcohol consumption differs in males and females. In general, men drink more, drink more frequently, and are more likely to be diagnosed with an alcohol use disorder than women.[46] [47] [48] However, epidemiologic data suggest that by consuming equal doses of alcohol, women are more likely to develop ALD with faster progression and worse prognosis.[13] [49]
#
Metabolic Dysfunction-Associated Fatty Liver Disease
Metabolic dysfunction-associated fatty liver disease (MAFLD) is considered a complex metabolic disease characterized by an excessive fat accumulation in hepatocytes, and refers to a wide spectrum of liver damages ranging from metabolic dysfunction-associated fatty liver to metabolic dysfunction-associated steatohepatitis (MASH), advanced fibrosis, cirrhosis, and HCC.[50] Studies to date have supported that there are substantial sex differences in the prevalence, progression, and risk factors of MAFLD. Men are reported to have a higher prevalence of MAFLD than women.[51] [52] Moreover, it has been observed that postmenopausal women are at increased risk of MAFLD compared with premenopausal, and have a comparable prevalence of MAFLD as men.[53] [54] Intriguingly, it was shown that the prevalence of MAFLD in women steadily increased with age, whereas men had a similar prevalence in all age groups.[55]
Of note, risk factors associated with MAFLD are increasingly proven to be sex dimorphic. For example, obese men have lower insulin sensitivity versus obese women,[56] and the mechanism of which is strongly associated with sex-specific risk factors, that is, fasting insulin and leptin levels in men versus the body mass index and transaminase levels in women.[57]
#
Autoimmune Liver Diseases
Autoimmune liver diseases (AILDs) are rare chronic immune-mediated hepatobiliary disorders, including primary biliary cholangitis (PBC), AIH, and primary sclerosing cholangitis (PSC). Strikingly, PBC and AIH are characterized by a strong female preponderance, in-line with trends reported for autoimmune diseases that affect more women than men.[58] It has been reported that women are 10 times more likely to have PBC and 4 times more likely to have AIH, compared with men.[59] [60] However, it is not the case in PSC, which presented a male predominance, and men with PSC tended to have worse survival outcomes than women.[61] [62] [63]
#
Liver Fibrosis
Liver fibrosis is a sustained injury-healing process characterized by progressive deposition of extracellular matrix (ECM), resulting from chronic liver injury of any etiology, and resulting in fibrotic scarring and tissue dysfunction.[64] [65] Males have significantly stronger progression and worse outcomes than females. Of note, the death rate among males was estimated to be 1.5 times higher than that among females globally.[28]
#
Hepatocellular Carcinoma
Primary liver cancer ranks fifth in incidence and third in mortality among all cancers.[66] HCC, accounting for 85 to 90% of primary liver cancer, is the most common type of liver cancer.[67] [68] Of note, men had three to four times higher incidence of HCC than women.[67] [69] In addition, men had an age-standardized mortality rate of 2.8 versus women at the global level.[70]
#
Hepatic Ischemia/Reperfusion Injury
Hepatic ischemia/reperfusion (I/R) injury is a common occurrence in liver transplant, hepatic resection (HR), and hypovolemic shock, with its clinical manifestations ranging from asymptomatic elevation of liver enzymes to liver dysfunction, multiple organ failure, and even death.[71] I/R injury could enhance the risk of morbidity and mortality after liver transplantation or HR and lead to donor organ shortage as well as poor early graft function and primary non-function.[18] [71] Notably, it has been increasingly appreciated that sex-based differences exist in the response to I/R injury in many organs, including heart, brain, kidney, intestines, and the liver.[72] [73] [74] [75] [76] Males are more susceptible to hepatic I/R injury than females.[27]
#
Liver Repair and Regeneration
Liver repair and regeneration is a complex process following hepatic injuries, including acute liver injury, chronic liver injury, and hepatectomy.[77] Studies have demonstrated that females and males have different regenerative capacities. It has been found that, after acute poisoning with CCl4, male mice showed a slower recovery capacity than females.[78] Regarding regression of liver fibrosis, female mice showed lower collagen content and α-smooth muscle actin protein levels than males, indicating a better regeneration ability.[79] Additionally, clinical studies have demonstrated that female patients recover better from HR compared with males.[27] [80] However, conflicting results on sex disparities in initiation and speed of hepatic regeneration were reported. Female patients and mice regenerated faster than males after liver partition and portal vein ligation,[80] whereas male zebrafish began liver regeneration earlier than females after partial hepatectomy (PH).[81] To confirm sex differences in liver regeneration, further clinical studies are required. Although studies conducted in zebrafish and rodents can provide valuable references for humans, the differences between species need to be considered.
#
Other Liver Diseases
Several benign liver tumors, including cavernous hemangioma, focal nodular hyperplasia, and hepatic adenoma, predominantly occur in women.[82] Hereditary hemochromatosis (HH) is a disease caused by the toxic effects of excess iron deposition in parenchymal organs due to partial or total loss of hepcidin activity,[83] and it has been found that male patients with HH tend to have higher levels of ferritin, experience more iron accumulation, and have a higher incidence of liver injury and progression to fibrosis compared with women.[84] [85] In addition, some vascular liver diseases show sex differences in prevalence. Budd–Chiari syndrome, a rare disorder featuring venous flow obstruction at the suprahepatic level, is more common in males than females (1.5:1).[86] Hereditary hemorrhagic telangiectasia (HHT), a rare genetic condition that affects the liver by the development of venous malformations, shows a strong and significant predominance of hepatic venous malformations in females who have HHT, with a male:female ratio varying from 1:2 to 1:4.5.[87]
#
#
Sex Hormones/Sex Hormone Receptors
Drug-Induced Liver Injury
Previous studies have reported that sex hormones play a role in the regulation of drug reactions, hepatic inflammation, as well as the immune response following drug exposure, contributing to sex disparity in DILI.[88] It has been found that estrogen could inhibit gastric emptying, which directly influences the bioavailability of drugs taken orally.[89] Moreover, sex-specific disparity in drug-metabolizing enzymes was demonstrated. There is evidence that females have higher activity of CYP3A4, CYP2B6, and CTP2A6, and lower activity of CYP1A2 and CYP2E1.[90] [91] Sex hormones can regulate the expression and activity of cytochrome P450 protein (CYP) enzymes.[92] For example, estrogen inhibits the expression of CYP1A2.[93] In addition, estrogen exerts a protective role in DILI by reducing inflammation and apoptosis, whereas androgen exacerbates DILI.[94] [95] Furthermore, the drug-induced immune response was found to be sex dimorphic, characterized by higher recruitment of proinflammatory immature monocytes to the male liver through the androgen receptor (AR) signaling pathway.[78]
#
Alcohol-Associated Liver Disease
There are multiple mechanisms accounting for sex differences in the development of ALDs ([Fig. 1]). Orally ingested alcohol is metabolized in the stomach with the enzyme gastric alcohol dehydrogenase (ADH), the activity and level of which is demonstrated to decrease in females compared with males, leading to higher blood alcohol levels in females.[96] Excessive alcohol consumption is known to disrupt the gastrointestinal barrier and increase gut-derived endotoxin, subsequently inducing Kupffer cell (KC) activation, which proved crucial for the development of ALD.[14] [97] Estrogen could exacerbate ethanol-associated liver injury by enhancing intestinal permeability and portal endotoxin levels, upregulating the expression of the endotoxin receptor CD14 and proinflammatory cytokine TNF-a, and enhancing KC sensitivity to endotoxin.[98] [99] [100] [101] Hepatic ADH activity is higher in female rats and mice than their male counterparts.[102] Increased rates of the resulting production of toxic acetaldehyde in females compared with males may be responsible for the known increased susceptibility to alcohol-induced liver injury by females. In fact, steady expression of group hormones, resembling that in females, could increase hepatic ADH activity, which may accordingly contribute to sex disparity in ALD that females are more likely to progress from AH to cirrhosis even if they abstain.[103] In addition, hepatic CYP2E1, an important resource of reactive oxygen species (ROS) during ethanol metabolism, could be activated by estrogen-related receptor γ (ERRγ) and aggravate oxidative liver injury, which may account for severe alcohol-induced injury in females.[104] Additionally, the antagonist of estrogen or CYP2E1 has been demonstrated to alleviate experimental alcoholic liver injury, suggesting their potential therapeutic role in ALD.[105] [106] [107]


#
Metabolic Dysfunction-Associated Fatty Liver Disease
Numerous studies have demonstrated the protective role of estrogen in the development of hepatic steatosis ([Fig. 2]).[20] [51] [53] [108] In mice without estrogen receptor α (ERα), higher levels of obesity, especially visceral fat accumulation were observed, suggesting the crucial role of ERα in preventing pathologic lipid synthesis and deposition.[109] Moreover, ERα exerts a prominent role in ROS detoxification and mitigating oxidative damage in the liver through hepatic peroxisome proliferator-activated receptor-γ coactivator 1α.[17] [110] In addition, M1 macrophage activation is inhibited by ERα.[111] It has recently been demonstrated that estrogen-related receptor α (ERRα) mediates the secretion of hepatic triglyceride-rich very low-density lipoprotein and halts fat accumulation in the liver.[112] Moreover, the G-protein-coupled estrogen receptor (GPER) has proven to increase high-density lipoprotein levels in females, reducing female hepatic lipid accumulation.[113] [114] [115] In addition, estrogen may ameliorate hepatic steatosis in female mice through upregulating miRNA-125b and miRNA-29.[116] [117] Formyl peptide receptor 2, regulated by estrogen, can also play a key role in protecting female mice from MAFLD/MASH development.[118]


Testosterone appears to have opposite effects on hepatic steatosis in males and females ([Fig. 2]). High levels of testosterone in females were reported to increase the risk of MAFLD,[119] while high serum testosterone level was associated with decreased risk of MAFLD in men.[120] [121] Furthermore, it has been demonstrated that androgen signaling could suppress lipid deposits and play a protective role in hepatic steatosis.[122] [123]
Moreover, a phase 2 clinical trial (NCT04134091) has reported that LPCN 1144, an oral testosterone prodrug, can improve MASH activity in adult men, indicating the therapeutic potential of LPCN 1144.
#
Autoimmune Liver Diseases
Although sex disparity is increasingly indicated in AILDs, the exact mechanisms underlying this difference in general are unknown. Recent studies have discovered that several sex-related factors, such as environmental factors, sex hormones, and X and Y chromosomes, may influence sex differences in AILDs.[124] Moreover, in patients with PBC, female individuals are observed to have increased levels of CD4+ T cells and antibody production with respect to males, implying the role of sex hormones in autoimmune response.[125]
#
Liver Fibrosis
Previous studies have suggested the protective role of estrogen in liver fibrosis ([Fig. 3]). It was widely reported that high estrogen could counteract hepatic fibrogenesis by reducing ECM deposition and inhibiting the transformation of quiescent hepatic stellate cells (HSCs) to myofibroblast-like HSCs.[126] [127] [128] [129] [130] [131] [132] Given that HSCs typically express only estrogen receptor β (ERβ), it has been suggested that the antifibrotic effect of estrogen may be mediated by ERβ through inhibiting HSCs activation and proliferation.[133] [134] Furthermore, studies have demonstrated that ERβ alleviates liver fibrosis by negatively regulating the ROS/NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome and blocking the transforming growth factor β (TGF-β)/Smad signaling pathway.[135] [136] [137] [138] ERβ selective agonists may be a potential therapeutic target for liver fibrosis. In addition, ERα could inhibit liver inflammation and protect against hepatic fibrogenesis.[139]


In contrast, few studies have investigated the role of androgen in hepatic fibrosis. Ma et al reported that androgen exacerbates hepatic fibrosis by modulating the activation of NLRP3 inflammatory vesicles ([Fig. 3]).[140]
#
Hepatocellular Carcinoma
Increased androgen levels, overexpression of ARs, and more active androgen response elements have been identified in HCC ([Fig. 4]).[16] [141] [142] Mechanically, androgen enhances the replication and transcription of hepatitis B virus (HBV) genes and HBV-induced hepatocarcinogenesis by binding to the ARE in the enhancer I (Enh I) of the HBV genome.[141] In addition, hepatitis B virus X protein (HBx) can induce AR transactivation through c-Src and glycogen synthase kinase-3 kinase, which could improve the N-terminal domain of AR activity.[143] [144] [145] [146] Meanwhile, AR promotes HCC development by upregulation of miR-216a and downregulation of the tumor suppressor in lung cancer-1 (TSLC1) gene.[147] It was found that AR, cell cycle-related kinase (CCRK), and β-catenin form a vicious cycle in HCC. Androgen enhances the CCRK transcription, subsequently activating the Wnt/β-catenin signaling pathway, and enhancing the expression and function of AR in HCC.[148] As such, some have proposed that HCC was an androgen-response tumor like prostate cancer. However, targeting androgen or receptor signaling has little impact on the survival outcome of patients with HCC.[149] [150] [151]


Numerous studies have demonstrated the protective role of estrogen in hepatocarcinogenesis ([Fig. 4]). It was found that estrogen may act as an inhibitor of HCC cell metastasis via downregulating IL-6 production from KCs in the diethylnitrosamine-treated male mice.[152] Moreover, ERα inhibits the hepatocarcinogenic effects of IL-6 by inhibiting nuclear factor kappa-B activation regulated by the Toll-like receptor articulating protein myeloid differentiation primary response protein 88.[146] Similarly, ERα can transcriptionally upregulate the O-type protein tyrosine phosphatase receptor (PTPRO), then inactivating signal transducer and activator of transcription 3 by downregulating phospho-Janus kinase 2 levels to promote HCC apoptosis.[153] The estrogen-ERα pathway can also promote p53 activation in SNU-387 cells, subsequently activating caspase3 and HCC apoptosis.[154] Furthermore, it has also been proved that p53 may facilitate miR-18a processing to decrease the expression level of ERα in female patients with HCC.[155] Interestingly, studies revealed that ERβ activated by E2 could inhibit the malignant behavior of HCC in three HCC cell lines, BEL7402, SMMC7721, and HepG2, through mitogen-activated protein kinase/extracellular regulated protein kinases pathway-mediated upregulation of the NLRP3 inflammasome.[156] Herein, some researchers advocate that HCC is an estrogen-responsive tumor like breast cancer. However, several randomized controlled trials showed no survival advantage of anti-estrogen treatments of HCC.[157] [158] [159] [160] As such, there is no robust evidence to consider HCC as a hormone-responsive tumor.
