Skip to main content

Application of metabolomics in intrahepatic cholestasis of pregnancy: a systematic review

Abstract

Background

Intrahepatic cholestasis of pregnancy (ICP) is a severe idiopathic disorder of bile metabolism; however, the etiology and pathogenesis of ICP remain unclear.

Aims

This study comprehensively reviewed metabolomics studies related to ICP, to help in identifying the pathophysiological changes of ICP and evaluating the potential application of metabolomics in its diagnosis.

Methods

Relevant articles were searched through 2 online databases (PubMed and Web of Science) from January 2000 to March 2022. The metabolites involved were systematically examined and compared. Pathway analysis was conducted through the online software MetaboAnalyst 5.0.

Results

A total of 14 papers reporting 212 metabolites were included in this study. There were several highly reported metabolites: bile acids, such as glycocholic acid, taurochenodeoxycholic acid, taurocholic acid, tauroursodeoxycholic acid, and glycochenodeoxycholic acid. Dysregulation of metabolic pathways involved bile acid metabolism and lipid metabolism. Metabolites related to lipid metabolism include phosphatidylcholine, phosphorylcholine, phosphatidylserine, sphingomyelin, and ceramide.

Conclusions

This study provides a systematic review of metabolomics of ICP and deepens our understanding of the etiology of ICP.

Introduction

Intrahepatic cholestasis of pregnancy (ICP) is a severe pregnancy complication, affecting 0.1–2% of pregnant women [1]. ICP is clinically characterized by pruritus and increased bile acids, and the symptoms usually disappear after labor [2]. Although essentially non-threatening from a maternal perspective, ICP is associated with an elevated risk of adverse fetal outcomes, including spontaneous preterm labor, meconium staining of the amniotic fluid, asphyxial events, and sudden intrauterine death [3, 4]. Despite that studies have shown that ICP is influenced by genetic predisposition, hormone levels, altered immunity, underlying liver disease, and environmental factors [5,6,7], the etiology and pathogenesis of ICP remain unclear. Meanwhile, the diagnosis of ICP mainly relies on detecting the serum concentration of total bile acid (TBA), but there are still several limitations [8]. For example, not all ICP patients have elevated TBA levels [9], and other liver diseases may also cause an increase in TBA [10]. Thus, enhanced research on the etiology, pathogenesis and diagnosis method is urgently required.

Metabolomics is a newly developed technology that can quantitatively analyze all metabolites in organisms and uncover the relative relationship between metabolites with physiological and pathological changes. Accordingly, it enables us to recognize the metabolites and metabolic pathways related to ICP, which could promote a deeper understanding of its etiology and pathophysiology, as well as boost its early prevention, diagnosis, and treatment.

In this study, we reviewed all metabolomics studies conducted on ICP over the last 20 years and systematically collected and analyzed the information from these researches. We summarized the significant changes in metabolic biomarkers and pathways of ICP to help us 1) understand the etiology and pathogenesis of ICP and 2) evaluate the potential application of metabolomics in ICP diagnosis.

Methods

Literature search

We obtained relevant publications from PubMed and Web of Science databases from January 2000 to March 2022, with the following searching terms: (“metabolome” or “metabolomics” or “metabolite” or “metabonomics” or “metabolic profiling” or “metabolic signature” or “metabolic biomarker” or “metabolic profile” or “metabolic portraits”) AND (“intrahepatic cholestasis of pregnancy”). All articles were searched and examined by two authors independently to assess their suitability for inclusion in the review, and a third researcher made a final decision in cases of disagreement.

Inclusion and exclusion criteria

The inclusion criteria were (1) metabolomics studies on ICP, (2) full text in English, and (3) studies recording the positive or negative relationship between metabolite markers and ICP. The exclusion criteria were as follows: (1) review articles, (2) animal and cell studies, and (3) studies evaluating drug effects.

Data extraction

We extracted the following information after reading the full articles and supplementary materials: (1) basic information of included studies, including first author, published date, and journal; (2) basic information of subjects, including sample size and singleton/twin; (3) study design, ICP diagnostic criteria, biological specimen, sampling time, and analytic platform; (4) the significant metabolites with changing trends. In addition, studies by the same first or corresponding author were checked whether there were overlaps in content.

