Comprehensive Analysis of Quantitative Proteomics With DIA Mass Spectrometry and ceRNA Network in Intrahepatic Cholestasis of Pregnancy

Background: Intrahepatic cholestasis of pregnancy (ICP) is a pregnancy-specific complication characterized by pruritus without skin damage and jaundice. The poor perinatal outcomes include fetal distress, preterm birth, and unexpected intrauterine death. However, the mechanism of ICP leading to poor prognosis is still unclear. Methods: We analyzed 10 ICP and 10 normal placental specimens through quantitative proteomics of data-independent acquisition (DIA) to screen and identify differentially expressed proteins. GO, KEGG, COG/KOG, StringDB, InterProScan, Metascape, BioGPS, and NetworkAnalyst databases were used in this study. PITA, miRanda, TargetScan, starBase, and LncBase Predicted v.2 were used for constructing a competing endogenous RNA (ceRNA) network. Cytoscape was used for drawing regulatory networks, and cytoHubba was used for screening core nodes. The ICP rat models were used to validate the pathological mechanism. Results: GO, KEGG, and COG/KOG functional enrichment analysis results showed the differentially expressed proteins participated in autophagy, autophagosome formation, cofactor binding, JAK-STAT signaling pathway, and coenzyme transport and metabolism. DisGeNET analysis showed that these differentially expressed proteins were associated with red blood cell disorder and slow progression. We further analyzed first 12 proteins in the upregulated and downregulated differentially expressed proteins and incorporated clinicopathologic parameters. Our results showed HBG1, SPI1, HBG2, HBE1, FOXK1, KRT72, SLC13A3, MBD2, SP9, GPLD1, MYH7, and BLOC1S1 were associated with ICP development. ceRNA network analysis showed that MBD2, SPI1, FOXK1, and SLC13A3 were regulated by multiple miRNAs and lncRNAs. Conclusion: ICP was associated with autophagy. The ceRNA network of MBD2, SPI1, FOXK1, and SLC13A3 was involved in ICP progression, and these core proteins might be potential target.

[1]  K. Rood,et al.  Intrahepatic cholestasis of pregnancy: risk factors for severe disease , 2021, The journal of maternal-fetal & neonatal medicine : the official journal of the European Association of Perinatal Medicine, the Federation of Asia and Oceania Perinatal Societies, the International Society of Perinatal Obstetricians.

[2]  Bowen Li,et al.  Expression, regulation, and function of exosome‐derived miRNAs in cancer progression and therapy , 2021, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[3]  Yayi Hu,et al.  Current understanding of autophagy in intrahepatic cholestasis of pregnancy. , 2021, Placenta.

[4]  Xiaoqian Zhang,et al.  Long non-coding RNA Xist regulates oocyte loss via suppressing miR-23b-3p/miR-29a-3p maturation and upregulating STX17 in perinatal mouse ovaries , 2021, Cell Death & Disease.

[5]  Xiaohuan Yuan,et al.  LncRNA XIST serves as a diagnostic biomarker in gestational diabetes mellitus and its regulatory effect on trophoblast cell via miR-497-5p/FOXO1 axis. , 2021, Cardiovascular diagnosis and therapy.

[6]  Minjian Chen,et al.  Diagnostic and Prognostic Value of Long Noncoding RNAs as Potential Novel Biomarkers in Intrahepatic Cholestasis of Pregnancy , 2021, BioMed research international.

[7]  A. Kourtidis,et al.  LNCcation: lncRNA localization and function , 2021, The Journal of cell biology.

[8]  Jinhong Li,et al.  An overview of autophagy: Mechanism, regulation and research progress. , 2021, Bulletin du cancer.

[9]  J. Fleckenstein,et al.  Intrahepatic Cholestasis of Pregnancy: Natural History and Current Management , 2021, Seminars in Liver Disease.

[10]  Nadezhda T. Doncheva,et al.  The STRING database in 2021: customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets , 2020, Nucleic Acids Res..

[11]  Dorte B. Bekker-Jensen,et al.  Data Processing and Analysis for DIA-Based Phosphoproteomics Using Spectronaut. , 2021, Methods in molecular biology.

[12]  S. Joshi,et al.  Cell death mechanisms and their roles in pregnancy related disorders. , 2021, Advances in protein chemistry and structural biology.