#
Hepatic Ischemia/Reperfusion Injury
Estrogen could significantly reduce injury after I/R to the liver ([Fig. 5]).[161] [162] Mechanistically, it was reported that ERα could enhance the activity of hepatic endothelial nitric oxide synthase (eNOS) and alleviate I/R-induced liver injury.[163] In addition, estrogens and ERs are involved in the regulation of mitochondrial permeability transitions (MPTs), which are associated with Ca2+ dysregulation and subsequent cell death in I/R injury, suggesting sex differences in MPTs may contribute to disparities in I/R injury between men and women.[164] In contrast, male-related hormones, such as testosterone, aggravate I/R injury through its proinflammatory and proapoptotic effects ([Fig. 5]).[165] [166]


#
Liver Repair and Regeneration
The correlation between estrogen and liver regeneration has been reported for decades.[167] [168] [169] [170] It has been found that ER is activated following PH and can accelerate hepatocyte proliferation.[167] [168] A recent study demonstrated that ERs orchestrate hepatic regeneration. ERα promotes hepatocyte proliferation by upregulating Ssxb6, Crygc, and Cst1, while ERβ mediates differentiation and metabolic function of hepatocytes through upregulating Ifna5.[171] Moreover, GPER1 is also responsible for cellular proliferation and cell cycle progression.[172] On the contrary, another study showed that activated ERs were associated with the later onset of hepatocyte proliferation in female zebrafish.[81]
Unlike estrogen, the specific functions of androgen in hepatic regeneration are poorly investigated. A study of rats showed that the regenerating liver takes up testosterone following PH, indicating the involvement of AR in liver regeneration.[173] Furthermore, it has been reported that AR regulates sex-biased liver regeneration through interacting with Hippo-Yap1 signaling, accounting for earlier initiation of hepatocyte proliferation and recovery in male zebrafish.[81]
#
#
X or Y Chromosome-Related Genes
Drug-Induced Liver Injury
Few types of research focus on sex chromosomes and drug toxicity. A study by Du et al showed that XX or XY contributes to sex-specific response to toxicity in neurons and splenocytes.[174] However, whether the sex chromosomes exert a similar role in hepatocytes or cholangiocytes requires further investigation.
#
Alcohol-Associated Liver Disease
There is little evidence investigating sex chromosomes and ALD. A study by Barker et al proposed that sex chromosome complement influences alcohol habit formation, independent of gonadal phenotype and hormone circulation, suggesting its potential role in the pathogenesis of ALD.[175]
#
Metabolic Dysfunction-Associated Fatty Liver Disease
Apart from hormonal factors, present studies have suggested the sex chromosome may account for sex differences seen in MAFLD ([Fig. 2]). Chen et al found that XX mice showed greater adiposity than XY mice, even after gonadectomy was carried out.[176] Furthermore, it was suggested that the increased adiposity was attributable to the presence of an extra X chromosome, rather than the absence of a Y chromosome.[176] Notch signaling is implicated to have metabolic effects, such as increasing intrahepatic triglyceride synthesis, thus being considered to be involved in MAFLD pathogenesis and progression.[177] A study by Zhu et al demonstrated that Notch activation could enhance SRY-Box transcription factor 9 (SOX9)-dependent osteopontin (OPN) expression and secretion from hepatocytes, which activates resident HSCs, thus inducing MASH-associated fibrosis.[178]
#
Autoimmune Liver Diseases
The involvement of sex chromosomes in the development of PBC has been suggested by findings of decreased DNA methylation of X chromosome-related genes, sex chromosome instability, and escape from X chromosome inactivation in patients with PBC.[124] [125]
#
Liver Fibrosis
Recently, we have identified that among all Y chromosome-encoded genes, SRY was the most upregulated gene in toxin- or cholestasis-induced liver fibrosis of mouse model, and SRY in hepatocytes can transcriptionally regulate platelet-derived growth factor receptor alpha (PDGFRα) expression, and promote high mobility group box 1 (HMGB1) release and subsequent HSCs activation ([Fig. 3]).[28] As a target gene of SRY, SOX9 is ectopically expressed during activation of HSCs and is responsible for the production of type 1 collagen, a major component of the fibrotic ECM, which leads to scarring in liver fibrosis.[179] It was shown that SOX9 could modulate OPN expression and promote liver fibrosis through Hedgehog (Hh) signaling.[180] In addition, it was suggested that SOX9 could be used to detect the severity of hepatic fibrosis in patients.[181] Moreover, several long non-coding RNAs (lncRNAs) and miRNAs, such as miR-15a, lncRNA H19, have been found to regulate SOX9-induced liver fibrosis.[182] [183]
#
Hepatocellular Carcinoma
Genes in the X chromosome may similarly have a role in this sex difference ([Fig. 4]). It was found that the dosage-sensitive sex-reversal adrenal insufficiency X chromosome congenital critical region, gene 1 (DAX1), was downregulated in HCC tissues and cell lines, and could interact with β-catenin and downregulates Wnt/β-catenin pathway.[184] Additionally, glypican-3 (GPC3) rs2267531, located in the Xq26 region, has been reported to be associated with the risk of HCC, the mutation of which may contribute to X-linked recessive inheritance, contributing to a male predominance in HCC.[185]
For Y chromosome-specific genes, we and others have demonstrated that the male-specific gene SRY is an oncogene, and promotes male-specific hepatocarcinogenesis ([Fig. 4]).[23] [24] [52] SRY-related genes, such as SOX9, SOX4, are also associated with HCC progression.[186] [187] [188] Liu et al proposed that activation of SOX9 and PDGFRa/phosphatidylin-ositol-3-kinase/protein kinase B pathways are commonly involved in male hepatocarcinogenesis.[22] Moreover, Tsai et al found that SOX4 could regulate chemokine C-X-C motif ligand 12 in HCC and promote distant metastasis of tumors.[189] In addition, some other Y chromosome-encoded genes, such as the RNA-binding motif gene on the Y chromosome (RBMY) and testis-specific protein Y-encoded (TSPY), have been considered new candidate oncogenes specific for male liver cancer.[26] [190] [191] [192]
#
Hepatic Ischemia/Reperfusion Injury
Recent study from our group has demonstrated that SRY was significantly upregulated in male mice throughout the entire process of hepatic I/R injury ([Fig. 5]).[27] SRY exacerbates hepatic I/R injury by promoting β-linker phosphorylation and subsequent degradation, which leads to inhibition of Wnt/β-catenin signaling, suppression of FOXOs, and activation of NF-kB and TLR4 signaling, resulting in increased oxidative stress, inflammation, and cell death.[27] In addition, SOX9, a direct target gene of SRY, has also been found to promote hepatic I/R injury via enhancing inflammation and apoptosis by activating TGF-β1.[193] Thus, suppressing SOX9-TGF-β1 may be an effective therapeutic strategy to prevent hepatic ischemia/reperfusion injury.
#
Liver Repair and Regeneration
Few studies concentrate on the role of sex chromosome-related genes in liver repair and regeneration. However, recent studies have reported that SOX9 is involved in liver regeneration. After chronic liver damage, SOX9+ hepatocytes expand from the original periportal area to all liver zones and contribute to hepatocyte regeneration.[194] During chronic biliary injury, SOX9 in periportal hepatocytes can be upregulated and promote hepatocyte-to-biliary epithelial cell differentiation.[195] Similarly, after PH, the conversion of mature hepatocytes into SOX9+ hepatocytes increased via an lipopolysaccharide/B-cell lymphoma 3/yes-associated protein1 signaling pathway, suggesting an important role of SOX9+ hepatocytes in liver regeneration.[196]
#
#
Other Potential Mechanisms
Drug-Induced Liver Injury
The sexually dimorphic gut bacteria may also account for sex differences in drug exposure by modulating the pharmacokinetic properties of drugs.[92] Similarly, Gong et al found that the gut microbiota metabolite deguelin is responsible for the resistance to APAP-induced hepatotoxicity in female mice.[197]
#
Autoimmune Liver Diseases
In the case of PBC, it has been demonstrated that there are no specific male-related risk factors for the development of PBC, while many risk factors, including hair dye use, recurrent urinary tract infections, smoking, and estrogen deficiency have been identified in females and at least in part responsible for increased disease in females.[125] [198] [199] [200]
In addition, a study by Karlsen et al suggested that AILDs carry a significant association with HLA haplotypes, while only in AIH, a relationship between HLA expression and sex is described.[201] Furthermore, in AIH patients, males showed an enhanced expression of HLA-DR3, whereas an increased prevalence of HLA-DR4 was observed in females.[124]
#
Hepatocellular Carcinoma
Several studies identified that CYP39A1, a liver-specific autosomal gene with a female-preferential expression, could inhibit hepatocarcinogenesis by blocking the transcriptional activation activity of c-Myc through its c-terminal region.[151] Additionally, adiponectin, which was shown to block IL-6 production in macrophages, thus providing potential crosstalk between adiponectin and sex steroid studies in HCC, could protect against cancer development by activating AMP-activated protein kinase and p38α.[202] [203] Notably, research on zebrafish demonstrated that higher serotonin synthesis and accumulation in male models could result in enhanced activation of HSCs and TGF-β1 expression, which accelerated the progression of HCC in males.[204]
#
Hepatic Ischemia/Reperfusion Injury
There are sex differences in the production of cytokines/chemokines (e.g., IL-6, macrophage inflammatory protein-2, and KC), all of which are observed to be more significantly elevated in I/R-treated males versus females.[73] Potassium voltage-gated channel subfamily E member 4 (KCNE4) is also an important sex-specific modulator of hepatic I/R injury, and its deletion may produce sex-dependent inhibition of the “reperfusion injury salvage kinase (RISK)” pathway in mice and exacerbate hepatic I/R injury.[205] In addition, autophagy, one of the mechanisms of hepatic I/R injury, could be modulated through the X chromosome, androgen and estrogen receptors, which may also contribute to the sex differences in hepatic I/R injury.[206] As such, the sex differences in the development and prognosis of hepatic I/R injury should be considered in the perioperative management of patients who underwent liver resection or transplantation.
#
Liver Repair and Regeneration
It is well-known that the recruitment of monocyte-derived macrophages (MoMFs) correlates with liver regeneration.[207] During fibrosis regression, a larger number of Ly6Clow MoMFs were observed in female mice, facilitating restoration of liver fibrosis through promoting ECM degradation and apoptosis of activated HSCs.[79]
#
#
Clinical Research and Guidelines
As major causes of illness and death worldwide, liver diseases have an important influence on human health. The fact that sex disparities exist in liver diseases cannot be disregarded. At the moment, the advocacy for sex-inclusive research and reporting has been proposed.[208] [209] [210] There are several sex-specific clinical studies in the management of liver diseases ([Table 2]).
Disease |
Type |
Study objective |
Study participants |
Main findings |
References |
---|---|---|---|---|---|
ALD |
Clinical trial |
Examine the long-term effects of baclofen in alcohol-dependent patients |
30 females 70 males |
Lower maximal daily dose of baclofen in females |
[211] |
MAFLD |
Clinical trial |
Investigate sex differences on weight loss and NAFLD improvement in patients with severe obesity undergoing a VLCKD |
43 females 28 males |
Better response to VLCKD in males |
[225] |
MAFLD |
Clinical trial |
Evaluate effect of quercetin in intrahepatic lipid content in patients with MAFLD |
14 females 27 males |
Larger reduction of intrahepatic lipid contents in females |
[214] |
MAFLD |
Clinical trial |
Evaluate the efficacy of berberine and pioglitazone on NAFLD |
70 females 85 males |
Further decreased liver fat content in females after PGZ + LSI treatment |
[215] |
PBC |
Cross-sectional study |
Obtain a better picture of PBC phenotypes |
2,132 females 221 males |
Better response to UDCA in females |
[216] |
AIH |
Comparative study |
Examine the effect of gender on the clinical features and treatment outcome of type 1 AIH |
144 females 41 males |
Similar response to therapy, in females and males |
[220] |
AIH |
Retrospective cohort study |
Evaluate the clinical course and outcomes of male patients |
187 females 51 males |
Higher relapse rate and better long-term survival and outcome in males |
[221] |
Abbreviations: AIH, autoimmune hepatitis; ALD, alcohol-associated liver disease; MAFLD, metabolic dysfunction-associated fatty liver disease; PBC, primary biliary cholangitis; PGZ + LSI, pioglitazone based on lifestyle intervention treatment; UDCA, ursodeoxycholic acid; VLCKD, very low-carbohydrate ketogenic diet.
Abstinence from alcohol is the cornerstone of therapy for ALD. An open-label trial showed that the necessary maximal daily dose of baclofen in females was significantly lower than that in men to achieve craving control, indicating better treatment response in women.[211] Practice guidance of ALD from American Association for the Study of Liver Diseases has suggested that particular attention should be paid to sex in patients with alcoholic hepatitis.[212] Currently, there are no clinically approved drugs for MAFLD, and the mainstay of treatment is lifestyle modifications, including diet, exercise, and weight loss.[213] D'Abbondanza et al have reported that in patients with severe obesity receiving a very low-carbohydrate ketogenic diet (VLCKD), males experienced a significantly larger excess body weight loss and a better MAFLD improvement compared with females. Apart from lifestyle intervention, clinical trials on effective pharmacological treatments have also been conducted. Quercetin was found to reduce intrahepatic lipid contents effectively and perform more outstanding effects in women.[214] It was also reported that females with MAFLD profited more than males from pioglitazone based on lifestyle intervention treatment.[215] At the moment, ursodeoxycholic acid (UDCA) is one of the licensed therapies of PBC, and it was found that males were significantly less likely to respond to UDCA than females.[216] [217] The British Society of Gastroenterology PBC treatment and management guidelines have recommended that male sex should be included in risk assessment at baseline and on treatment.[218] Regarding AIH, prednisolone alone or in combination with azathioprine is an effective treatment.[219] However, in terms of sex-related responses to the therapy, studies to date have reported controversial results. For example, Czaja and Donaldson did not identify sex-related responses to steroids,[220] while Al-Chalabi et al found a higher relapse rate among reduction or withdrawal of treatment in male versus female patients.[221] Moreover, given the significant sex disparity in the development and prognosis of liver fibrosis, guidelines of hepatic fibrosis have proposed that patient sex needs to be considered when interpreting vibration-controlled transient elastography measurements and selecting patients in clinical trials of anti-liver fibrosis drugs.[222] [223] Additionally, gender dimorphism in the efficacy and toxicity of oncology treatments has been reported. A recent data analysis article showed that females have better outcomes and treatment responses, while males underwent fewer side effects, in the majority of oncology clinical trials.[224]
Although the sex differences have been widely reported, few sex-specific clinical studies have been conducted. Current evidence in liver diseases may be insufficient to propose sex-specific recommendations. As such, guidelines highlight the importance of sex and call on the research community to provide the evidence required.
#
Conclusion
Despite increasing evidence indicating that many diseases manifest differently in men and women, female individuals have long been underestimated in basic and clinical research. Exploring sex disparities in diseases is essential for getting the science right for the benefit of all. However, at present, sex-specific studies in liver diseases are still limited. Some controversial results are reported on sex-associated epidemiological characteristics in liver diseases. The underlying mechanisms of sex differences are not elucidated. Clinical trials rarely focus on disparities between males and females. Therefore, large-scale clinical and basic studies on sex differences are required to further explore sex differences and reveal underlying mechanisms. Clinical trials should incorporate women to observe the effects on the efficacy and safety of treatment. All these approaches can contribute to developing personalized therapies for liver diseases in both sexes.
#
#
Conflict of Interest
None declared.
Acknowledgments
All the figures in the manuscript were drawn in Figdraw.
Authors' Contributions
X-F.Z. and Y.L. conceived and mentored this study. J-W.Z. and N.Z. acquired and analyzed data, and wrote the draft of the manuscript. X-F.Z. and Y.L. wrote and revised the manuscript.
