Original Article
Hepatology
Metabolic Dysfunction-Associated Fatty Liver Disease and Subsequent Development of Adverse Pregnancy Outcomes

https://doi.org/10.1016/j.cgh.2021.11.007Get rights and content

Background & Aims

Recently, metabolic dysfunction–associated fatty liver disease (MAFLD), rather than nonalcoholic fatty liver disease (NAFLD), was proposed to better describe liver disease associated with metabolic dysfunction (MD). In this study, we attempted to investigate the impact of MAFLD on pregnancy complications.

Methods

The current study is a secondary analysis of a multicenter prospective cohort designed to examine the risk of NAFLD during pregnancy. In the first trimester, enrolled pregnant women were evaluated for hepatic steatosis by liver ultrasonography, and blood samples were collected for biochemical measurements. The study population was divided into 3 groups: no NAFLD, hepatic steatosis but without metabolic dysfunction (non-MD NAFLD), and MAFLD. The primary outcome was the subsequent development of adverse pregnancy outcomes, including gestational diabetes mellitus, pregnancy-associated hypertension, preterm birth, and fetal growth abnormalities.

Results

The study population consisted of 1744 pregnant women, including 1523 with no NAFLD, 43 with non-MD NAFLD, and 178 with MAFLD. The risk of subsequent development of adverse pregnancy outcomes was higher in MAFLD than in non-MD NAFLD (adjusted odds ratio, 4.03; 95% CI, 1.68–9.67), whereas the risk was not significantly different between no NAFLD and non-MD NAFLD. Among women with no NAFLD, the presence of MD increased the risk of adverse pregnancy outcomes. However, women with MAFLD were at higher risk for adverse pregnancy outcomes than women with no NAFLD without MD or those with no NAFLD with MD.

Conclusions

In pregnant women, MAFLD may be associated with an increased risk of subsequent adverse pregnancy outcomes.

Section snippets

Methods

The cohort included singleton pregnant women who were enrolled during their first trimester between October 2014 and August 2020 and followed up until delivery. After enrollment, baseline demographics and prepregnancy data were retrieved using questionnaires. At 10 to 14 weeks of gestation, participants underwent physical measurements and liver ultrasonography for evaluation of hepatic steatosis, and a fasting blood sample was collected for blood chemistry analysis. The study was approved by

Subject Population

During the study period, a total of 1922 pregnant women were enrolled. After excluding subjects with hepatitis B or C (n = 17), incomplete evaluation for MD (n = 11), withdrawal of consent (n = 43), previable birth (n = 14), and loss to follow-up evaluation (n = 93), the remaining 1744 pregnant women were included in the final analysis. According to the presence or absence of hepatic steatosis and MD, the study population was divided into 3 groups: no hepatic steatosis (no NAFLD, n = 1523);

Discussion

The principal findings of the current study were as follows: (1) among pregnant women, MAFLD was present in 10% of the study population, and among women with hepatic steatosis, MD was present in 81%; (2) the risk of subsequent development of adverse pregnancy outcomes was higher in women with MAFLD than in non-MD NAFLD women, whereas the risk was not significantly different between women without NAFLD and women with non-MD NAFLD; and (3) among women with no NAFLD, MD itself increased the risk

CRediT Authorship Contributions

Seung Mi Lee (Conceptualization: Equal; Formal analysis: Lead; Investigation: Equal; Writing – original draft: Lead; Writing – review & editing: Lead)

Young Mi Jung (Investigation: Equal; Writing – review & editing: Supporting)

Eun Saem Choi (Investigation: Supporting; Writing – review & editing: Supporting)

Soo Heon Kwak (Formal analysis: Supporting; Writing – review & editing: Supporting)

Ja Nam Koo (Investigation: Equal; Writing – review & editing: Supporting)

Ig Hwan Oh (Investigation:

References (28)

  • S.M. Lee et al.

    Non-alcoholic fatty liver disease in the first trimester and subsequent development of gestational diabetes mellitus

    Diabetologia

    (2019)
  • S.M. Lee et al.

    Nonalcoholic fatty liver disease is a risk factor for large-for-gestational-age birthweight

    PLoS One

    (2019)
  • Y.M. Jung et al.

    The risk of pregnancy-associated hypertension in women with nonalcoholic fatty liver disease

    Liver Int

    (2020)
  • S. Hong et al.

    A comparison of predictive performances between old versus new criteria in a risk-based screening strategy for gestational diabetes mellitus

    Diabetes Metab J

    (2020)
  • Cited by (10)

    • Pregnancy and Metabolic-Associated Fatty Liver Disease

      2023, Endocrinology and Metabolism Clinics of North America
    • Transcriptional pathways linked to fetal and maternal hepatic dysfunction caused by gestational exposure to perfluorooctanoic acid (PFOA) or hexafluoropropylene oxide-dimer acid (HFPO-DA or GenX) in CD-1 mice

      2022, Ecotoxicology and Environmental Safety
      Citation Excerpt :

      These biological processes are compromised in MAFLD (Eslam et al., 2020; Shao et al., 2020). Furthermore, MAFLD during pregnancy is associated with increased risk for preeclampsia, pregnancy-induced hypertension, and gestational diabetes mellitus (Lee et al., 2021). The ability to connect gene expression to phenotype is critical for establishing causal relationships between exposures and outcomes and for building confidence in toxicological mechanisms of action.

    • MAFLD and Pregnancy: What are the Consequences?

      2022, Clinical Gastroenterology and Hepatology
    • Reply

      2022, Clinical Gastroenterology and Hepatology
    • The prevalence of metabolic dysfunction–associated fatty liver disease and its association on adverse pregnancy outcomes in women with gestational diabetes mellitus

      2022, Diabetes Research and Clinical Practice
      Citation Excerpt :

      However, within this GDM cohort, the presence of MAFLD was not found to be associated with worse pregnancy outcomes. This is in contrast to prior studies which identified MAFLD as a potential risk factor for both adverse maternal and neonatal pregnancy outcomes, with a 2–3 fold higher risk of gestational hypertension and pre–eclampsia and a 2–fold increased risk of having both SGA or LGA neonates as well as pre–term birth and extreme prematurity (<32 weeks gestation) [26–28]. The lack of association of MAFLD with adverse pregnancy outcomes demonstrated in our study may be related to the predominance of participant recruitment occurring from a high–risk antenatal clinic, where closer monitoring and more intensive management is already in place for these at–risk group of women.

    • Fatty liver indices and their association with glucose metabolism in pregnancy – An observational cohort study

      2022, Diabetes Research and Clinical Practice
      Citation Excerpt :

      Metabolic dysfunction could be considered to be the underlying risk factor for both, the development of GDM and fatty liver disease; however, another Korean study found that pregnant women with MAFLD had an approximately twofold higher risk for developing GDM compared to women with no fatty liver disease but metabolic dysfunction, even after adjustment for age, prior history of GDM or family history of type II diabetes. This suggests that hepatic steatosis has an adverse impact on glycemic regulation in pregnancy, independently of the presence of metabolic dysfunction [32]. Some mechanistic studies suggest that increased ectopic lipid contents, specifically intracellular lipid metabolites, inhibit insulin signaling leading to hepatic insulin resistance [33].

    View all citing articles on Scopus

    Conflicts of interest The authors disclose no conflicts.

    Funding This work was supported by the Seoul National University Hospital research fund (0320212200), and two National Research Foundation of Korea grants funded by the Ministry of Education, Science and Technology of the Korean government (2021R1A2C2005820 and 2021M3A9E4021818).

    b

    Authors share co-corresponding authorship.

    View full text