Statistical analysis

The frequencies on biological specimens, targeted/untargeted, analytic platforms, sample sizes, and frequently reported biomarkers were computed and charted. Pathway enrichment analysis and topology analysis were performed by the MetaboAnalyst 5.0 online software (https://www.metaboanalyst.ca) [11].

Results

Study characteristics

A total of 14 articles [9, 12,13,14,15,16,17,18,19,20,21,22,23,24] were included in this systematic review (Fig. 1). The characteristics of the 14 studies are presented in Table 1. Nine studies were performed with blood samples (serum and plasma), three with urine samples, one with hair samples, and one simultaneously collected placenta and serum (Fig. 2a). Besides, 8 metabolomics studies were targeted and 6 were untargeted (Fig. 2b). Twelve studies used liquid chromatography–mass spectrometry (LC–MS) and the others used gas chromatography–mass spectrometry (GC–MS) (Fig. 2c). As for sample sizes, the majority of the studies ranged from 50 to 100 subjects (Fig. 2d).

Fig. 1
figure 1

Flow diagram of literature search and study selection for metabolite markers of ICP

Table 1 Characteristics of the 14 Included Studies
Fig. 2
figure 2

Numbers of the included studies according to biospecimen (a), targeted/untargeted (b), analytic platform (c), and sample size (d)

Analysis of metabolic biomarkers of ICP

Of the 14 studies included, only one metabolomics article used hair and did not identify statistically meaningful metabolites [14]. The other 13 found 212 metabolic biomarkers that were significantly associated with ICP. In addition, since most studies focused on the metabolic profile of bile acids, the high-frequency biomarkers (reported in ≥ 4 studies) involved were limited and were all bile acids. Glycocholic acid (GCA) was totally reported ten times, which was the most frequently reported metabolite. Interestingly, except for taurochenodeoxycholic acid (TCDCA), which showed a down-regulated trend in one study [20], all other significant bile acids showed an up-regulated trend in ICP (Table 2).

Table 2 High-frequency metabolic biomarkers of ICP

Analysis of metabolic pathways

To understand the metabolic pathways that these potential biomarkers are involved in, we imported all the reported metabolites except bile acids to MetaboAnalyst for pathway analysis, as bile acid metabolism is well-known to be related to ICP. As a result, 98 non-bile acid metabolites were finally selected for the enrichment analysis. Detailed information regarding the analysis result is shown in Table 3. Two pathways were significantly enriched at the significance level of 0.05, namely, glycerophospholipid metabolism and sphingolipid metabolism, which are both lipid metabolism-related pathways (Fig. 3). The glycerophospholipid metabolism pathway includes phosphatidylcholine (PC), phosphorylcholine, and phosphatidylserine (PS), and the sphingolipid metabolism pathway contains sphingomyelin (SM) and ceramide (Cer).

Table 3 Results of the Pathway Analysis
Fig. 3
figure 3

Overview of Pathway Analysis of ICP

Predictive and diagnostic potential of metabolite markers for discriminating ICP

Five studies [9, 13, 15, 16, 18] evaluated the potential of metabolic biomarkers or biomarker panels to predict and diagnose ICP (Table 4). These studies all calculated the area under the receiver operating curve (AUC) of single metabolites, resulting AUC values ranging from 0.642 to 1.000. Besides, Cui et al. [9] and Zheng et al. [18] both used bile acids panels to predict or diagnose ICP. Adding Complementary other biomarkers to bile acids was also shown to be effective in Dong et al. and Ma et al.’s studies [13, 16]. In addition, Dong et al. [13] indicated that metabolites at different stages of pregnancy had different predictive and diagnostic abilities.

Table 4 Potential of metabolic markers for the prediction or diagnosis of ICP

Discussion

In this study, 14 metabolomics studies on ICP were comprehensively reviewed and analyzed. To identify valuable metabolic biomarkers, seven high-frequency metabolites (reported in ≥ 4 studies) were listed. Pathway analysis results indicated two metabolic pathways involved in ICP and suggested a series of metabolic dysregulations in ICP patients.

