Tian Lan’s research while affiliated with Charité Universitätsmedizin Berlin and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (30)


FRI-301 Reactive cholangiocyte-derived orosomucoid-2 (ORM2) drives a pathogenic modulation of the injured biliary niche through macrophage reprogramming
  • Article

May 2025

·

8 Reads

Journal of Hepatology

·

·

Tian Lan

·

[...]

·

Adrien Guillot

Figure 1 ORM2 is upregulated in cholangiocytes but downregulated in hepatocytes during liver injury. (A) Ckb, Fam166b, Onecut1, Orm2 and Adam11 were upregulated in both acutely (ihCD59 BEG-TG mice challenged by acute BEC injury for 48 hours, data set ID: PRJNA510784) and chronically (32-week-old dnTGFRII mice, data set ID: GSE202676; 29-week HFD fed mice, data set ID: GSE217739) injured mouse BECs. Upregulated DEGs were identified as log 2 (fold change) >0.5, p<0.05. (B) Gene expression changes of Adam11 and Orm2 in injured livers from diverse mouse models (injured vs healthy) and human samples (diseases vs healthy). Bulk RNA sequencing data was obtained from the GepLiver database and GEO data sets. (C) Gene expression changes of Orm2 in single-cell resolved human liver samples with MASLD, liver cirrhosis and PSC. scRNA-seq and snRNAseq data were obtained from the GepLiver database and GEO data sets (ID: GSE247128 and GSE243977). 'X' in the block represents undetectable data. (D) Immunofluorescent staining illustrated the associations among hepatocytes (HepPar1 + ), BECs (CK7 + ), monocytes/macrophages (IBA1 + ) and ORM2 expression in human liver samples (normal, MALSD and PSC). Relative ORM2 staining was illustrated with highlighted portal areas. (E) ORM2 expression in single hepatocytes and BECs in human liver samples (normal, MALSD and PSC) was assessed according to the immunofluorescent staining analyses. (F) Significant correlations (Spearman's, p<0.05) among staining intensity of ORM2, CK7 and CK19, neighbouring IBA1 + cells (<100 μm) and minimum distance to IBA1 + cells were illustrated in a heatmap. (G) Gene expression of Orm2 in WT mouse primary BECs on biliatresone and CDCA treatments (vs untreated). (H) Protein expression of ORM2 in mouse primary BECs on biliatresone illustrated in fluorescent staining and (I) quantitative analysis. (J) Gene expression of Orm2 in WT mouse primary hepatocytes on FFAs, APAP and biliatresone treatments (vs untreated). (K) Protein expression of ORM2 in mouse primary hepatocytes on biliatresone illustrated in fluorescent staining and (L) quantitative analysis. Sample sizes: (D-F) n=1-4 per group; (G-K) n=4 per group. APAP, acetaminophen; B/(m)BEC, (mouse) biliary epithelial cell; BSA, bovine serum albumin; CDCA, chenodeoxycholic acid; DDC, 3,5-diethoxycarbonyl-1,4-dihydrocollidine; DEG, differentially expressed gene; F0/2/4, fibrosis stage-0/2/4; FFAs, free fatty acids; H/(m)HepaC, (mouse) hepatocyte; HFD, high-fat diet; IntraC/ExtraC, intracellular/extracellular; MASLD, metabolic dysfunction associated steatotic liver diseases; MASH, metabolic dysfunction associated steatohepatitis; PSC, primary sclerosing cholangitis; OA, oleic acid; ORM2, orosomucoid 2; sc/snRNA, single-cell/single-nuclei RNA; WT, wild-type. One-way analysis of variance followed by Tukey's multiple comparison test and unpaired Student's t-tests were performed. *p<0.05 as indicated or as compared with controls.
Figure 4 ORM2 reshapes transcriptome profiles in liver macrophages. (A) Gene expression of macrophage phenotype-associated markers was measured in human THP-1 cell line, hMoMF, mLMF and mBMDM (ORM2-treated vs Ctrl). (B) Significantly upregulated and downregulated DEGs in mLMF by bulk RNA-seq analysis were illustrated in the volcano plot (ORM2-treated vs Ctrl). (C) Function enrichment analysis (GO-BP and KEGG) on significantly upregulated and downregulated DEGs in mLMF by bulk RNA-seq analysis was illustrated (ORM2-treated vs Ctrl). (D) Gene expression changes of pro-inflammatory and anti-inflammatory macrophage phenotype-associated markers in mLMF by bulk RNA-seq analysis were illustrated in the radar plot (ORM2-treated vs Ctrl). (E) Overlapped significantly upregulated DEGs in mLMF from diverse bulk RNA-seq data sets (GSE273509 and GSE86397) were illustrated (vs Ctrl). Sample sizes: (A) n=4 per group; (B-E) ORM2-treated (n=4) versus Ctrl (n=5). DEG, differentially expressed gene; GO-BP, Gene Ontology-biological processes; hMoMF, human monocyte-derived macrophages; KEGG, Kyoto Encyclopedia of Genes and Genomes; mLMF, mouse liver macrophages; mBMDM, mouse bone marrow-derived macrophages; mRNA, messenger RNA; ORM2, orosomucoid 2; RNA-seq, RNA sequencing. Unpaired Student's t-tests were used. *Represents p<0.05. Protected by copyright, including for uses related to text and data mining, AI training, and similar technologies.
Figure 6 ORM2 promotes phagocytosis, lipid deposition and cell stress in liver macrophages. (A) Gene expression of Cd163, Timd4, Cyp2e1, Cd36, Cdkn1a and Casp1 in mLMF on ORM2 treatments (0, 0.5, 1.0, 2.0 and 4.0 μg/mL) was measured (ORM2-treated vs Ctrl). (B) Phagocytosis/bead capture (YG + ) of mLMF on Ctrl, ORM2 and LPS treatments was illustrated in fluorescent staining and (C) quantitative analysis. (D) Protein expression of TIM4 in mLMF on Ctrl, ORM2 and LPS treatments was illustrated in fluorescent staining and (E) quantitative analysis. (F) Lipid deposition of mLMF on Ctrl, ORM2, FFAs and FFAs+ORM2 treatments was illustrated in fluorescent staining and (G) quantitative analysis. (H) Protein expression of CD36 in mLMF on Ctrl, ORM2 and LPS treatments was assessed by western blot in two independent experiments (both displayed) and (I) quantitative analysis was performed. (J) DNA damage (Sa-β-Gal + ) of mLMF on Ctrl, ORM2 and LPS treatments was illustrated in fluorescent staining and (K) quantitative analysis. (L) Cell apoptosis (Apopxin + ) of mLMF on Ctrl, ORM2 and LPS treatments was illustrated in fluorescent staining and (M) quantitative analysis. Sample sizes: n=3-4 per group. CD36, cluster of differentiation 36; FFAs, free fatty acids; ITPR2, inositol 1,4,5-trisphosphate receptor type 2; LPS, lipopolysaccharides; mLMF, mouse liver macrophages; ORM2, orosomucoid 2; SA-β-Gal, senescence-associated betagalactosidase; TIM4, T-cell/transmembrane immunoglobulin and mucin domain containing 4; YG, yellow green fluorescence. One-way analysis of variance followed by Tukey's multiple comparison tests were performed. *p<0.05 as indicated or as compared with controls.
Figure 7 Multiplex immunofluorescence and single-cell transcriptomics reveal heterogeneous ITPR2 expression in liver macrophages during liver diseases. (A) Multiplex immunofluorescence was applied to identify hepatocytes (HepPar1 + ), cholangiocytes (CK7 + ) and macrophages (IBA1 + ), together with ITPR2 expression in human normal, PSC, MASLD-F2 and MASH-F4 liver samples. Red arrows point toward ITPR2-expressing macrophages. (B) Large scan multiplex immunofluorescence images shown in (A) were used for CK7 + and IBA1 + cell distribution analysis (heatmaps), and singular IBA1 + cell relative staining intensities for ITPR2, CD16 and CD163 (dot plots, green: high staining intensity, blue: low staining intensity). (C) Spearman's correlation was used to evaluate the correlation between single cell immunostaining intensities of the indicated proteins in IBA1 + cells present in the images shown in panels A and B. All markers showed significant correlations. Differential gene expressions were investigated in the indicated data sets for ITPR2, CALM1 and CALM2 in monocyte and macrophage populations in human (D) MASH, (E) PSC and (F) liver cirrhosis (all vs healthy). F2/4, fibrosis stage 2/4; KC, Kupffer cells; ITPR2, inositol 1,4,5-trisphosphate receptor type 2; MASH, metabolic dysfunction-associated steatohepatitis; MASLD, metabolic dysfunction-associated steatotic liver disease; MF, macrophages; Mono: monocytes; PSC, primary sclerosing cholangitis; sc/snRNA-seq, single cell/single nuclei RNA sequencing. (D-F) Unpaired Student's t-tests were used. *Represents p<0.05.
Reactive cholangiocyte-derived ORM2 drives a pathogenic modulation of the injured biliary niche through macrophage reprogramming
  • Article
  • Full-text available