-
References
- 1 Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E, Kamath PS. Global burden of liver disease: 2023 update. J Hepatol 2023; 79 (02) 516-537
- 2 Heron M. Deaths: leading causes for 2017. Natl Vital Stat Rep 2019; 68 (06) 1-77
- 3 Woitowich NC, Woodruff TK. Opinion: Research community needs to better appreciate the value of sex-based research. Proc Natl Acad Sci U S A 2019; 116 (15) 7154-7156
- 4 Mauvais-Jarvis F, Bairey Merz N, Barnes PJ. et al. Sex and gender: modifiers of health, disease, and medicine. Lancet 2020; 396 (10250): 565-582
- 5 Roy AK, Chatterjee B. Sexual dimorphism in the liver. Annu Rev Physiol 1983; 45: 37-50
- 6 Yang X, Schadt EE, Wang S. et al. Tissue-specific expression and regulation of sexually dimorphic genes in mice. Genome Res 2006; 16 (08) 995-1004
- 7 Man S, Deng Y, Ma Y. et al. Prevalence of liver steatosis and fibrosis in the general population and various high-risk populations: a nationwide study with 5.7 million adults in China. Gastroenterology 2023; 165 (04) 1025-1040
- 8 Lefebvre P, Staels B. Hepatic sexual dimorphism - implications for non-alcoholic fatty liver disease. Nat Rev Endocrinol 2021; 17 (11) 662-670
- 9 Caballería L, Pera G, Arteaga I. et al. High prevalence of liver fibrosis among european adults with unknown liver disease: a population-based study. Clin Gastroenterol Hepatol 2018; 16 (07) 1138-1145.e5
- 10 Lonardo A, Nascimbeni F, Ballestri S. et al. Sex differences in nonalcoholic fatty liver disease: state of the art and identification of research gaps. Hepatology 2019; 70 (04) 1457-1469
- 11 Poynard T, Bedossa P, Opolon P. Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 1997; 349 (9055): 825-832
- 12 Floreani A, Bizzaro D, Shalaby S, Taliani G, Burra P. Special Interest Group Gender in Hepatology of the Italian Association for the Study of the Liver (AISF). Sex disparity and drug-induced liver injury. Dig Liver Dis 2023; 55 (01) 21-28
- 13 Szabo G. Women and alcoholic liver disease - warning of a silent danger. Nat Rev Gastroenterol Hepatol 2018; 15 (05) 253-254
- 14 Kezer CA, Simonetto DA, Shah VH. Sex differences in alcohol consumption and alcohol-associated liver disease. Mayo Clin Proc 2021; 96 (04) 1006-1016
- 15 Invernizzi F, Cilla M, Trapani S. et al; Special Interest Group Gender in Hepatology of the Italian Association for the Study of the Liver (AISF). Gender and autoimmune liver diseases: relevant aspects in clinical practice. J Pers Med 2022; 12 (06) 12
- 16 Barone M, Margiotta M, Scavo MP. et al. Possible involvement of androgen receptor alterations in hepatocarcinogenesis. Dig Liver Dis 2009; 41 (09) 665-670
- 17 Besse-Patin A, Léveillé M, Oropeza D, Nguyen BN, Prat A, Estall JL. Estrogen signals through peroxisome proliferator-activated receptor-γ coactivator 1α to reduce oxidative damage associated with diet-induced fatty liver disease. Gastroenterology 2017; 152 (01) 243-256
- 18 Li W, Li D, Sun L, Li Z, Yu L, Wu S. The protective effects of estrogen on hepatic ischemia-reperfusion injury in rats by downregulating the Ang II/AT1R pathway. Biochem Biophys Res Commun 2018; 503 (04) 2543-2548
- 19 Wang SH, Yeh SH, Lin WH. et al. Estrogen receptor α represses transcription of HBV genes via interaction with hepatocyte nuclear factor 4α. Gastroenterology 2012; 142 (04) 989-998.e4
- 20 Klair JS, Yang JD, Abdelmalek MF. et al; Nonalcoholic Steatohepatitis Clinical Research Network. A longer duration of estrogen deficiency increases fibrosis risk among postmenopausal women with nonalcoholic fatty liver disease. Hepatology 2016; 64 (01) 85-91
- 21 Nuermaimaiti A, Chang L, Yan Y. et al. The role of sex hormones and receptors in HBV infection and development of HBV-related HCC. J Med Virol 2023; 95 (12) e29298
- 22 Liu C, Ren YF, Dong J. et al. Activation of SRY accounts for male-specific hepatocarcinogenesis: Implication in gender disparity of hepatocellular carcinoma. Cancer Lett 2017; 410: 20-31
- 23 Murakami S, Chishima S, Uemoto H. et al. The male-specific factor Sry harbors an oncogenic function. Oncogene 2014; 33 (23) 2978-2986
- 24 Murakami S, Ninomiya W, Sakamoto E, Shibata T, Akiyama H, Tashiro F. SRY and OCT4 are required for the acquisition of cancer stem cell-like properties and are potential differentiation therapy targets. Stem Cells 2015; 33 (09) 2652-2663
- 25 Tsuei DJ, Lee PH, Peng HY. et al. Male germ cell-specific RNA binding protein RBMY: a new oncogene explaining male predominance in liver cancer. PLoS One 2011; 6 (11) e26948
- 26 Yin YH, Li YY, Qiao H. et al. TSPY is a cancer testis antigen expressed in human hepatocellular carcinoma. Br J Cancer 2005; 93 (04) 458-463
- 27 Dong J, Ke MY, Wu XN. et al. SRY is a key mediator of sexual dimorphism in hepatic ischemia/reperfusion injury. Ann Surg 2022; 276 (02) 345-356
- 28 Wu XN, Wang MZ, Zhang N. et al. Sex-determining region Y gene promotes liver fibrosis and accounts for sexual dimorphism in its pathophysiology. J Hepatol 2024; 80 (06) 928-940
- 29 Donnelly MC, Davidson JS, Martin K, Baird A, Hayes PC, Simpson KJ. Acute liver failure in Scotland: changes in aetiology and outcomes over time (the Scottish Look-Back Study). Aliment Pharmacol Ther 2017; 45 (06) 833-843
- 30 Weaver RJ, Blomme EA, Chadwick AE. et al. Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nat Rev Drug Discov 2020; 19 (02) 131-148
- 31 Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 2005; 4 (06) 489-499
- 32 Chalasani N, Bonkovsky HL, Fontana R. et al; United States Drug Induced Liver Injury Network. Features and outcomes of 899 patients with drug-induced liver injury: The DILIN Prospective Study. Gastroenterology 2015; 148 (07) 1340-52.e7
- 33 Amacher DE. Female gender as a susceptibility factor for drug-induced liver injury. Hum Exp Toxicol 2014; 33 (09) 928-939
- 34 Shen T, Liu Y, Shang J. et al. Incidence and etiology of drug-induced liver injury in Mainland China. Gastroenterology 2019; 156 (08) 2230-2241.e11
- 35 Sgro C, Clinard F, Ouazir K. et al. Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology 2002; 36 (02) 451-455
- 36 Lucena MI, Andrade RJ, Kaplowitz N. et al; Spanish Group for the Study of Drug-Induced Liver Disease. Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and sex. Hepatology 2009; 49 (06) 2001-2009
- 37 Andrade RJ, Lucena MI, Fernández MC. et al; Spanish Group for the Study of Drug-Induced Liver Disease. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology 2005; 129 (02) 512-521
- 38 Reuben A, Koch DG, Lee WM. Acute Liver Failure Study Group. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology 2010; 52 (06) 2065-2076
- 39 Dai G, He L, Chou N, Wan YJ. Acetaminophen metabolism does not contribute to gender difference in its hepatotoxicity in mouse. Toxicol Sci 2006; 92 (01) 33-41
- 40 Du K, Williams CD, McGill MR, Jaeschke H. Lower susceptibility of female mice to acetaminophen hepatotoxicity: role of mitochondrial glutathione, oxidant stress and c-jun N-terminal kinase. Toxicol Appl Pharmacol 2014; 281 (01) 58-66
- 41 Rohrer PR, Rudraiah S, Goedken MJ, Manautou JE. Is nuclear factor erythroid 2-related factor 2 responsible for sex differences in susceptibility to acetaminophen-induced hepatotoxicity in mice?. Drug Metab Dispos 2014; 42 (10) 1663-1674
- 42 Rubin JB, Hameed B, Gottfried M, Lee WM, Sarkar M. Acute Liver Failure Study Group. Acetaminophen-induced acute liver failure is more common and more severe in women. Clin Gastroenterol Hepatol 2018; 16 (06) 936-946
- 43 Kjartansdottir I, Bergmann OM, Arnadottir RS, Björnsson ES. Paracetamol intoxications: a retrospective population-based study in Iceland. Scand J Gastroenterol 2012; 47 (11) 1344-1352
- 44 Sami F, Berg S, Manadan AM, Mycyk MB. Acetaminophen overdose: analysis of 2018 US nationwide emergency database. Intern Emerg Med 2024; 19 (06) 1727-1732
- 45 Singal AK, Bataller R, Ahn J, Kamath PS, Shah VH. ACG Clinical Guideline: alcoholic liver disease. Am J Gastroenterol 2018; 113 (02) 175-194
- 46 Müller C. Liver, alcohol and gender. Wien Med Wochenschr 2006; 156 (19–20): 523-526
- 47 Sayaf K, Gabbia D, Russo FP, De Martin S. The role of sex in acute and chronic liver damage. Int J Mol Sci 2022; 23 (18) 23
- 48 Erol A, Karpyak VM. Sex and gender-related differences in alcohol use and its consequences: contemporary knowledge and future research considerations. Drug Alcohol Depend 2015; 156: 1-13
- 49 Gramenzi A, Caputo F, Biselli M. et al. Review article: alcoholic liver disease. –pathophysiological aspects and risk factors. Aliment Pharmacol Ther 2006; 24 (08) 1151-1161
- 50 Makri E, Goulas A, Polyzos SA. Epidemiology, pathogenesis, diagnosis and emerging treatment of nonalcoholic fatty liver disease. Arch Med Res 2021; 52 (01) 25-37
- 51 Nagral A, Bangar M, Menezes S. et al. Gender differences in nonalcoholic fatty liver disease. Euroasian J Hepatogastroenterol 2022; 12 (Suppl. 01) S19-S25
- 52 Balakrishnan M, Patel P, Dunn-Valadez S. et al. Women have a lower risk of nonalcoholic fatty liver disease but a higher risk of progression vs men: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 2021; 19 (01) 61-71.e15
- 53 Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver disease. Gastroenterology 2002; 122 (06) 1649-1657
- 54 Yang JD, Abdelmalek MF, Pang H. et al. Gender and menopause impact severity of fibrosis among patients with nonalcoholic steatohepatitis. Hepatology 2014; 59 (04) 1406-1414
- 55 Hamaguchi M, Kojima T, Ohbora A, Takeda N, Fukui M, Kato T. Aging is a risk factor of nonalcoholic fatty liver disease in premenopausal women. World J Gastroenterol 2012; 18 (03) 237-243
- 56 Ter Horst KW, Gilijamse PW, de Weijer BA. et al. Sexual dimorphism in hepatic, adipose tissue, and peripheral tissue insulin sensitivity in obese humans. Front Endocrinol (Lausanne) 2015; 6: 182
- 57 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
- 58 Conrad N, Misra S, Verbakel JY. et al. Incidence, prevalence, and co-occurrence of autoimmune disorders over time and by age, sex, and socioeconomic status: a population-based cohort study of 22 million individuals in the UK. Lancet 2023; 401 (10391): 1878-1890
- 59 Parikh-Patel A, Gold EB, Worman H, Krivy KE, Gershwin ME. Risk factors for primary biliary cirrhosis in a cohort of patients from the United States. Hepatology 2001; 33 (01) 16-21
- 60 Manns MP, Czaja AJ, Gorham JD. et al; American Association for the Study of Liver Diseases. Diagnosis and management of autoimmune hepatitis. Hepatology 2010; 51 (06) 2193-2213
- 61 Bambha K, Kim WR, Talwalkar J. et al. Incidence, clinical spectrum, and outcomes of primary sclerosing cholangitis in a United States community. Gastroenterology 2003; 125 (05) 1364-1369
- 62 Singh S, Talwalkar JA. Primary sclerosing cholangitis: diagnosis, prognosis, and management. Clin Gastroenterol Hepatol 2013; 11 (08) 898-907
- 63 Weismüller TJ, Trivedi PJ, Bergquist A. et al; International PSC Study Group. Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology 2017; 152 (08) 1975-1984.e8
- 64 Bataller R, Brenner DA. Liver fibrosis. J Clin Invest 2005; 115 (02) 209-218
- 65 Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol 2017; 14 (07) 397-411
- 66 Parkin DM. Global cancer statistics in the year 2000. Lancet Oncol 2001; 2 (09) 533-543
- 67 El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 2007; 132 (07) 2557-2576
- 68 Mittal S, El-Serag HB. Epidemiology of hepatocellular carcinoma: consider the population. J Clin Gastroenterol 2013; 47 Suppl (00) S2-S6
- 69 Zhang XF, Liu Y, Li JH. et al. [Effect of splenectomy on the risk of hepatocellular carcinoma development among patients with liver cirrhosis and portal hypertension: a multi-institutional cohort study]. Zhonghua Wai Ke Za Zhi 2021; 59 (10) 821-828
- 70 Akinyemiju T, Abera S, Ahmed M. et al; Global Burden of Disease Liver Cancer Collaboration. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the Global Burden of Disease Study 2015. JAMA Oncol 2017; 3 (12) 1683-1691
- 71 Zhai Y, Petrowsky H, Hong JC, Busuttil RW, Kupiec-Weglinski JW. Ischaemia-reperfusion injury in liver transplantation. –from bench to bedside. Nat Rev Gastroenterol Hepatol 2013; 10 (02) 79-89
- 72 Chen C, Hu LX, Dong T. et al. Apoptosis and autophagy contribute to gender difference in cardiac ischemia-reperfusion induced injury in rats. Life Sci 2013; 93 (07) 265-270
- 73 Crockett ET, Spielman W, Dowlatshahi S, He J. Sex differences in inflammatory cytokine production in hepatic ischemia-reperfusion. J Inflamm (Lond) 2006; 3: 16
- 74 Hundscheid IHR, Schellekens DHSM, Grootjans J. et al. Females are more resistant to ischemia-reperfusion-induced intestinal injury than males: a human study. Ann Surg 2020; 272 (06) 1070-1079
- 75 Rodríguez F, Nieto-Cerón S, Fenoy FJ. et al. Sex differences in nitrosative stress during renal ischemia. Am J Physiol Regul Integr Comp Physiol 2010; 299 (05) R1387-R1395
- 76 Zuo W, Zhang W, Chen NH. Sexual dimorphism in cerebral ischemia injury. Eur J Pharmacol 2013; 711 (1-3): 73-79
- 77 Michalopoulos GK, Bhushan B. Liver regeneration: biological and pathological mechanisms and implications. Nat Rev Gastroenterol Hepatol 2021; 18 (01) 40-55