Bile acid metabolism and ICP

Several bile acids were repeatedly identified across these studies. Bile acids, the main component of bile, are steroidal C24 carboxylic acids formed from cholesterol metabolism [25]. Based on synthetic pathways, bile acids can be classified into primary bile acids (i.e., cholic acid and chenodeoxycholic acid), secondary bile acids, and tertiary bile acids [26]. Bile acids can also be divided into 2 categories: free and conjugated bile acids (conjugated with taurine or glycine) [27].

Under normal conditions, bile acids synthesized in the liver are secreted into the bile, stored in the gallbladder, reabsorbed in the intestine, and transported back to the liver. This process is known as the reabsorption of bile acids, the so-called enterohepatic circulation [28]. Therefore, almost all bile acids are sustained in the enterohepatic system and maintained “sequestered”. In ICP, due to disruption of bile acid transport, bile acids are accumulated in liver cells, increasing their flow to the maternal systemic blood circulation and elevating circulating bile acid concentration [29]. In our results, high-frequency biomarkers were almost bile acids that showed an up-regulated trend in ICP. Only 1 study suggested that the concentration of TCDCA in ICP was significantly lower than in healthy controls [20]; however, the article did not mention the ICP diagnostic criteria and sampling time, and used a new quantitative method at that time, which may lead to the inconsistence. In addition, it is not difficult to find that these high-frequency bile acids were all conjugated bile acids, including four taurine conjugated and three glycine conjugated. Conjugated bile acids are more hydrophilic and less toxic than free bile acids [30]. Extravasation of bile acids and elevation of conjugated bile acids may be maternal adaptive mechanisms during cholestasis to reduce bile acids toxicity to the liver [31]. Studies have also demonstrated that serum conjugated bile acids significantly increased in various hepatopathies [32,33,34]. In ICP, especially severe ICP, fetal complications are more closely associated with serum TBA levels. It has been reported that for every 1 μmol/L increase in serum TBA, the incidence of fetal complications (including preterm delivery, asphyxial events, and meconium staining) increases by 1–2% [35]. Therefore, the disorders of bile acid metabolism and variation in bile acid profile require further exploration of methods to reduce serum TBA concentration and improve pregnant outcomes of ICP patients.

Though serum TBA concentration has been the most commonly used criterion for the diagnosis of ICP, metabolomics may provide more specific disease information. Single [9, 13, 15, 16, 18] or panels [9, 18] of bile acids, or combining bile acids with other biomarkers [13, 16], all have the potential to effectively predict and diagnose ICP. However, the related studies generally had a small sample size and limited validation. Further studies, such as independent external cohorts with a large sample size, are needed for obtaining reliable conclusions. In addition, Dong and coworkers [13] suggested that biomarkers’ discriminating ability differed according to stages of pregnancy. Therefore, clinical applications of bile acids panels require further exploration and optimization.

Lipid metabolism and ICP

Although the changes in bile acid metabolites were the most significant, lipid alterations would also help in the understanding of ICP pathogenesis. As basic components of cellular membranes, lipids are crucial for maintaining cellular structure, function, signaling, and energy storage [36]. Therefore, disruptions of lipid metabolism and transport exert a certain effect on human disease. In a study with a sample size of 63 ICP patients [37], results showed that ICP was associated with an abnormal lipid profile: low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, total cholesterol, and other plasma lipid concentrations were all significantly changed. Similarly, a lipidomics study [38] observed that multiple lipid components had been altered in mice with alpha-naphthyl isothiocyanate-induced intrahepatic cholestasis, suggesting that lipid metabolism disorder might be the underlying pathogenesis of intrahepatic cholestasis.

PC is a structural lipid and shows a downward trend in ICP patients [17]. Disorders in the secretion of PC into bile, as well as a significant decrease in phospholipid concentrations in bile, can lead to cholangiocyte luminal membrane injury and biliary lesions that cause cholestasis [39]. PS is also a glycerophospholipid, and its metabolism is associated with ATP8B1, which acts as the flippase for PS [40]. Studies have demonstrated that mutations in ATP8B1 could cause cholestatic disease [41] and increase the risk of ICP [42]. The pathogenic mechanism lies in that the mutations in ATP8B1 could lead to the loss of phospholipid asymmetry and subsequently undermined bile salt transport [43].