[13]  F. Jin,et al.  A novel autophagy‐related lncRNA prognostic risk model for breast cancer , 2020, Journal of cellular and molecular medicine.

[14]  R. Xu,et al.  LncRNA‐mediated posttranslational modifications and reprogramming of energy metabolism in cancer , 2020, Cancer communications.

[15]  C. Yuan,et al.  Oncogenic LncRNA CASC9 in Cancer Progression. , 2020, Current pharmaceutical design.

[16]  P. Middleton,et al.  Pharmacological interventions for treating intrahepatic cholestasis of pregnancy. , 2020, The Cochrane database of systematic reviews.

[17]  Lin Zhang,et al.  LncRNA: A Potential Research Direction in Intestinal Barrier Function , 2020, Digestive Diseases and Sciences.

[18]  Tianyu Jiang,et al.  Physiological and pathological regulation of autophagy in pregnancy , 2020, Archives of Gynecology and Obstetrics.

[19]  Alex Alves Freitas,et al.  Comparing enrichment analysis and machine learning for identifying gene properties that discriminate between gene classes , 2020, Briefings Bioinform..

[20]  Shi-kun He,et al.  Autophagy and autophagy-related proteins in cancer , 2020, Molecular Cancer.

[21]  Howard Y. Chang,et al.  Structural modularity of the XIST ribonucleoprotein complex , 2019, Nature Communications.

[22]  Peiyue Jiang,et al.  MicroRNAs derived from urinary exosomes act as novel biomarkers in the diagnosis of intrahepatic cholestasis of pregnancy. , 2019, American journal of translational research.

[23]  Othman Soufan,et al.  NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis , 2019, Nucleic Acids Res..

[24]  T. Kawakita,et al.  Association of adverse perinatal outcomes of intrahepatic cholestasis of pregnancy with biochemical markers: results of aggregate and individual patient data meta-analyses , 2019, The Lancet.

[25]  Minjian Chen,et al.  The Serum microRNA Profile of Intrahepatic Cholestasis of Pregnancy: Identification of Novel Noninvasive Biomarkers , 2018, Cellular Physiology and Biochemistry.

[26]  Li'na Hu,et al.  Linc02527 promoted autophagy in Intrahepatic cholestasis of pregnancy , 2018, Cell Death & Disease.

[27]  Xuan Huang,et al.  Placental Expressions of CDKN1C and KCNQ1OT1 in Monozygotic Twins with Selective Intrauterine Growth Restriction , 2017, Twin Research and Human Genetics.

[28]  Yi-Ling Ding,et al.  miR-148a-mediated estrogen-induced cholestasis in intrahepatic cholestasis of pregnancy: Role of PXR/MRP3 , 2017, PloS one.

[29]  Y. Le Bouc,et al.  Imprinted disorders and growth. , 2017, Annales d'endocrinologie.

[30]  Ting Yang,et al.  Feasibility of urinary microRNA profiling detection in intrahepatic cholestasis of pregnancy and its potential as a non-invasive biomarker , 2016, Scientific Reports.

[31]  Robert Powers,et al.  PCA as a practical indicator of OPLS-DA model reliability. , 2016, Current Metabolomics.

[32]  M. Karin,et al.  Autophagy, Inflammation, and Immunity: A Troika Governing Cancer and Its Treatment , 2016, Cell.

[33]  C. Williamson,et al.  The pathophysiology of intrahepatic cholestasis of pregnancy. , 2016, Clinics and research in hepatology and gastroenterology.

[34]  Chunlei Wu,et al.  BioGPS: building your own mash-up of gene annotations and expression profiles , 2015, Nucleic Acids Res..

[35]  Ling Yu,et al.  Human leukocyte antigen G and miR-148a are associated with the pathogenesis of intrahepatic cholestasis of pregnancy , 2014, Experimental and therapeutic medicine.

[36]  I. Netchine,et al.  Beckwith–Wiedemann and Russell–Silver Syndromes: from new molecular insights to the comprehension of imprinting regulation , 2014, Current opinion in endocrinology, diabetes, and obesity.

[37]  T. Pusl,et al.  Intrahepatic cholestasis of pregnancy , 2007, Orphanet journal of rare diseases.

[38]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[39]  M. Biteker Current understanding , 1990, The Lancet.