- 78 Bizzaro D, Crescenzi M, Di Liddo R. et al. Sex-dependent differences in inflammatory responses during liver regeneration in a murine model of acute liver injury. Clin Sci (Lond) 2018; 132 (02) 255-272
- 79 Sayaf K, Zanotto I, Gabbia D. et al. Sex drives functional changes in the progression and regression of liver fibrosis. Int J Mol Sci 2023; 24 (22) 24
- 80 Birrer DL, Linecker M, López-López V. et al. Sex disparities in outcomes following major liver surgery: new powers of estrogen?. Ann Surg 2022; 276 (05) 875-881
- 81 Zhu M, Li Y, Shen Q, Gong Z, Liu D. Sex hormone receptors, calcium-binding protein and Yap1 signaling regulate sex-dependent liver cell proliferation following partial hepatectomy. Dis Model Mech 2024; 17 (10) 17
- 82 Guy J, Peters MG. Liver disease in women: the influence of gender on epidemiology, natural history, and patient outcomes. Gastroenterol Hepatol (N Y) 2013; 9 (10) 633-639
- 83 Pietrangelo A. Genetics, genetic testing, and management of hemochromatosis: 15 years since hepcidin. Gastroenterology 2015; 149 (05) 1240-1251.e4
- 84 Allen KJ, Gurrin LC, Constantine CC. et al. Iron-overload-related disease in HFE hereditary hemochromatosis. N Engl J Med 2008; 358 (03) 221-230
- 85 Cooper KM, Delk M, Devuni D, Sarkar M. Sex differences in chronic liver disease and benign liver lesions. JHEP Rep Innov Hepatol 2023; 5 (11) 100870
- 86 Shin N, Kim YH, Xu H. et al. Redefining Budd-Chiari syndrome: a systematic review. World J Hepatol 2016; 8 (16) 691-702
- 87 Buzzetti E, Parikh PM, Gerussi A, Tsochatzis E. Gender differences in liver disease and the drug-dose gender gap. Pharmacol Res 2017; 120: 97-108
- 88 Sutti S, Tacke F. Liver inflammation and regeneration in drug-induced liver injury: sex matters!. Clin Sci (Lond) 2018; 132 (05) 609-613
- 89 Chen TS, Doong ML, Chang FY, Lee SD, Wang PS. Effects of sex steroid hormones on gastric emptying and gastrointestinal transit in rats. Am J Physiol 1995; 268 (1 Pt 1): G171-G176
- 90 Yang L, Li Y, Hong H. et al. Sex differences in the expression of drug-metabolizing and transporter genes in human liver. J Drug Metab Toxicol 2012; 3 (03) 1000119
- 91 Lamba V, Lamba J, Yasuda K. et al. Hepatic CYP2B6 expression: gender and ethnic differences and relationship to CYP2B6 genotype and CAR (constitutive androstane receptor) expression. J Pharmacol Exp Ther 2003; 307 (03) 906-922
- 92 Mauvais-Jarvis F, Berthold HK, Campesi I. et al. Sex- and gender-based pharmacological response to drugs. Pharmacol Rev 2021; 73 (02) 730-762
- 93 Lai KP, Wong MH, Wong CK. Modulation of AhR-mediated CYP1A1 mRNA and EROD activities by 17beta-estradiol and dexamethasone in TCDD-induced H411E cells. Toxicol Sci 2004; 78 (01) 41-49
- 94 Koyuncuoğlu T, Yıldırım A, Dertsiz EK, Yüksel M, Ercan F, Yeğen BÇ. Estrogen receptor agonists protect against acetaminophen-induced hepatorenal toxicity in rats. Life Sci 2020; 263: 118561
- 95 Win S, Min RW, Chen CQ. et al. Expression of mitochondrial membrane-linked SAB determines severity of sex-dependent acute liver injury. J Clin Invest 2019; 129 (12) 5278-5293
- 96 Frezza M, di Padova C, Pozzato G, Terpin M, Baraona E, Lieber CS. High blood alcohol levels in women. The role of decreased gastric alcohol dehydrogenase activity and first-pass metabolism. N Engl J Med 1990; 322 (02) 95-99
- 97 Thurman RG. II. Alcoholic liver injury involves activation of Kupffer cells by endotoxin. Am J Physiol 1998; 275 (04) G605-G611
- 98 Yin M, Ikejima K, Wheeler MD. et al. Estrogen is involved in early alcohol-induced liver injury in a rat enteral feeding model. Hepatology 2000; 31 (01) 117-123
- 99 Ikejima K, Enomoto N, Iimuro Y. et al. Estrogen increases sensitivity of hepatic Kupffer cells to endotoxin. Am J Physiol 1998; 274 (04) G669-G676
- 100 Mishra P, Davies DA, Albensi BC. The interaction between NF-κB and estrogen in Alzheimer's disease. Mol Neurobiol 2023; 60 (03) 1515-1526
- 101 Purawarga Matada GS, Dhiwar PS, Abbas N. et al. Molecular docking and molecular dynamic studies: screening of phytochemicals against EGFR, HER2, estrogen and NF-KB receptors for their potential use in breast cancer. J Biomol Struct Dyn 2022; 40 (13) 6183-6192
- 102 Mezey E. Influence of sex hormones on alcohol metabolism. Alcohol Clin Exp Res 2000; 24 (04) 421
- 103 Ichiro S, Kamochi M, Yoshikawa H, Nakayama Y. Gender Difference in Alcoholic Liver Disease. IntechOpen; 2012
- 104 Kim DK, Kim YH, Jang HH. et al. Estrogen-related receptor γ controls hepatic CB1 receptor-mediated CYP2E1 expression and oxidative liver injury by alcohol. Gut 2013; 62 (07) 1044-1054
- 105 Järveläinen HA, Lukkari TA, Heinaro S, Sippel H, Lindros KO. The antiestrogen toremifene protects against alcoholic liver injury in female rats. J Hepatol 2001; 35 (01) 46-52
- 106 Fang C, Lindros KO, Badger TM, Ronis MJ, Ingelman-Sundberg M. Zonated expression of cytokines in rat liver: effect of chronic ethanol and the cytochrome P450 2E1 inhibitor, chlormethiazole. Hepatology 1998; 27 (05) 1304-1310
- 107 Gouillon Z, Lucas D, Li J. et al. Inhibition of ethanol-induced liver disease in the intragastric feeding rat model by chlormethiazole. Proc Soc Exp Biol Med 2000; 224 (04) 302-308
- 108 Zhu L, Brown WC, Cai Q. et al. Estrogen treatment after ovariectomy protects against fatty liver and may improve pathway-selective insulin resistance. Diabetes 2013; 62 (02) 424-434
- 109 Skubic C, Drakulić Ž, Rozman D. Personalized therapy when tackling nonalcoholic fatty liver disease: a focus on sex, genes, and drugs. Expert Opin Drug Metab Toxicol 2018; 14 (08) 831-841
- 110 Schiffrin M, Winkler C, Quignodon L. et al. Sex dimorphism of nonalcoholic fatty liver disease (NAFLD) in Pparg-null mice. Int J Mol Sci 2021; 22 (18) 22
- 111 Shu Z, Zhang G, Zhu X, Xiong W. Estrogen receptor α mediated M1/M2 macrophages polarization plays a critical role in NASH of female mice. Biochem Biophys Res Commun 2022; 596: 63-70
- 112 Yang M, Liu Q, Huang T. et al. Dysfunction of estrogen-related receptor alpha-dependent hepatic VLDL secretion contributes to sex disparity in NAFLD/NASH development. Theranostics 2020; 10 (24) 10874-10891
- 113 Della Torre S. Beyond the X factor: relevance of sex hormones in NAFLD pathophysiology. Cells 2021; 10 (09) 10
- 114 Sharma G, Prossnitz ER. G-protein-coupled estrogen receptor (GPER) and sex-specific metabolic homeostasis. Adv Exp Med Biol 2017; 1043: 427-453
- 115 Meoli L, Isensee J, Zazzu V. et al. Sex- and age-dependent effects of Gpr30 genetic deletion on the metabolic and cardiovascular profiles of diet-induced obese mice. Gene 2014; 540 (02) 210-216
- 116 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 (18) 14851-14862
- 117 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 (06) 1466-1475
- 118 Lee C, Kim J, Han J. et al. Formyl peptide receptor 2 determines sex-specific differences in the progression of nonalcoholic fatty liver disease and steatohepatitis. Nat Commun 2022; 13 (01) 578
- 119 Wu J, Yao XY, Shi RX, Liu SF, Wang XY. A potential link between polycystic ovary syndrome and non-alcoholic fatty liver disease: an update meta-analysis. Reprod Health 2018; 15 (01) 77
- 120 Sarkar M, Yates K, Suzuki A. et al. Low testosterone is associated with nonalcoholic steatohepatitis and fibrosis severity in men. Clin Gastroenterol Hepatol 2021; 19 (02) 400-402.e2
- 121 Jaruvongvanich V, Sanguankeo A, Riangwiwat T, Upala S. Testosterone, sex hormone-binding globulin and nonalcoholic fatty liver disease: a systematic review and meta-analysis. Ann Hepatol 2017; 16 (03) 382-394
- 122 Jia Y, Yee JK, Wang C. et al. Testosterone protects high-fat/low-carbohydrate diet-induced nonalcoholic fatty liver disease in castrated male rats mainly via modulating endoplasmic reticulum stress. Am J Physiol Endocrinol Metab 2018; 314 (04) E366-E376
- 123 Nikolaenko L, Jia Y, Wang C. et al. Testosterone replacement ameliorates nonalcoholic fatty liver disease in castrated male rats. Endocrinology 2014; 155 (02) 417-428
- 124 Schwinge D, Schramm C. Sex-related factors in autoimmune liver diseases. Semin Immunopathol 2019; 41 (02) 165-175
- 125 Smyk DS, Rigopoulou EI, Pares A. et al. Sex differences associated with primary biliary cirrhosis. Clin Dev Immunol 2012; 2012: 610504
- 126 Yasuda M, Shimizu I, Shiba M, Ito S. Suppressive effects of estradiol on dimethylnitrosamine-induced fibrosis of the liver in rats. Hepatology 1999; 29 (03) 719-727
- 127 Xu J-W, Gong J, Chang X-M. et al. Estrogen reduces CCL4- induced liver fibrosis in rats. World J Gastroenterol 2002; 8 (05) 883-887
- 128 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 (07) 897-907
- 129 Zhou YJ, Yin DM, Chen HS, Shi JH, Sha BX, Wang X. Inhibitory effects of idoxifene on hepatic fibrosis in rats. Acta Pharmacol Sin 2005; 26 (05) 581-586
- 130 Shimizu I, Ito S. Protection of estrogens against the progression of chronic liver disease. Hepatol Res 2007; 37 (04) 239-247
- 131 Shimizu I, Kohno N, Tamaki K. et al. Female hepatology: favorable role of estrogen in chronic liver disease with hepatitis B virus infection. World J Gastroenterol 2007; 13 (32) 4295-4305
- 132 Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 2017; 121: 27-42
- 133 Zhou Y, Shimizu I, Lu G. et al. Hepatic stellate cells contain the functional estrogen receptor beta but not the estrogen receptor alpha in male and female rats. Biochem Biophys Res Commun 2001; 286 (05) 1059-1065
- 134 Zhang B, Zhang CG, Ji LH, Zhao G, Wu ZY. Estrogen receptor β selective agonist ameliorates liver cirrhosis in rats by inhibiting the activation and proliferation of hepatic stellate cells. J Gastroenterol Hepatol 2018; 33 (03) 747-755
- 135 Zhang K, Lin L, Zhu Y, Zhang N, Zhou M, Li Y. Saikosaponin d alleviates liver fibrosis by negatively regulating the ROS/NLRP3 inflammasome through activating the ERβ pathway. Front Pharmacol 2022; 13: 894981
- 136 Inagaki Y, Okazaki I. Emerging insights into transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut 2007; 56 (02) 284-292
- 137 McCarty MF, Barroso-Aranda J, Contreras F. Genistein and phycocyanobilin may prevent hepatic fibrosis by suppressing proliferation and activation of hepatic stellate cells. Med Hypotheses 2009; 72 (03) 330-332
- 138 Ganai AA, Husain M. Genistein attenuates D-GalN induced liver fibrosis/chronic liver damage in rats by blocking the TGF-β/Smad signaling pathways. Chem Biol Interact 2017; 261: 80-85
- 139 Evans MJ, Lai K, Shaw LJ, Harnish DC, Chadwick CC. Estrogen receptor alpha inhibits IL-1beta induction of gene expression in the mouse liver. Endocrinology 2002; 143 (07) 2559-2570
- 140 Ma X, Zhou Y, Qiao B. et al. Androgen aggravates liver fibrosis by activation of NLRP3 inflammasome in CCl4-induced liver injury mouse model. Am J Physiol Endocrinol Metab 2020; 318 (05) E817-E829
- 141 Wang S-H, Yeh S-H, Lin W-H, Wang HY, Chen DS, Chen PJ. Identification of androgen response elements in the enhancer I of hepatitis B virus: a mechanism for sex disparity in chronic hepatitis B. Hepatology 2009; 50 (05) 1392-1402
- 142 Wu M-H, Ma W-L, Hsu C-L. et al. Androgen receptor promotes hepatitis B virus-induced hepatocarcinogenesis through modulation of hepatitis B virus RNA transcription. Sci Transl Med 2010; 2 (32) 32ra35
- 143 Cha MY, Kim CM, Park YM, Ryu WS. Hepatitis B virus X protein is essential for the activation of Wnt/beta-catenin signaling in hepatoma cells. Hepatology 2004; 39 (06) 1683-1693
- 144 Ding Q, Xia W, Liu J-C. et al. Erk associates with and primes GSK-3beta for its inactivation resulting in upregulation of beta-catenin. Mol Cell 2005; 19 (02) 159-170
- 145 Bouchard MJ, Wang L, Schneider RJ. Activation of focal adhesion kinase by hepatitis B virus HBx protein: multiple functions in viral replication. J Virol 2006; 80 (09) 4406-4414
- 146 Yeh S-H, Chen P-J. Gender disparity of hepatocellular carcinoma: the roles of sex hormones. Oncology 2010; 78 (Suppl. 01) 172-179
- 147 Chen P-J, Yeh S-H, Liu W-H. et al. Androgen pathway stimulates microRNA-216a transcription to suppress the tumor suppressor in lung cancer-1 gene in early hepatocarcinogenesis. Hepatology 2012; 56 (02) 632-643
- 148 Feng H, Cheng ASL, Tsang DP. et al. Cell cycle-related kinase is a direct androgen receptor-regulated gene that drives β-catenin/T cell factor-dependent hepatocarcinogenesis. J Clin Invest 2011; 121 (08) 3159-3175
- 149 Llovet JM. Updated treatment approach to hepatocellular carcinoma. J Gastroenterol 2005; 40 (03) 225-235
- 150 Zhang H, Spencer K, Burley SK, Zheng XFS. Toward improving androgen receptor-targeted therapies in male-dominant hepatocellular carcinoma. Drug Discov Today 2021; 26 (06) 1539-1546
- 151 Ji F, Zhang J, Liu N. et al. Blocking hepatocarcinogenesis by a cytochrome P450 family member with female-preferential expression. Gut 2022; 71 (11) 2313-2324
- 152 Naugler WE, Sakurai T, Kim S. et al. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 2007; 317 (5834): 121-124
- 153 Hou J, Xu J, Jiang R. et al. Estrogen-sensitive PTPRO expression represses hepatocellular carcinoma progression by control of STAT3. Hepatology 2013; 57 (02) 678-688
- 154 Huang F-Y, Wong DK-H, Seto W-K, Lai C-L, Yuen M-F. Estradiol induces apoptosis via activation of miRNA-23a and p53: implication for gender difference in liver cancer development. Oncotarget 2015; 6 (33) 34941-34952