SM is a type of sphingolipid found in animal cell membranes and usually consists of phosphorylcholine and Cer. Serving as modulators of liver regeneration, sphingolipids and their enzymes play a key role in repairing liver injury [44]. Cer is one of the hydrolysis byproducts of SM by the enzyme sphingomyelinase. A study [45] focusing on the role of sphingolipids in the pathogenesis of intrahepatic cholestasis found that the levels of Cers were significantly elevated in the ICP group, and Cers could be potentially used as early biomarkers of ICP. An animal experiment [46] noticed that Cer/SM imbalance would promote lipid metabolism disorder and apoptosis and gradually cause liver injury. Thus, there is a strong relationship between SM and Cer, and the changes in their levels can reflect the health status of human live, which can be regarded as a potential therapeutic target of ICP.

Limitations of current metabolomics studies on ICP

Several limitations of the existing metabolomics studies on ICP should be noted. First, the majority of subjects were recruited in China, which may result in the limitation on study population. Moreover, most researchers [9, 12, 15, 18,19,20, 23, 24] focused on bile acid metabolism. Second, many studies possessed relatively small sample sizes, which may influence statistical power and the credibility of their research results. Third, when researchers used biofluids as samples, profiling of metabolites may be greatly dynamic and influenced by multiple factors including diet, immune status, lifestyle, and so on [47]. For example, bile acid profiles often varied from fasting to non-fasting conditions. In addition, the metabolomics on ICP is still in the preliminary stage of development. Before translating the results into clinical practice, more independent validations are needed. Sufficient external cohorts or animal/cell experiments are necessary to verify, complement and deepen current findings. Finally, the integration of metabolomics with other omics (e.g., genomics, transcriptomics, and proteomics) may help researchers to obtain a comprehensive understanding of the complexity of ICP.

Conclusions

To sum up, this study conducted a systematic review and analysis of metabolomics research on different aspects of ICP. Except for bile acid metabolism, glycerophospholipid metabolism and sphingolipid metabolism were suggested to be changed in patients with ICP. The reported metabolic biomarkers suggested potential applications of metabolomics in the clinical prediction and diagnosis of ICP. Therefore, more comprehensive and improved metabolomic studies should be encouraged to provide more valuable information for the exploration and understanding of ICP.

Availability of data and materials

Not applicable.

Abbreviations

ICP:

Intrahepatic cholestasis of pregnancy

TBA:

Total bile acid

LC–MS:

Liquid Chromatography–mass spectrometry

GC–MS:

Gas chromatography–mass spectrometry

GCA:

Glycocholic acid

TCDCA:

Taurochenodeoxycholic acid

PC:

Phosphatidylcholine

PS:

Phosphatidylserine

SM:

Sphingomyelin

Cer:

Ceramide

AUC:

Area under the receiver operating curve

References

  1. Ovadia C, Seed PT, Sklavounos A, Geenes V, Di Ilio C, Chambers J, et al. Association of adverse perinatal outcomes of intrahepatic cholestasis of pregnancy with biochemical markers: results of aggregate and individual patient data meta-analyses. Lancet. 2019;393(10174):899–909.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Smith DD, Rood KM. Intrahepatic cholestasis of pregnancy. Clin Obstet Gynecol. 2020;63(1):134–51.

    Article  PubMed  Google Scholar 

  3. Geenes V, Williamson C. Intrahepatic cholestasis of pregnancy. World J Gastroenterol. 2009;15(17):2049–66.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Geenes V, Chappell LC, Seed PT, Steer PJ, Knight M, Williamson C. Association of severe intrahepatic cholestasis of pregnancy with adverse pregnancy outcomes: a prospective population-based case-control study. Hepatology. 2014;59(4):1482–91.

    Article  PubMed  Google Scholar 

  5. Kondrackiene J, Kupcinskas L. Intrahepatic cholestasis of pregnancy-current achievements and unsolved problems. World J Gastroenterol. 2008;14(38):5781–8.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Rook M, Vargas J, Caughey A, Bacchetti P, Rosenthal P, Bull L. Fetal outcomes in pregnancies complicated by intrahepatic cholestasis of pregnancy in a Northern California cohort. PLoS ONE. 2012;7(3):e28343.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Floreani A, Caroli D, Lazzari R, Memmo A, Vidali E, Colavito D, et al. Intrahepatic cholestasis of pregnancy: new insights into its pathogenesis. J Matern Fetal Neonatal Med. 2013;26(14):1410–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Manzotti C, Casazza G, Stimac T, Nikolova D, Gluud C. Total serum bile acids or serum bile acid profile, or both, for the diagnosis of intrahepatic cholestasis of pregnancy. Cochrane Database Syst Rev. 2019;7(7):CD012546.