- 155 Li C-L, Yeh K-H, Liu W-H. et al. Elevated p53 promotes the processing of miR-18a to decrease estrogen receptor-α in female hepatocellular carcinoma. Int J Cancer 2015; 136 (04) 761-770
- 156 Wei Q, Guo P, Mu K. et al. Estrogen suppresses hepatocellular carcinoma cells through ERβ-mediated upregulation of the NLRP3 inflammasome. Lab Invest 2015; 95 (07) 804-816
- 157 Chow PK, Tai BC, Tan CK. et al; Asian-Pacific Hepatocellular Carcinoma Trials Group. High-dose tamoxifen in the treatment of inoperable hepatocellular carcinoma: a multicenter randomized controlled trial. Hepatology 2002; 36 (05) 1221-1226
- 158 Riestra S, Rodriguez M, Delgado M. et al. Tamoxifen does not improve survival of patients with advanced hepatocellular carcinoma. J Clin Gastroenterol 1998; 26 (03) 200-203
- 159 Shimizu I. CLIP Group (Cancer of the Liver Italian Programme). Tamoxifen in treatment of hepatocellular carcinoma: a randomised controlled trial. Lancet 1998; 352 (9121): 17-20
- 160 Barbare JC, Bouché O, Bonnetain F. et al. Randomized controlled trial of tamoxifen in advanced hepatocellular carcinoma. J Clin Oncol 2005; 23 (19) 4338-4346
- 161 Harada H, Pavlick KP, Hines IN. et al. Selected contribution: effects of gender on reduced-size liver ischemia and reperfusion injury. J Appl Physiol 2001; 91 (06) 2816-2822
- 162 Harada H, Pavlick KP, Hines IN. et al. Sexual dimorphism in reduced-size liver ischemia and reperfusion injury in mice: role of endothelial cell nitric oxide synthase. Proc Natl Acad Sci U S A 2003; 100 (02) 739-744
- 163 Harada H, Bharwani S, Pavlick KP, Korach KS, Grisham MB. Estrogen receptor-alpha, sexual dimorphism and reduced-size liver ischemia and reperfusion injury in mice. Pediatr Res 2004; 55 (03) 450-456
- 164 Fels JA, Manfredi G. Sex differences in ischemia/reperfusion injury: the role of mitochondrial permeability transition. Neurochem Res 2019; 44 (10) 2336-2345
- 165 Huang C, Gu H, Zhang W, Herrmann JL, Wang M. Testosterone-down-regulated Akt pathway during cardiac ischemia/reperfusion: a mechanism involving BAD, Bcl-2 and FOXO3a. J Surg Res 2010; 164 (01) e1-e11
- 166 Wang M, Tsai BM, Kher A, Baker LB, Wairiuko GM, Meldrum DR. Role of endogenous testosterone in myocardial proinflammatory and proapoptotic signaling after acute ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2005; 288 (01) H221-H226
- 167 Liddle C, Farrell GC. Role of the oestrogen receptor in liver regeneration in the male rat. J Gastroenterol Hepatol 1993; 8 (06) 524-529
- 168 Francavilla A, di Leo A, Eagon PK. et al. Regenerating rat liver: correlations between estrogen receptor localization and deoxyribonucleic acid synthesis. Gastroenterology 1984; 86 (03) 552-557
- 169 Fisher B, Gunduz N, Saffer EA, Zheng S. Relation of estrogen and its receptor to rat liver growth and regeneration. Cancer Res 1984; 44 (06) 2410-2415
- 170 Francavilla A, Eagon PK, DiLeo A. et al. Sex hormone-related functions in regenerating male rat liver. Gastroenterology 1986; 91 (05) 1263-1270
- 171 Kao TL, Kuan YP, Cheng WC. et al. Estrogen receptors orchestrate cell growth and differentiation to facilitate liver regeneration. Theranostics 2018; 8 (10) 2672-2682
- 172 Chaturantabut S, Shwartz A, Evason KJ. et al. Estrogen activation of G-Protein-coupled estrogen receptor 1 regulates phosphoinositide 3-kinase and mTOR signaling to promote liver growth in zebrafish and proliferation of human hepatocytes. Gastroenterology 2019; 156 (06) 1788-1804.e13
- 173 Yamaguchi M, Yu L, Nazmy El-Assal O. et al. Androgen metabolism in regenerating liver of male rats: evidence for active uptake and utilization of testosterone. Hepatol Res 2001; 20 (01) 114-127
- 174 Du L, Bayir H, Lai Y. et al. Innate gender-based proclivity in response to cytotoxicity and programmed cell death pathway. J Biol Chem 2004; 279 (37) 38563-38570
- 175 Barker JM, Torregrossa MM, Arnold AP, Taylor JR. Dissociation of genetic and hormonal influences on sex differences in alcoholism-related behaviors. J Neurosci 2010; 30 (27) 9140-9144
- 176 Chen X, McClusky R, Chen J. et al. The number of x chromosomes causes sex differences in adiposity in mice. PLoS Genet 2012; 8 (05) e1002709
- 177 Romeo S. Notch and nonalcoholic fatty liver and fibrosis. N Engl J Med 2019; 380 (07) 681-683
- 178 Zhu C, Kim K, Wang X. et al. Hepatocyte Notch activation induces liver fibrosis in nonalcoholic steatohepatitis. Sci Transl Med 2018; 10 (468) 10
- 179 Hanley KP, Oakley F, Sugden S, Wilson DI, Mann DA, Hanley NA. Ectopic SOX9 mediates extracellular matrix deposition characteristic of organ fibrosis. J Biol Chem 2008; 283 (20) 14063-14071
- 180 Pritchett J, Harvey E, Athwal V. et al. Osteopontin is a novel downstream target of SOX9 with diagnostic implications for progression of liver fibrosis in humans. Hepatology 2012; 56 (03) 1108-1116
- 181 Athwal VS, Pritchett J, Llewellyn J. et al. SOX9 predicts progression toward cirrhosis in patients while its loss protects against liver fibrosis. EMBO Mol Med 2017; 9 (12) 1696-1710
- 182 Fu M, Yin W, Zhang W, Zhu Y, Ni H, Gong L. MicroRNA-15a inhibits hepatic stellate cell activation and proliferation via targeting SRY-box transcription factor 9. Bioengineered 2022; 13 (05) 13011-13020
- 183 Wang C, Deng J, Deng H. et al. A novel Sox9/lncRNA H19 axis contributes to hepatocyte death and liver fibrosis. Toxicol Sci 2020; 177 (01) 214-225
- 184 Jiang H-L, Xu D, Yu H. et al. DAX-1 inhibits hepatocellular carcinoma proliferation by inhibiting β-catenin transcriptional activity. Cell Physiol Biochem 2014; 34: 734-742
- 185 Motawi TMK, Sadik NAH, Sabry D, Shahin NN, Fahim SA. rs2267531, a promoter SNP within glypican-3 gene in the X chromosome, is associated with hepatocellular carcinoma in Egyptians. Sci Rep 2019; 9 (01) 6868
- 186 Liao YL, Sun YM, Chau GY. et al. Identification of SOX4 target genes using phylogenetic footprinting-based prediction from expression microarrays suggests that overexpression of SOX4 potentiates metastasis in hepatocellular carcinoma. Oncogene 2008; 27 (42) 5578-5589
- 187 Wang W, Zhang J, Zhan X. et al. SOX4 is associated with poor prognosis in cholangiocarcinoma. Biochem Biophys Res Commun 2014; 452 (03) 614-621
- 188 Kawaguchi Y. Sox9 and programming of liver and pancreatic progenitors. J Clin Invest 2013; 123 (05) 1881-1886
- 189 Tsai C-N, Yu S-C, Lee C-W. et al. SOX4 activates CXCL12 in hepatocellular carcinoma cells to modulate endothelial cell migration and angiogenesis in vivo. Oncogene 2020; 39 (24) 4695-4710
- 190 Tsuei D-J, Hsu H-C, Lee P-H. et al. RBMY, a male germ cell-specific RNA-binding protein, activated in human liver cancers and transforms rodent fibroblasts. Oncogene 2004; 23 (34) 5815-5822
- 191 Chua H-H, Tsuei D-J, Lee P-H. et al. RBMY, a novel inhibitor of glycogen synthase kinase 3β, increases tumor stemness and predicts poor prognosis of hepatocellular carcinoma. Hepatology 2015; 62 (05) 1480-1496
- 192 Li S, Mo C, Huang S. et al. Over-expressed Testis-specific Protein Y-encoded 1 as a novel biomarker for male hepatocellular carcinoma. PLoS One 2014; 9 (02) e89219
- 193 Fan XD, Zheng HB, Fan XS, Lu S. Increase of SOX9 promotes hepatic ischemia/reperfusion (IR) injury by activating TGF-β1. Biochem Biophys Res Commun 2018; 503 (01) 215-221
- 194 Han X, Wang Y, Pu W. et al. Lineage tracing reveals the bipotency of SOX9+ hepatocytes during liver regeneration. Stem Cell Reports 2019; 12 (03) 624-638
- 195 Yovchev MI, Locker J, Oertel M. Biliary fibrosis drives liver repopulation and phenotype transition of transplanted hepatocytes. J Hepatol 2016; 64 (06) 1348-1357
- 196 Shao C, Jing Y, Zhao S. et al. LPS/Bcl3/YAP1 signaling promotes Sox9+HNF4α+ hepatocyte-mediated liver regeneration after hepatectomy. Cell Death Dis 2022; 13 (03) 277
- 197 Gong S, Zeng Y, Wang Z. et al. Intestinal deguelin drives resistance to acetaminophen-induced hepatotoxicity in female mice. Gut Microbes 2024; 16 (01) 2404138
- 198 Corpechot C, Chrétien Y, Chazouillères O, Poupon R. Demographic, lifestyle, medical and familial factors associated with primary biliary cirrhosis. J Hepatol 2010; 53 (01) 162-169
- 199 Gershwin ME, Selmi C, Worman HJ. et al; USA PBC Epidemiology Group. Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients. Hepatology 2005; 42 (05) 1194-1202
- 200 Prince MI, Ducker SJ, James OF. Case-control studies of risk factors for primary biliary cirrhosis in two United Kingdom populations. Gut 2010; 59 (04) 508-512
- 201 Karlsen TH, Lammert F, Thompson RJ. Genetics of liver disease: from pathophysiology to clinical practice. J Hepatol 2015; 62 (1, Suppl): S6-S14
- 202 Greten TF. Gender disparity in HCC: is it the fat and not the sex?. J Exp Med 2019; 216 (05) 1014-1015
- 203 Manieri E, Herrera-Melle L, Mora A. et al. Adiponectin accounts for gender differences in hepatocellular carcinoma incidence. J Exp Med 2019; 216 (05) 1108-1119
- 204 Yang Q, Yan C, Yin C, Gong Z. Serotonin activated hepatic stellate cells contribute to sex disparity in hepatocellular carcinoma. Cell Mol Gastroenterol Hepatol 2017; 3 (03) 484-499
- 205 Hu Z, Jepps TA, Zhou L, Liu J, Li M, Abbott GW. Kcne4 deletion sex dependently inhibits the RISK pathway response and exacerbates hepatic ischemia-reperfusion injury in mice. Am J Physiol Regul Integr Comp Physiol 2019; 316 (05) R552-R562
- 206 Shang D, Wang L, Klionsky DJ, Cheng H, Zhou R. Sex differences in autophagy-mediated diseases: toward precision medicine. Autophagy 2021; 17 (05) 1065-1076
- 207 Campana L, Esser H, Huch M, Forbes S. Liver regeneration and inflammation: from fundamental science to clinical applications. Nat Rev Mol Cell Biol 2021; 22 (09) 608-624
- 208 Shimizu I. Accounting for sex and gender makes for better science. Nature 2020; 588 (7837): 196
- 209 Hunt L, Nielsen MW, Schiebinger L. A framework for sex, gender, and diversity analysis in research. Science 2022; 377 (6614): 1492-1495
- 210 Peters SAE, Woodward M. A roadmap for sex- and gender-disaggregated health research. BMC Med 2023; 21 (01) 354
- 211 de Beaurepaire R. Suppression of alcohol dependence using baclofen: a 2-year observational study of 100 patients. Front Psychiatry 2012; 3: 103
- 212 Crabb DW, Im GY, Szabo G, Mellinger JL, Lucey MR. Diagnosis and treatment of alcohol-associated liver diseases: 2019 Practice Guidance From the American Association for the Study of Liver Diseases. Hepatology 2020; 71 (01) 306-333
- 213 Cotter TG, Rinella M. Nonalcoholic fatty liver disease 2020: the state of the disease. Gastroenterology 2020; 158 (07) 1851-1864
- 214 Li N, Cui C, Xu J, Mi M, Wang J, Qin Y. Quercetin intervention reduced hepatic fat deposition in patients with nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled crossover clinical trial. Am J Clin Nutr 2024; 120 (03) 507-517
- 215 Yan H, Wu W, Chang X. et al. Gender differences in the efficacy of pioglitazone treatment in nonalcoholic fatty liver disease patients with abnormal glucose metabolism. Biol Sex Differ 2021; 12 (01) 1
- 216 Carbone M, Mells GF, Pells G. et al; UK PBC Consortium. Sex and age are determinants of the clinical phenotype of primary biliary cirrhosis and response to ursodeoxycholic acid. Gastroenterology 2013; 144 (03) 560-569.e7 , quiz e13–e14
- 217 Carey EJ, Ali AH, Lindor KD. Primary biliary cirrhosis. Lancet 2015; 386 (10003): 1565-1575
- 218 Hirschfield GM, Dyson JK, Alexander GJM. et al. The British Society of Gastroenterology/UK-PBC primary biliary cholangitis treatment and management guidelines. Gut 2018; 67 (09) 1568-1594
- 219 Muratori L, Lohse AW, Lenzi M. Diagnosis and management of autoimmune hepatitis. BMJ 2023; 380: e070201
- 220 Czaja AJ, Donaldson PT. Gender effects and synergisms with histocompatibility leukocyte antigens in type 1 autoimmune hepatitis. Am J Gastroenterol 2002; 97 (08) 2051-2057
- 221 Al-Chalabi T, Underhill JA, Portmann BC, McFarlane IG, Heneghan MA. Impact of gender on the long-term outcome and survival of patients with autoimmune hepatitis. J Hepatol 2008; 48 (01) 140-147
- 222 Kim MN, Han JW, An J. et al; Korean Association for the Study of the Liver (KASL). KASL clinical practice guidelines for noninvasive tests to assess liver fibrosis in chronic liver disease. Clin Mol Hepatol 2024; 30 (Suppl): S5-S105
- 223 Shimizu I. Chinese Society of Hepatology Chinese Medical Association, Chinese Society of Gastroenterology Chinese Medical Association, Chinese Society of Infectious Diseases, Chinese Medical Association. [Consensus on the diagnosis and therapy of hepatic fibrosis in]. Zhonghua Gan Zang Bing Za Zhi 2019; 27 (09) 657-667
- 224 Kammula AV, Schäffer AA, Rajagopal PS, Kurzrock R, Ruppin E. Outcome differences by sex in oncology clinical trials. Nat Commun 2024; 15 (01) 2608
- 225
D'Abbondanza M,
Ministrini S,
Pucci G.
et al.
Very Low-Carbohydrate Ketogenic Diet for the Treatment of Severe Obesity and Associated
Non-Alcoholic Fatty Liver Disease: The Role of Sex Differences. Nutrients 2020;12
Address for correspondence
Publication History
Accepted Manuscript online:
14 January 2025
Article published online:
11 February 2025
© 2025. Thieme. All rights reserved.
Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA
-
References
- 1 Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E, Kamath PS. Global burden of liver disease: 2023 update. J Hepatol 2023; 79 (02) 516-537
- 2 Heron M. Deaths: leading causes for 2017. Natl Vital Stat Rep 2019; 68 (06) 1-77
- 3 Woitowich NC, Woodruff TK. Opinion: Research community needs to better appreciate the value of sex-based research. Proc Natl Acad Sci U S A 2019; 116 (15) 7154-7156
- 4 Mauvais-Jarvis F, Bairey Merz N, Barnes PJ. et al. Sex and gender: modifiers of health, disease, and medicine. Lancet 2020; 396 (10250): 565-582
- 5 Roy AK, Chatterjee B. Sexual dimorphism in the liver. Annu Rev Physiol 1983; 45: 37-50
- 6 Yang X, Schadt EE, Wang S. et al. Tissue-specific expression and regulation of sexually dimorphic genes in mice. Genome Res 2006; 16 (08) 995-1004
- 7 Man S, Deng Y, Ma Y. et al. Prevalence of liver steatosis and fibrosis in the general population and various high-risk populations: a nationwide study with 5.7 million adults in China. Gastroenterology 2023; 165 (04) 1025-1040
- 8 Lefebvre P, Staels B. Hepatic sexual dimorphism - implications for non-alcoholic fatty liver disease. Nat Rev Endocrinol 2021; 17 (11) 662-670
- 9 Caballería L, Pera G, Arteaga I. et al. High prevalence of liver fibrosis among european adults with unknown liver disease: a population-based study. Clin Gastroenterol Hepatol 2018; 16 (07) 1138-1145.e5
- 10 Lonardo A, Nascimbeni F, Ballestri S. et al. Sex differences in nonalcoholic fatty liver disease: state of the art and identification of research gaps. Hepatology 2019; 70 (04) 1457-1469
- 11 Poynard T, Bedossa P, Opolon P. Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 1997; 349 (9055): 825-832
- 12 Floreani A, Bizzaro D, Shalaby S, Taliani G, Burra P. Special Interest Group Gender in Hepatology of the Italian Association for the Study of the Liver (AISF). Sex disparity and drug-induced liver injury. Dig Liver Dis 2023; 55 (01) 21-28
- 13 Szabo G. Women and alcoholic liver disease - warning of a silent danger. Nat Rev Gastroenterol Hepatol 2018; 15 (05) 253-254
- 14 Kezer CA, Simonetto DA, Shah VH. Sex differences in alcohol consumption and alcohol-associated liver disease. Mayo Clin Proc 2021; 96 (04) 1006-1016
- 15 Invernizzi F, Cilla M, Trapani S. et al; Special Interest Group Gender in Hepatology of the Italian Association for the Study of the Liver (AISF). Gender and autoimmune liver diseases: relevant aspects in clinical practice. J Pers Med 2022; 12 (06) 12
- 16 Barone M, Margiotta M, Scavo MP. et al. Possible involvement of androgen receptor alterations in hepatocarcinogenesis. Dig Liver Dis 2009; 41 (09) 665-670
- 17 Besse-Patin A, Léveillé M, Oropeza D, Nguyen BN, Prat A, Estall JL. Estrogen signals through peroxisome proliferator-activated receptor-γ coactivator 1α to reduce oxidative damage associated with diet-induced fatty liver disease. Gastroenterology 2017; 152 (01) 243-256
- 18 Li W, Li D, Sun L, Li Z, Yu L, Wu S. The protective effects of estrogen on hepatic ischemia-reperfusion injury in rats by downregulating the Ang II/AT1R pathway. Biochem Biophys Res Commun 2018; 503 (04) 2543-2548
- 19 Wang SH, Yeh SH, Lin WH. et al. Estrogen receptor α represses transcription of HBV genes via interaction with hepatocyte nuclear factor 4α. Gastroenterology 2012; 142 (04) 989-998.e4
- 20 Klair JS, Yang JD, Abdelmalek MF. et al; Nonalcoholic Steatohepatitis Clinical Research Network. A longer duration of estrogen deficiency increases fibrosis risk among postmenopausal women with nonalcoholic fatty liver disease. Hepatology 2016; 64 (01) 85-91
- 21 Nuermaimaiti A, Chang L, Yan Y. et al. The role of sex hormones and receptors in HBV infection and development of HBV-related HCC. J Med Virol 2023; 95 (12) e29298
- 22 Liu C, Ren YF, Dong J. et al. Activation of SRY accounts for male-specific hepatocarcinogenesis: Implication in gender disparity of hepatocellular carcinoma. Cancer Lett 2017; 410: 20-31
- 23 Murakami S, Chishima S, Uemoto H. et al. The male-specific factor Sry harbors an oncogenic function. Oncogene 2014; 33 (23) 2978-2986
- 24 Murakami S, Ninomiya W, Sakamoto E, Shibata T, Akiyama H, Tashiro F. SRY and OCT4 are required for the acquisition of cancer stem cell-like properties and are potential differentiation therapy targets. Stem Cells 2015; 33 (09) 2652-2663
- 25 Tsuei DJ, Lee PH, Peng HY. et al. Male germ cell-specific RNA binding protein RBMY: a new oncogene explaining male predominance in liver cancer. PLoS One 2011; 6 (11) e26948
- 26 Yin YH, Li YY, Qiao H. et al. TSPY is a cancer testis antigen expressed in human hepatocellular carcinoma. Br J Cancer 2005; 93 (04) 458-463
- 27 Dong J, Ke MY, Wu XN. et al. SRY is a key mediator of sexual dimorphism in hepatic ischemia/reperfusion injury. Ann Surg 2022; 276 (02) 345-356
- 28 Wu XN, Wang MZ, Zhang N. et al. Sex-determining region Y gene promotes liver fibrosis and accounts for sexual dimorphism in its pathophysiology. J Hepatol 2024; 80 (06) 928-940
- 29 Donnelly MC, Davidson JS, Martin K, Baird A, Hayes PC, Simpson KJ. Acute liver failure in Scotland: changes in aetiology and outcomes over time (the Scottish Look-Back Study). Aliment Pharmacol Ther 2017; 45 (06) 833-843
- 30 Weaver RJ, Blomme EA, Chadwick AE. et al. Managing the challenge of drug-induced liver injury: a roadmap for the development and deployment of preclinical predictive models. Nat Rev Drug Discov 2020; 19 (02) 131-148
- 31 Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 2005; 4 (06) 489-499
- 32 Chalasani N, Bonkovsky HL, Fontana R. et al; United States Drug Induced Liver Injury Network. Features and outcomes of 899 patients with drug-induced liver injury: The DILIN Prospective Study. Gastroenterology 2015; 148 (07) 1340-52.e7
- 33 Amacher DE. Female gender as a susceptibility factor for drug-induced liver injury. Hum Exp Toxicol 2014; 33 (09) 928-939
- 34 Shen T, Liu Y, Shang J. et al. Incidence and etiology of drug-induced liver injury in Mainland China. Gastroenterology 2019; 156 (08) 2230-2241.e11
- 35 Sgro C, Clinard F, Ouazir K. et al. Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology 2002; 36 (02) 451-455
- 36 Lucena MI, Andrade RJ, Kaplowitz N. et al; Spanish Group for the Study of Drug-Induced Liver Disease. Phenotypic characterization of idiosyncratic drug-induced liver injury: the influence of age and sex. Hepatology 2009; 49 (06) 2001-2009
- 37 Andrade RJ, Lucena MI, Fernández MC. et al; Spanish Group for the Study of Drug-Induced Liver Disease. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology 2005; 129 (02) 512-521
- 38 Reuben A, Koch DG, Lee WM. Acute Liver Failure Study Group. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology 2010; 52 (06) 2065-2076
- 39 Dai G, He L, Chou N, Wan YJ. Acetaminophen metabolism does not contribute to gender difference in its hepatotoxicity in mouse. Toxicol Sci 2006; 92 (01) 33-41
- 40 Du K, Williams CD, McGill MR, Jaeschke H. Lower susceptibility of female mice to acetaminophen hepatotoxicity: role of mitochondrial glutathione, oxidant stress and c-jun N-terminal kinase. Toxicol Appl Pharmacol 2014; 281 (01) 58-66
- 41 Rohrer PR, Rudraiah S, Goedken MJ, Manautou JE. Is nuclear factor erythroid 2-related factor 2 responsible for sex differences in susceptibility to acetaminophen-induced hepatotoxicity in mice?. Drug Metab Dispos 2014; 42 (10) 1663-1674
- 42 Rubin JB, Hameed B, Gottfried M, Lee WM, Sarkar M. Acute Liver Failure Study Group. Acetaminophen-induced acute liver failure is more common and more severe in women. Clin Gastroenterol Hepatol 2018; 16 (06) 936-946
- 43 Kjartansdottir I, Bergmann OM, Arnadottir RS, Björnsson ES. Paracetamol intoxications: a retrospective population-based study in Iceland. Scand J Gastroenterol 2012; 47 (11) 1344-1352
- 44 Sami F, Berg S, Manadan AM, Mycyk MB. Acetaminophen overdose: analysis of 2018 US nationwide emergency database. Intern Emerg Med 2024; 19 (06) 1727-1732
- 45 Singal AK, Bataller R, Ahn J, Kamath PS, Shah VH. ACG Clinical Guideline: alcoholic liver disease. Am J Gastroenterol 2018; 113 (02) 175-194
- 46 Müller C. Liver, alcohol and gender. Wien Med Wochenschr 2006; 156 (19–20): 523-526
- 47 Sayaf K, Gabbia D, Russo FP, De Martin S. The role of sex in acute and chronic liver damage. Int J Mol Sci 2022; 23 (18) 23
- 48 Erol A, Karpyak VM. Sex and gender-related differences in alcohol use and its consequences: contemporary knowledge and future research considerations. Drug Alcohol Depend 2015; 156: 1-13
- 49 Gramenzi A, Caputo F, Biselli M. et al. Review article: alcoholic liver disease. –pathophysiological aspects and risk factors. Aliment Pharmacol Ther 2006; 24 (08) 1151-1161
- 50 Makri E, Goulas A, Polyzos SA. Epidemiology, pathogenesis, diagnosis and emerging treatment of nonalcoholic fatty liver disease. Arch Med Res 2021; 52 (01) 25-37
- 51 Nagral A, Bangar M, Menezes S. et al. Gender differences in nonalcoholic fatty liver disease. Euroasian J Hepatogastroenterol 2022; 12 (Suppl. 01) S19-S25
- 52 Balakrishnan M, Patel P, Dunn-Valadez S. et al. Women have a lower risk of nonalcoholic fatty liver disease but a higher risk of progression vs men: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 2021; 19 (01) 61-71.e15
- 53 Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver disease. Gastroenterology 2002; 122 (06) 1649-1657
- 54 Yang JD, Abdelmalek MF, Pang H. et al. Gender and menopause impact severity of fibrosis among patients with nonalcoholic steatohepatitis. Hepatology 2014; 59 (04) 1406-1414
- 55 Hamaguchi M, Kojima T, Ohbora A, Takeda N, Fukui M, Kato T. Aging is a risk factor of nonalcoholic fatty liver disease in premenopausal women. World J Gastroenterol 2012; 18 (03) 237-243
- 56 Ter Horst KW, Gilijamse PW, de Weijer BA. et al. Sexual dimorphism in hepatic, adipose tissue, and peripheral tissue insulin sensitivity in obese humans. Front Endocrinol (Lausanne) 2015; 6: 182
- 57 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
- 58 Conrad N, Misra S, Verbakel JY. et al. Incidence, prevalence, and co-occurrence of autoimmune disorders over time and by age, sex, and socioeconomic status: a population-based cohort study of 22 million individuals in the UK. Lancet 2023; 401 (10391): 1878-1890
- 59 Parikh-Patel A, Gold EB, Worman H, Krivy KE, Gershwin ME. Risk factors for primary biliary cirrhosis in a cohort of patients from the United States. Hepatology 2001; 33 (01) 16-21
- 60 Manns MP, Czaja AJ, Gorham JD. et al; American Association for the Study of Liver Diseases. Diagnosis and management of autoimmune hepatitis. Hepatology 2010; 51 (06) 2193-2213
- 61 Bambha K, Kim WR, Talwalkar J. et al. Incidence, clinical spectrum, and outcomes of primary sclerosing cholangitis in a United States community. Gastroenterology 2003; 125 (05) 1364-1369
- 62 Singh S, Talwalkar JA. Primary sclerosing cholangitis: diagnosis, prognosis, and management. Clin Gastroenterol Hepatol 2013; 11 (08) 898-907
- 63 Weismüller TJ, Trivedi PJ, Bergquist A. et al; International PSC Study Group. Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology 2017; 152 (08) 1975-1984.e8
- 64 Bataller R, Brenner DA. Liver fibrosis. J Clin Invest 2005; 115 (02) 209-218
- 65 Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol 2017; 14 (07) 397-411
- 66 Parkin DM. Global cancer statistics in the year 2000. Lancet Oncol 2001; 2 (09) 533-543
- 67 El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 2007; 132 (07) 2557-2576
- 68 Mittal S, El-Serag HB. Epidemiology of hepatocellular carcinoma: consider the population. J Clin Gastroenterol 2013; 47 Suppl (00) S2-S6
- 69 Zhang XF, Liu Y, Li JH. et al. [Effect of splenectomy on the risk of hepatocellular carcinoma development among patients with liver cirrhosis and portal hypertension: a multi-institutional cohort study]. Zhonghua Wai Ke Za Zhi 2021; 59 (10) 821-828
- 70 Akinyemiju T, Abera S, Ahmed M. et al; Global Burden of Disease Liver Cancer Collaboration. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the Global Burden of Disease Study 2015. JAMA Oncol 2017; 3 (12) 1683-1691
- 71 Zhai Y, Petrowsky H, Hong JC, Busuttil RW, Kupiec-Weglinski JW. Ischaemia-reperfusion injury in liver transplantation. –from bench to bedside. Nat Rev Gastroenterol Hepatol 2013; 10 (02) 79-89
- 72 Chen C, Hu LX, Dong T. et al. Apoptosis and autophagy contribute to gender difference in cardiac ischemia-reperfusion induced injury in rats. Life Sci 2013; 93 (07) 265-270
- 73 Crockett ET, Spielman W, Dowlatshahi S, He J. Sex differences in inflammatory cytokine production in hepatic ischemia-reperfusion. J Inflamm (Lond) 2006; 3: 16
- 74 Hundscheid IHR, Schellekens DHSM, Grootjans J. et al. Females are more resistant to ischemia-reperfusion-induced intestinal injury than males: a human study. Ann Surg 2020; 272 (06) 1070-1079
- 75 Rodríguez F, Nieto-Cerón S, Fenoy FJ. et al. Sex differences in nitrosative stress during renal ischemia. Am J Physiol Regul Integr Comp Physiol 2010; 299 (05) R1387-R1395
- 76 Zuo W, Zhang W, Chen NH. Sexual dimorphism in cerebral ischemia injury. Eur J Pharmacol 2013; 711 (1-3): 73-79