    PubMed  Google Scholar 

  9. Cui Y, Xu B, Zhang X, He Y, Shao Y, Ding M. Diagnostic and therapeutic profiles of serum bile acids in women with intrahepatic cholestasis of pregnancy-a pseudo-targeted metabolomics study. Clin Chim Acta. 2018;483:135–41.

    Article  CAS  PubMed  Google Scholar 

  10. Martinefski M, Contin M, Lucangioli S, Di Carlo MB, Tripodi V. In search of an accurate evaluation of intrahepatic cholestasis of pregnancy. Scientifica. 2012;2012:496489.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Pang Z, Chong J, Zhou G, de Lima Morais DA, Chang L, Barrette M, et al. MetaboAnalyst 5.0: narrowing the gap between raw spectra and functional insights. Nucleic Acids Res. 2021;49(W1):W388–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. He Y, Zhang X, Shao Y, Xu B, Cui Y, Chen X, et al. Recognition of asymptomatic hypercholanemia of pregnancy: different clinical features, fetal outcomes and bile acids metabolism from intrahepatic cholestasis of pregnancy. Biochim Biophys Acta Mol Basis Dis. 2022;1868(1):166269.

    Article  CAS  PubMed  Google Scholar 

  13. Dong R, Ye N, Zhao S, Wang G, Zhang Y, Wang T, et al. Studies on novel diagnostic and predictive biomarkers of intrahepatic cholestasis of pregnancy through metabolomics and proteomics. Front Immunol. 2021;12:733225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. de Seymour JV, Tu S, He X, Zhang H, Han TL, Baker PN, et al. Metabolomic profiling of maternal hair suggests rapid development of intrahepatic cholestasis of pregnancy. Metabolomics. 2018;14(6):79.

    Article  PubMed  CAS  Google Scholar 

  15. Li Y, Zhang X, Chen J, Feng C, He Y, Shao Y, et al. Targeted metabolomics of sulfated bile acids in urine for the diagnosis and grading of intrahepatic cholestasis of pregnancy. Genes Dis. 2018;5(4):358–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ma L, Zhang X, Pan F, Cui Y, Yang T, Deng L, et al. Urinary metabolomic analysis of intrahepatic cholestasis of pregnancy based on high performance liquid chromatography/mass spectrometry. Clin Chim Acta. 2017;471:292–7.

    Article  CAS  PubMed  Google Scholar 

  17. Sun X, Qu T, Wang W, Li C, Yang X, He X, et al. Untargeted lipidomics analysis in women with intrahepatic cholestasis of pregnancy: a cross-sectional study. BJOG. 2021;129:880.

    Article  PubMed  CAS  Google Scholar 

  18. Zheng Q, Shen L, Zhao D, Zhang H, Liang Y, Zhu Y, et al. Metabolic characteristics of plasma bile acids in patients with intrahepatic cholestasis of pregnancy-mass spectrometric study. Metabolomics. 2021;17(10):93.

    Article  CAS  PubMed  Google Scholar 

  19. Chen J, Deng W, Wang J, Shao Y, Ou M, Ding M. Primary bile acids as potential biomarkers for the clinical grading of intrahepatic cholestasis of pregnancy. Int J Gynaecol Obstet. 2013;122(1):5–8.

    Article  CAS  PubMed  Google Scholar 

  20. Ye L, Liu S, Wang M, Shao Y, Ding M. High-performance liquid chromatography-tandem mass spectrometry for the analysis of bile acid profiles in serum of women with intrahepatic cholestasis of pregnancy. J Chromatogr B Analyt Technol Biomed Life Sci. 2007;860(1):10–7.