- 77 Michalopoulos GK, Bhushan B. Liver regeneration: biological and pathological mechanisms and implications. Nat Rev Gastroenterol Hepatol 2021; 18 (01) 40-55
- 78 Bizzaro D, Crescenzi M, Di Liddo R. et al. Sex-dependent differences in inflammatory responses during liver regeneration in a murine model of acute liver injury. Clin Sci (Lond) 2018; 132 (02) 255-272
- 79 Sayaf K, Zanotto I, Gabbia D. et al. Sex drives functional changes in the progression and regression of liver fibrosis. Int J Mol Sci 2023; 24 (22) 24
- 80 Birrer DL, Linecker M, López-López V. et al. Sex disparities in outcomes following major liver surgery: new powers of estrogen?. Ann Surg 2022; 276 (05) 875-881
- 81 Zhu M, Li Y, Shen Q, Gong Z, Liu D. Sex hormone receptors, calcium-binding protein and Yap1 signaling regulate sex-dependent liver cell proliferation following partial hepatectomy. Dis Model Mech 2024; 17 (10) 17
- 82 Guy J, Peters MG. Liver disease in women: the influence of gender on epidemiology, natural history, and patient outcomes. Gastroenterol Hepatol (N Y) 2013; 9 (10) 633-639
- 83 Pietrangelo A. Genetics, genetic testing, and management of hemochromatosis: 15 years since hepcidin. Gastroenterology 2015; 149 (05) 1240-1251.e4
- 84 Allen KJ, Gurrin LC, Constantine CC. et al. Iron-overload-related disease in HFE hereditary hemochromatosis. N Engl J Med 2008; 358 (03) 221-230
- 85 Cooper KM, Delk M, Devuni D, Sarkar M. Sex differences in chronic liver disease and benign liver lesions. JHEP Rep Innov Hepatol 2023; 5 (11) 100870
- 86 Shin N, Kim YH, Xu H. et al. Redefining Budd-Chiari syndrome: a systematic review. World J Hepatol 2016; 8 (16) 691-702
- 87 Buzzetti E, Parikh PM, Gerussi A, Tsochatzis E. Gender differences in liver disease and the drug-dose gender gap. Pharmacol Res 2017; 120: 97-108
- 88 Sutti S, Tacke F. Liver inflammation and regeneration in drug-induced liver injury: sex matters!. Clin Sci (Lond) 2018; 132 (05) 609-613
- 89 Chen TS, Doong ML, Chang FY, Lee SD, Wang PS. Effects of sex steroid hormones on gastric emptying and gastrointestinal transit in rats. Am J Physiol 1995; 268 (1 Pt 1): G171-G176
- 90 Yang L, Li Y, Hong H. et al. Sex differences in the expression of drug-metabolizing and transporter genes in human liver. J Drug Metab Toxicol 2012; 3 (03) 1000119
- 91 Lamba V, Lamba J, Yasuda K. et al. Hepatic CYP2B6 expression: gender and ethnic differences and relationship to CYP2B6 genotype and CAR (constitutive androstane receptor) expression. J Pharmacol Exp Ther 2003; 307 (03) 906-922
- 92 Mauvais-Jarvis F, Berthold HK, Campesi I. et al. Sex- and gender-based pharmacological response to drugs. Pharmacol Rev 2021; 73 (02) 730-762
- 93 Lai KP, Wong MH, Wong CK. Modulation of AhR-mediated CYP1A1 mRNA and EROD activities by 17beta-estradiol and dexamethasone in TCDD-induced H411E cells. Toxicol Sci 2004; 78 (01) 41-49
- 94 Koyuncuoğlu T, Yıldırım A, Dertsiz EK, Yüksel M, Ercan F, Yeğen BÇ. Estrogen receptor agonists protect against acetaminophen-induced hepatorenal toxicity in rats. Life Sci 2020; 263: 118561
- 95 Win S, Min RW, Chen CQ. et al. Expression of mitochondrial membrane-linked SAB determines severity of sex-dependent acute liver injury. J Clin Invest 2019; 129 (12) 5278-5293
- 96 Frezza M, di Padova C, Pozzato G, Terpin M, Baraona E, Lieber CS. High blood alcohol levels in women. The role of decreased gastric alcohol dehydrogenase activity and first-pass metabolism. N Engl J Med 1990; 322 (02) 95-99
- 97 Thurman RG. II. Alcoholic liver injury involves activation of Kupffer cells by endotoxin. Am J Physiol 1998; 275 (04) G605-G611
- 98 Yin M, Ikejima K, Wheeler MD. et al. Estrogen is involved in early alcohol-induced liver injury in a rat enteral feeding model. Hepatology 2000; 31 (01) 117-123
- 99 Ikejima K, Enomoto N, Iimuro Y. et al. Estrogen increases sensitivity of hepatic Kupffer cells to endotoxin. Am J Physiol 1998; 274 (04) G669-G676
- 100 Mishra P, Davies DA, Albensi BC. The interaction between NF-κB and estrogen in Alzheimer's disease. Mol Neurobiol 2023; 60 (03) 1515-1526
- 101 Purawarga Matada GS, Dhiwar PS, Abbas N. et al. Molecular docking and molecular dynamic studies: screening of phytochemicals against EGFR, HER2, estrogen and NF-KB receptors for their potential use in breast cancer. J Biomol Struct Dyn 2022; 40 (13) 6183-6192
- 102 Mezey E. Influence of sex hormones on alcohol metabolism. Alcohol Clin Exp Res 2000; 24 (04) 421
- 103 Ichiro S, Kamochi M, Yoshikawa H, Nakayama Y. Gender Difference in Alcoholic Liver Disease. IntechOpen; 2012
- 104 Kim DK, Kim YH, Jang HH. et al. Estrogen-related receptor γ controls hepatic CB1 receptor-mediated CYP2E1 expression and oxidative liver injury by alcohol. Gut 2013; 62 (07) 1044-1054
- 105 Järveläinen HA, Lukkari TA, Heinaro S, Sippel H, Lindros KO. The antiestrogen toremifene protects against alcoholic liver injury in female rats. J Hepatol 2001; 35 (01) 46-52
- 106 Fang C, Lindros KO, Badger TM, Ronis MJ, Ingelman-Sundberg M. Zonated expression of cytokines in rat liver: effect of chronic ethanol and the cytochrome P450 2E1 inhibitor, chlormethiazole. Hepatology 1998; 27 (05) 1304-1310
- 107 Gouillon Z, Lucas D, Li J. et al. Inhibition of ethanol-induced liver disease in the intragastric feeding rat model by chlormethiazole. Proc Soc Exp Biol Med 2000; 224 (04) 302-308
- 108 Zhu L, Brown WC, Cai Q. et al. Estrogen treatment after ovariectomy protects against fatty liver and may improve pathway-selective insulin resistance. Diabetes 2013; 62 (02) 424-434
- 109 Skubic C, Drakulić Ž, Rozman D. Personalized therapy when tackling nonalcoholic fatty liver disease: a focus on sex, genes, and drugs. Expert Opin Drug Metab Toxicol 2018; 14 (08) 831-841
- 110 Schiffrin M, Winkler C, Quignodon L. et al. Sex dimorphism of nonalcoholic fatty liver disease (NAFLD) in Pparg-null mice. Int J Mol Sci 2021; 22 (18) 22
- 111 Shu Z, Zhang G, Zhu X, Xiong W. Estrogen receptor α mediated M1/M2 macrophages polarization plays a critical role in NASH of female mice. Biochem Biophys Res Commun 2022; 596: 63-70
- 112 Yang M, Liu Q, Huang T. et al. Dysfunction of estrogen-related receptor alpha-dependent hepatic VLDL secretion contributes to sex disparity in NAFLD/NASH development. Theranostics 2020; 10 (24) 10874-10891
- 113 Della Torre S. Beyond the X factor: relevance of sex hormones in NAFLD pathophysiology. Cells 2021; 10 (09) 10
- 114 Sharma G, Prossnitz ER. G-protein-coupled estrogen receptor (GPER) and sex-specific metabolic homeostasis. Adv Exp Med Biol 2017; 1043: 427-453
- 115 Meoli L, Isensee J, Zazzu V. et al. Sex- and age-dependent effects of Gpr30 genetic deletion on the metabolic and cardiovascular profiles of diet-induced obese mice. Gene 2014; 540 (02) 210-216
- 116 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 (18) 14851-14862
- 117 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 (06) 1466-1475
- 118 Lee C, Kim J, Han J. et al. Formyl peptide receptor 2 determines sex-specific differences in the progression of nonalcoholic fatty liver disease and steatohepatitis. Nat Commun 2022; 13 (01) 578
- 119 Wu J, Yao XY, Shi RX, Liu SF, Wang XY. A potential link between polycystic ovary syndrome and non-alcoholic fatty liver disease: an update meta-analysis. Reprod Health 2018; 15 (01) 77
- 120 Sarkar M, Yates K, Suzuki A. et al. Low testosterone is associated with nonalcoholic steatohepatitis and fibrosis severity in men. Clin Gastroenterol Hepatol 2021; 19 (02) 400-402.e2
- 121 Jaruvongvanich V, Sanguankeo A, Riangwiwat T, Upala S. Testosterone, sex hormone-binding globulin and nonalcoholic fatty liver disease: a systematic review and meta-analysis. Ann Hepatol 2017; 16 (03) 382-394
- 122 Jia Y, Yee JK, Wang C. et al. Testosterone protects high-fat/low-carbohydrate diet-induced nonalcoholic fatty liver disease in castrated male rats mainly via modulating endoplasmic reticulum stress. Am J Physiol Endocrinol Metab 2018; 314 (04) E366-E376
- 123 Nikolaenko L, Jia Y, Wang C. et al. Testosterone replacement ameliorates nonalcoholic fatty liver disease in castrated male rats. Endocrinology 2014; 155 (02) 417-428
- 124 Schwinge D, Schramm C. Sex-related factors in autoimmune liver diseases. Semin Immunopathol 2019; 41 (02) 165-175
- 125 Smyk DS, Rigopoulou EI, Pares A. et al. Sex differences associated with primary biliary cirrhosis. Clin Dev Immunol 2012; 2012: 610504
- 126 Yasuda M, Shimizu I, Shiba M, Ito S. Suppressive effects of estradiol on dimethylnitrosamine-induced fibrosis of the liver in rats. Hepatology 1999; 29 (03) 719-727
- 127 Xu J-W, Gong J, Chang X-M. et al. Estrogen reduces CCL4- induced liver fibrosis in rats. World J Gastroenterol 2002; 8 (05) 883-887
- 128 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 (07) 897-907
- 129 Zhou YJ, Yin DM, Chen HS, Shi JH, Sha BX, Wang X. Inhibitory effects of idoxifene on hepatic fibrosis in rats. Acta Pharmacol Sin 2005; 26 (05) 581-586
- 130 Shimizu I, Ito S. Protection of estrogens against the progression of chronic liver disease. Hepatol Res 2007; 37 (04) 239-247
- 131 Shimizu I, Kohno N, Tamaki K. et al. Female hepatology: favorable role of estrogen in chronic liver disease with hepatitis B virus infection. World J Gastroenterol 2007; 13 (32) 4295-4305
- 132 Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 2017; 121: 27-42
- 133 Zhou Y, Shimizu I, Lu G. et al. Hepatic stellate cells contain the functional estrogen receptor beta but not the estrogen receptor alpha in male and female rats. Biochem Biophys Res Commun 2001; 286 (05) 1059-1065
- 134 Zhang B, Zhang CG, Ji LH, Zhao G, Wu ZY. Estrogen receptor β selective agonist ameliorates liver cirrhosis in rats by inhibiting the activation and proliferation of hepatic stellate cells. J Gastroenterol Hepatol 2018; 33 (03) 747-755
- 135 Zhang K, Lin L, Zhu Y, Zhang N, Zhou M, Li Y. Saikosaponin d alleviates liver fibrosis by negatively regulating the ROS/NLRP3 inflammasome through activating the ERβ pathway. Front Pharmacol 2022; 13: 894981
- 136 Inagaki Y, Okazaki I. Emerging insights into transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut 2007; 56 (02) 284-292
- 137 McCarty MF, Barroso-Aranda J, Contreras F. Genistein and phycocyanobilin may prevent hepatic fibrosis by suppressing proliferation and activation of hepatic stellate cells. Med Hypotheses 2009; 72 (03) 330-332
- 138 Ganai AA, Husain M. Genistein attenuates D-GalN induced liver fibrosis/chronic liver damage in rats by blocking the TGF-β/Smad signaling pathways. Chem Biol Interact 2017; 261: 80-85
- 139 Evans MJ, Lai K, Shaw LJ, Harnish DC, Chadwick CC. Estrogen receptor alpha inhibits IL-1beta induction of gene expression in the mouse liver. Endocrinology 2002; 143 (07) 2559-2570
- 140 Ma X, Zhou Y, Qiao B. et al. Androgen aggravates liver fibrosis by activation of NLRP3 inflammasome in CCl4-induced liver injury mouse model. Am J Physiol Endocrinol Metab 2020; 318 (05) E817-E829
- 141 Wang S-H, Yeh S-H, Lin W-H, Wang HY, Chen DS, Chen PJ. Identification of androgen response elements in the enhancer I of hepatitis B virus: a mechanism for sex disparity in chronic hepatitis B. Hepatology 2009; 50 (05) 1392-1402
- 142 Wu M-H, Ma W-L, Hsu C-L. et al. Androgen receptor promotes hepatitis B virus-induced hepatocarcinogenesis through modulation of hepatitis B virus RNA transcription. Sci Transl Med 2010; 2 (32) 32ra35
- 143 Cha MY, Kim CM, Park YM, Ryu WS. Hepatitis B virus X protein is essential for the activation of Wnt/beta-catenin signaling in hepatoma cells. Hepatology 2004; 39 (06) 1683-1693
- 144 Ding Q, Xia W, Liu J-C. et al. Erk associates with and primes GSK-3beta for its inactivation resulting in upregulation of beta-catenin. Mol Cell 2005; 19 (02) 159-170
- 145 Bouchard MJ, Wang L, Schneider RJ. Activation of focal adhesion kinase by hepatitis B virus HBx protein: multiple functions in viral replication. J Virol 2006; 80 (09) 4406-4414
- 146 Yeh S-H, Chen P-J. Gender disparity of hepatocellular carcinoma: the roles of sex hormones. Oncology 2010; 78 (Suppl. 01) 172-179
- 147 Chen P-J, Yeh S-H, Liu W-H. et al. Androgen pathway stimulates microRNA-216a transcription to suppress the tumor suppressor in lung cancer-1 gene in early hepatocarcinogenesis. Hepatology 2012; 56 (02) 632-643
- 148 Feng H, Cheng ASL, Tsang DP. et al. Cell cycle-related kinase is a direct androgen receptor-regulated gene that drives β-catenin/T cell factor-dependent hepatocarcinogenesis. J Clin Invest 2011; 121 (08) 3159-3175
- 149 Llovet JM. Updated treatment approach to hepatocellular carcinoma. J Gastroenterol 2005; 40 (03) 225-235
- 150 Zhang H, Spencer K, Burley SK, Zheng XFS. Toward improving androgen receptor-targeted therapies in male-dominant hepatocellular carcinoma. Drug Discov Today 2021; 26 (06) 1539-1546
- 151 Ji F, Zhang J, Liu N. et al. Blocking hepatocarcinogenesis by a cytochrome P450 family member with female-preferential expression. Gut 2022; 71 (11) 2313-2324
- 152 Naugler WE, Sakurai T, Kim S. et al. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 2007; 317 (5834): 121-124
- 153 Hou J, Xu J, Jiang R. et al. Estrogen-sensitive PTPRO expression represses hepatocellular carcinoma progression by control of STAT3. Hepatology 2013; 57 (02) 678-688
- 154 Huang F-Y, Wong DK-H, Seto W-K, Lai C-L, Yuen M-F. Estradiol induces apoptosis via activation of miRNA-23a and p53: implication for gender difference in liver cancer development. Oncotarget 2015; 6 (33) 34941-34952
- 155 Li C-L, Yeh K-H, Liu W-H. et al. Elevated p53 promotes the processing of miR-18a to decrease estrogen receptor-α in female hepatocellular carcinoma. Int J Cancer 2015; 136 (04) 761-770
- 156 Wei Q, Guo P, Mu K. et al. Estrogen suppresses hepatocellular carcinoma cells through ERβ-mediated upregulation of the NLRP3 inflammasome. Lab Invest 2015; 95 (07) 804-816
- 157 Chow PK, Tai BC, Tan CK. et al; Asian-Pacific Hepatocellular Carcinoma Trials Group. High-dose tamoxifen in the treatment of inoperable hepatocellular carcinoma: a multicenter randomized controlled trial. Hepatology 2002; 36 (05) 1221-1226
- 158 Riestra S, Rodriguez M, Delgado M. et al. Tamoxifen does not improve survival of patients with advanced hepatocellular carcinoma. J Clin Gastroenterol 1998; 26 (03) 200-203
- 159 Shimizu I. CLIP Group (Cancer of the Liver Italian Programme). Tamoxifen in treatment of hepatocellular carcinoma: a randomised controlled trial. Lancet 1998; 352 (9121): 17-20
- 160 Barbare JC, Bouché O, Bonnetain F. et al. Randomized controlled trial of tamoxifen in advanced hepatocellular carcinoma. J Clin Oncol 2005; 23 (19) 4338-4346
- 161 Harada H, Pavlick KP, Hines IN. et al. Selected contribution: effects of gender on reduced-size liver ischemia and reperfusion injury. J Appl Physiol 2001; 91 (06) 2816-2822
- 162 Harada H, Pavlick KP, Hines IN. et al. Sexual dimorphism in reduced-size liver ischemia and reperfusion injury in mice: role of endothelial cell nitric oxide synthase. Proc Natl Acad Sci U S A 2003; 100 (02) 739-744
- 163 Harada H, Bharwani S, Pavlick KP, Korach KS, Grisham MB. Estrogen receptor-alpha, sexual dimorphism and reduced-size liver ischemia and reperfusion injury in mice. Pediatr Res 2004; 55 (03) 450-456
- 164 Fels JA, Manfredi G. Sex differences in ischemia/reperfusion injury: the role of mitochondrial permeability transition. Neurochem Res 2019; 44 (10) 2336-2345
- 165 Huang C, Gu H, Zhang W, Herrmann JL, Wang M. Testosterone-down-regulated Akt pathway during cardiac ischemia/reperfusion: a mechanism involving BAD, Bcl-2 and FOXO3a. J Surg Res 2010; 164 (01) e1-e11
- 166 Wang M, Tsai BM, Kher A, Baker LB, Wairiuko GM, Meldrum DR. Role of endogenous testosterone in myocardial proinflammatory and proapoptotic signaling after acute ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2005; 288 (01) H221-H226
- 167 Liddle C, Farrell GC. Role of the oestrogen receptor in liver regeneration in the male rat. J Gastroenterol Hepatol 1993; 8 (06) 524-529
- 168 Francavilla A, di Leo A, Eagon PK. et al. Regenerating rat liver: correlations between estrogen receptor localization and deoxyribonucleic acid synthesis. Gastroenterology 1984; 86 (03) 552-557
- 169 Fisher B, Gunduz N, Saffer EA, Zheng S. Relation of estrogen and its receptor to rat liver growth and regeneration. Cancer Res 1984; 44 (06) 2410-2415
- 170 Francavilla A, Eagon PK, DiLeo A. et al. Sex hormone-related functions in regenerating male rat liver. Gastroenterology 1986; 91 (05) 1263-1270
- 171 Kao TL, Kuan YP, Cheng WC. et al. Estrogen receptors orchestrate cell growth and differentiation to facilitate liver regeneration. Theranostics 2018; 8 (10) 2672-2682
- 172 Chaturantabut S, Shwartz A, Evason KJ. et al. Estrogen activation of G-Protein-coupled estrogen receptor 1 regulates phosphoinositide 3-kinase and mTOR signaling to promote liver growth in zebrafish and proliferation of human hepatocytes. Gastroenterology 2019; 156 (06) 1788-1804.e13
- 173 Yamaguchi M, Yu L, Nazmy El-Assal O. et al. Androgen metabolism in regenerating liver of male rats: evidence for active uptake and utilization of testosterone. Hepatol Res 2001; 20 (01) 114-127
- 174 Du L, Bayir H, Lai Y. et al. Innate gender-based proclivity in response to cytotoxicity and programmed cell death pathway. J Biol Chem 2004; 279 (37) 38563-38570
- 175 Barker JM, Torregrossa MM, Arnold AP, Taylor JR. Dissociation of genetic and hormonal influences on sex differences in alcoholism-related behaviors. J Neurosci 2010; 30 (27) 9140-9144
- 176 Chen X, McClusky R, Chen J. et al. The number of x chromosomes causes sex differences in adiposity in mice. PLoS Genet 2012; 8 (05) e1002709
- 177 Romeo S. Notch and nonalcoholic fatty liver and fibrosis. N Engl J Med 2019; 380 (07) 681-683
- 178 Zhu C, Kim K, Wang X. et al. Hepatocyte Notch activation induces liver fibrosis in nonalcoholic steatohepatitis. Sci Transl Med 2018; 10 (468) 10
- 179 Hanley KP, Oakley F, Sugden S, Wilson DI, Mann DA, Hanley NA. Ectopic SOX9 mediates extracellular matrix deposition characteristic of organ fibrosis. J Biol Chem 2008; 283 (20) 14063-14071
- 180 Pritchett J, Harvey E, Athwal V. et al. Osteopontin is a novel downstream target of SOX9 with diagnostic implications for progression of liver fibrosis in humans. Hepatology 2012; 56 (03) 1108-1116
- 181 Athwal VS, Pritchett J, Llewellyn J. et al. SOX9 predicts progression toward cirrhosis in patients while its loss protects against liver fibrosis. EMBO Mol Med 2017; 9 (12) 1696-1710
- 182 Fu M, Yin W, Zhang W, Zhu Y, Ni H, Gong L. MicroRNA-15a inhibits hepatic stellate cell activation and proliferation via targeting SRY-box transcription factor 9. Bioengineered 2022; 13 (05) 13011-13020
- 183 Wang C, Deng J, Deng H. et al. A novel Sox9/lncRNA H19 axis contributes to hepatocyte death and liver fibrosis. Toxicol Sci 2020; 177 (01) 214-225
- 184 Jiang H-L, Xu D, Yu H. et al. DAX-1 inhibits hepatocellular carcinoma proliferation by inhibiting β-catenin transcriptional activity. Cell Physiol Biochem 2014; 34: 734-742
- 185 Motawi TMK, Sadik NAH, Sabry D, Shahin NN, Fahim SA. rs2267531, a promoter SNP within glypican-3 gene in the X chromosome, is associated with hepatocellular carcinoma in Egyptians. Sci Rep 2019; 9 (01) 6868
- 186 Liao YL, Sun YM, Chau GY. et al. Identification of SOX4 target genes using phylogenetic footprinting-based prediction from expression microarrays suggests that overexpression of SOX4 potentiates metastasis in hepatocellular carcinoma. Oncogene 2008; 27 (42) 5578-5589
- 187 Wang W, Zhang J, Zhan X. et al. SOX4 is associated with poor prognosis in cholangiocarcinoma. Biochem Biophys Res Commun 2014; 452 (03) 614-621
- 188 Kawaguchi Y. Sox9 and programming of liver and pancreatic progenitors. J Clin Invest 2013; 123 (05) 1881-1886
- 189 Tsai C-N, Yu S-C, Lee C-W. et al. SOX4 activates CXCL12 in hepatocellular carcinoma cells to modulate endothelial cell migration and angiogenesis in vivo. Oncogene 2020; 39 (24) 4695-4710
- 190 Tsuei D-J, Hsu H-C, Lee P-H. et al. RBMY, a male germ cell-specific RNA-binding protein, activated in human liver cancers and transforms rodent fibroblasts. Oncogene 2004; 23 (34) 5815-5822
- 191 Chua H-H, Tsuei D-J, Lee P-H. et al. RBMY, a novel inhibitor of glycogen synthase kinase 3β, increases tumor stemness and predicts poor prognosis of hepatocellular carcinoma. Hepatology 2015; 62 (05) 1480-1496
- 192 Li S, Mo C, Huang S. et al. Over-expressed Testis-specific Protein Y-encoded 1 as a novel biomarker for male hepatocellular carcinoma. PLoS One 2014; 9 (02) e89219
- 193 Fan XD, Zheng HB, Fan XS, Lu S. Increase of SOX9 promotes hepatic ischemia/reperfusion (IR) injury by activating TGF-β1. Biochem Biophys Res Commun 2018; 503 (01) 215-221
- 194 Han X, Wang Y, Pu W. et al. Lineage tracing reveals the bipotency of SOX9+ hepatocytes during liver regeneration. Stem Cell Reports 2019; 12 (03) 624-638
- 195 Yovchev MI, Locker J, Oertel M. Biliary fibrosis drives liver repopulation and phenotype transition of transplanted hepatocytes. J Hepatol 2016; 64 (06) 1348-1357
- 196 Shao C, Jing Y, Zhao S. et al. LPS/Bcl3/YAP1 signaling promotes Sox9+HNF4α+ hepatocyte-mediated liver regeneration after hepatectomy. Cell Death Dis 2022; 13 (03) 277
- 197 Gong S, Zeng Y, Wang Z. et al. Intestinal deguelin drives resistance to acetaminophen-induced hepatotoxicity in female mice. Gut Microbes 2024; 16 (01) 2404138
- 198 Corpechot C, Chrétien Y, Chazouillères O, Poupon R. Demographic, lifestyle, medical and familial factors associated with primary biliary cirrhosis. J Hepatol 2010; 53 (01) 162-169
- 199 Gershwin ME, Selmi C, Worman HJ. et al; USA PBC Epidemiology Group. Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients. Hepatology 2005; 42 (05) 1194-1202
- 200 Prince MI, Ducker SJ, James OF. Case-control studies of risk factors for primary biliary cirrhosis in two United Kingdom populations. Gut 2010; 59 (04) 508-512
- 201 Karlsen TH, Lammert F, Thompson RJ. Genetics of liver disease: from pathophysiology to clinical practice. J Hepatol 2015; 62 (1, Suppl): S6-S14
- 202 Greten TF. Gender disparity in HCC: is it the fat and not the sex?. J Exp Med 2019; 216 (05) 1014-1015
- 203 Manieri E, Herrera-Melle L, Mora A. et al. Adiponectin accounts for gender differences in hepatocellular carcinoma incidence. J Exp Med 2019; 216 (05) 1108-1119
- 204 Yang Q, Yan C, Yin C, Gong Z. Serotonin activated hepatic stellate cells contribute to sex disparity in hepatocellular carcinoma. Cell Mol Gastroenterol Hepatol 2017; 3 (03) 484-499
- 205 Hu Z, Jepps TA, Zhou L, Liu J, Li M, Abbott GW. Kcne4 deletion sex dependently inhibits the RISK pathway response and exacerbates hepatic ischemia-reperfusion injury in mice. Am J Physiol Regul Integr Comp Physiol 2019; 316 (05) R552-R562
- 206 Shang D, Wang L, Klionsky DJ, Cheng H, Zhou R. Sex differences in autophagy-mediated diseases: toward precision medicine. Autophagy 2021; 17 (05) 1065-1076
- 207 Campana L, Esser H, Huch M, Forbes S. Liver regeneration and inflammation: from fundamental science to clinical applications. Nat Rev Mol Cell Biol 2021; 22 (09) 608-624
- 208 Shimizu I. Accounting for sex and gender makes for better science. Nature 2020; 588 (7837): 196
- 209 Hunt L, Nielsen MW, Schiebinger L. A framework for sex, gender, and diversity analysis in research. Science 2022; 377 (6614): 1492-1495
- 210 Peters SAE, Woodward M. A roadmap for sex- and gender-disaggregated health research. BMC Med 2023; 21 (01) 354
- 211 de Beaurepaire R. Suppression of alcohol dependence using baclofen: a 2-year observational study of 100 patients. Front Psychiatry 2012; 3: 103
- 212 Crabb DW, Im GY, Szabo G, Mellinger JL, Lucey MR. Diagnosis and treatment of alcohol-associated liver diseases: 2019 Practice Guidance From the American Association for the Study of Liver Diseases. Hepatology 2020; 71 (01) 306-333
- 213 Cotter TG, Rinella M. Nonalcoholic fatty liver disease 2020: the state of the disease. Gastroenterology 2020; 158 (07) 1851-1864
- 214 Li N, Cui C, Xu J, Mi M, Wang J, Qin Y. Quercetin intervention reduced hepatic fat deposition in patients with nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled crossover clinical trial. Am J Clin Nutr 2024; 120 (03) 507-517
- 215 Yan H, Wu W, Chang X. et al. Gender differences in the efficacy of pioglitazone treatment in nonalcoholic fatty liver disease patients with abnormal glucose metabolism. Biol Sex Differ 2021; 12 (01) 1
- 216 Carbone M, Mells GF, Pells G. et al; UK PBC Consortium. Sex and age are determinants of the clinical phenotype of primary biliary cirrhosis and response to ursodeoxycholic acid. Gastroenterology 2013; 144 (03) 560-569.e7 , quiz e13–e14
- 217 Carey EJ, Ali AH, Lindor KD. Primary biliary cirrhosis. Lancet 2015; 386 (10003): 1565-1575
- 218 Hirschfield GM, Dyson JK, Alexander GJM. et al. The British Society of Gastroenterology/UK-PBC primary biliary cholangitis treatment and management guidelines. Gut 2018; 67 (09) 1568-1594
- 219 Muratori L, Lohse AW, Lenzi M. Diagnosis and management of autoimmune hepatitis. BMJ 2023; 380: e070201
- 220 Czaja AJ, Donaldson PT. Gender effects and synergisms with histocompatibility leukocyte antigens in type 1 autoimmune hepatitis. Am J Gastroenterol 2002; 97 (08) 2051-2057
- 221 Al-Chalabi T, Underhill JA, Portmann BC, McFarlane IG, Heneghan MA. Impact of gender on the long-term outcome and survival of patients with autoimmune hepatitis. J Hepatol 2008; 48 (01) 140-147
- 222 Kim MN, Han JW, An J. et al; Korean Association for the Study of the Liver (KASL). KASL clinical practice guidelines for noninvasive tests to assess liver fibrosis in chronic liver disease. Clin Mol Hepatol 2024; 30 (Suppl): S5-S105
- 223 Shimizu I. Chinese Society of Hepatology Chinese Medical Association, Chinese Society of Gastroenterology Chinese Medical Association, Chinese Society of Infectious Diseases, Chinese Medical Association. [Consensus on the diagnosis and therapy of hepatic fibrosis in]. Zhonghua Gan Zang Bing Za Zhi 2019; 27 (09) 657-667
- 224 Kammula AV, Schäffer AA, Rajagopal PS, Kurzrock R, Ruppin E. Outcome differences by sex in oncology clinical trials. Nat Commun 2024; 15 (01) 2608
- 225
D'Abbondanza M,
Ministrini S,
Pucci G.
et al.
Very Low-Carbohydrate Ketogenic Diet for the Treatment of Severe Obesity and Associated
Non-Alcoholic Fatty Liver Disease: The Role of Sex Differences. Nutrients 2020;12