    Article  CAS  PubMed  Google Scholar 

  21. Parizek A, Hill M, Duskova M, Vitek L, Velikova M, Kancheva R, et al. A comprehensive evaluation of steroid metabolism in women with intrahepatic cholestasis of pregnancy. PLoS ONE. 2016;11(8): e0159203.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Li GH, Huang SJ, Li X, Liu XS, Du QL. Response of gut microbiota to serum metabolome changes in intrahepatic cholestasis of pregnant patients. World J Gastroenterol. 2020;26(46):7338–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen X, Zhang X, Xu B, Cui Y, He Y, Yang T, et al. The urinary bile acid profiling analysis of asymptomatic hypercholanemia of pregnancy: a pseudo-targeted metabolomics study. Clin Chim Acta. 2019;497:67–75.

    Article  CAS  PubMed  Google Scholar 

  24. Tribe RM, Dann AT, Kenyon AP, Seed P, Shennan AH, Mallet A. Longitudinal profiles of 15 serum bile acids in patients with intrahepatic cholestasis of pregnancy. Am J Gastroenterol. 2010;105(3):585–95.

    Article  CAS  PubMed  Google Scholar 

  25. Gao J, Xu B, Zhang X, Cui Y, Deng L, Shi Z, et al. Association between serum bile acid profiles and gestational diabetes mellitus: a targeted metabolomics study. Clin Chim Acta. 2016;459:63–72.

    Article  CAS  PubMed  Google Scholar 

  26. Di Ciaula A, Garruti G, Lunardi Baccetto R, Molina-Molina E, Bonfrate L, Wang DQH, et al. Bile acid physiology. Ann Hepatol. 2017;16:S4–14.

    Article  PubMed  CAS  Google Scholar 

  27. Yang T, Shu T, Liu G, Mei H, Zhu X, Huang X, et al. Quantitative profiling of 19 bile acids in rat plasma, liver, bile and different intestinal section contents to investigate bile acid homeostasis and the application of temporal variation of endogenous bile acids. J Steroid Biochem Mol Biol. 2017;172:69–78.

    Article  CAS  PubMed  Google Scholar 

  28. Chiang JY. Bile acid metabolism and signaling. Compr Physiol. 2013;3(3):1191–212.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Gabzdyl EM, Schlaeger JM. Intrahepatic cholestasis of pregnancy: a critical clinical review. J Perinat Neonatal Nurs. 2015;29(1):41–50.

    Article  PubMed  Google Scholar 

  30. Li M, Cai SY, Boyer JL. Mechanisms of bile acid mediated inflammation in the liver. Mol Aspects Med. 2017;56:45–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li T, Chiang JYL. Bile acid-induced liver injury in cholestasis. In: Ding W-X, Yin X-M, editors. Cellular injury in liver diseases. Cham: Springer International Publishing; 2017. p. 143–72.

    Chapter  Google Scholar 

  32. Wang X, Xie G, Zhao A, Zheng X, Huang F, Wang Y, et al. Serum bile acids are associated with pathological progression of hepatitis B-induced cirrhosis. J Proteome Res. 2016;15(4):1126–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sydor S, Best J, Messerschmidt I, Manka P, Vilchez-Vargas R, Brodesser S, et al. Altered microbiota diversity and bile acid signaling in cirrhotic and noncirrhotic NASH-HCC. Clin Transl Gastroenterol. 2020;11(3):e00131.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Zhang K, Yao Y, Wang M, Liu F, Wang Q, Ma H, et al. A UPLC-MS/MS-based metabolomics analysis of the pharmacological mechanisms of rabdosia serra against cholestasis. Phytomedicine. 2021;91:153683.

    Article  CAS  PubMed  Google Scholar 

  35. Glantz A, Marschall HU, Mattsson LA. Intrahepatic cholestasis of pregnancy: Relationships between bile acid levels and fetal complication rates. Hepatology. 2004;40(2):467–74.

    Article  PubMed  Google Scholar 

  36. Kloska A, Wesierska M, Malinowska M, Gabig-Ciminska M, Jakobkiewicz-Banecka J. Lipophagy and lipolysis status in lipid storage and lipid metabolism diseases. Int J Mol Sci. 2020;21(17):6113.

    Article  CAS  PubMed Central  Google Scholar 

  37. Dann AT, Kenyon AP, Wierzbicki AS, Seed PT, Shennan AH, Tribe RM. Plasma lipid profiles of women with intrahepatic cholestasis of pregnancy. Obstet Gynecol. 2006;107(1):106–14.

    Article  CAS  PubMed  Google Scholar 

  38. Wang B-L, Zhang C-W, Wang L, Tang K-L, Tanaka N, Gonzalez FJ, et al. Lipidomics reveal aryl hydrocarbon receptor (Ahr)-regulated lipid metabolic pathway in alpha-naphthyl isothiocyanate (ANIT)-induced intrahepatic cholestasis. Xenobiotica. 2019;49(5):591–601.

    Article  CAS  PubMed  Google Scholar 

  39. Erlinger S. Low phospholipid-associated cholestasis and cholelithiasis. Clin Res Hepatol Gastroenterol. 2012;36(Suppl 1):S36-40.

    Article  CAS  PubMed  Google Scholar 

  40. Groen A, Kunne C, Jongsma G, Van Den Oever K, Mok KS, Petruzzelli M, et al. Abcg5/8 independent biliary cholesterol excretion in Atp8b1-deficient mice. Gastroenterology. 2008;134(7):2091–100.

    Article  CAS  PubMed  Google Scholar 

  41. Folmer DE, van der Mark VA, Ho-Mok KS, Elferink R, Paulusma CC. Differential effects of progressive familial intrahepatic cholestasis type 1 and benign recurrent intrahepatic cholestasis type 1 mutations on canalicular localization of ATP8B1. Hepatology. 2009;50(5):1597–605.

    Article  CAS  PubMed  Google Scholar 

  42. Horgan RP, Broadhurst DI, Walsh SK, Dunn WB, Brown M, Roberts CT, et al. Metabolic profiling uncovers a phenotypic signature of small for gestational age in early pregnancy. J Proteome Res. 2011;10(8):3660–73.

    Article  CAS  PubMed  Google Scholar 

  43. Paulusma CC, Groen A, Kunne C, Ho-Mok KS, Spijkerboer AL, Rudi de Waart D, et al. Atp8b1 deficiency in mice reduces resistance of the canalicular membrane to hydrophobic bile salts and impairs bile salt transport. Hepatology. 2006;44(1):195–204.

    Article  CAS  PubMed  Google Scholar 

  44. Nojima H, Freeman CM, Gulbins E, Lentsch AB. Sphingolipids in liver injury, repair and regeneration. Biol Chem. 2015;396(6–7):633–43.

    Article  CAS  PubMed  Google Scholar 

  45. Mikucka-Niczyporuk A, Pierzynski P, Lemancewicz A, Kosinski P, Charkiewicz K, Knas M, et al. Role of sphingolipids in the pathogenesis of intrahepatic cholestasis. Prostaglandins Other Lipid Mediat. 2020;147:106399.

    Article  CAS  PubMed  Google Scholar 

  46. Su D, Liao Z, Feng B, Wang T, Shan B, Zeng Q, et al. Pulsatilla chinensis saponins cause liver injury through interfering ceramide/sphingomyelin balance that promotes lipid metabolism dysregulation and apoptosis. Phytomedicine. 2020;76:153265.

    Article  CAS  PubMed  Google Scholar 

  47. Souza RT, Mayrink U, Leite DF, Costa ML, Calderon IM, Rocha Filho EA, et al. Metabolomics applied to maternal and perinatal health: a review of new frontiers with a translation potential. Clinics. 2019;74:e894.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (grant: 82273635).

Author information

Authors and Affiliations

Authors

Contributions

ZY: writing—original draft; MY: writing—original draft; XH: review and editing; YZ: review and editing; CZ: review and editing; XX: conceptualization, supervision, writing—review and editing; JY: conceptualization, supervision, writing—review and editing. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Xiangxiang Xu or Jieyun Yin.

Ethics declarations

Ethical approval and consent to participate

Ethical approval was not required, because the analysis under consideration is from data already publicly available in published studies.

Consent for publication

The author consents to the publication of the manuscript in European Journal of Medical Research.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, Z., Yao, M., Zhang, C. et al. Application of metabolomics in intrahepatic cholestasis of pregnancy: a systematic review. Eur J Med Res 27, 178 (2022). https://doi.org/10.1186/s40001-022-00802-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-022-00802-z

Keywords