April 2025

·

150 Reads

·

4 Citations

Gut

Background Injured or reactive biliary epithelial cells participate in most chronic liver injuries in a process referred to as ductular reaction, which involves multicellular interactions with marked local infiltration of macrophages and fibrogenic cell activation. The direct roles of biliary epithelial cells in shaping their cellular niche remain unknown. Objective We aimed at investigating the effects of biliary epithelial cell-derived acute phase response protein orosomucoid 2 (ORM2) in shaping monocyte/macrophage response to liver injury. Design Transcriptome data sets from human and mouse livers were used, results were confirmed with multiplex immunofluorescence. A multicellular biliary-niche-on-a-chip derived from primary liver and blood cells (wild-type, Mdr2 −/− mice) was established to model ductular reaction. Human blood cells collected from healthy donors and intrahepatic cholangiocyte organoids derived from normal and cirrhotic liver patients were used. Results Our transcriptome data set and multiplex immunofluorescence analyses indicated a previously unrecognised involvement of the acute phase response protein ORM2 in ductular reactions in both human and mouse livers. ORM2 gene expression was increased in biliatresone-challenged, bile acid-challenged and acetaminophen-challenged cholangiocytes. Cholangiocyte-derived ORM2 induced unique transcriptome changes and functional adaptation of liver macrophages. ORM2-activated macrophages exacerbated cholangiocyte cell stress and Orm2 expression, but also tended to promote fibrogenic activation of hepatic stellate cells. Mechanistically, ORM2 effects were mediated by an inositol 1,4,5-trisphosphate receptor type 2-dependent calcium pathway. Conclusion This study reveals a paracrine communication circuit during ductular reaction, in which reactive cholangiocyte-derived ORM2 reprogrammes liver macrophages, participating in a pathogenic remodelling of the immune biliary niche.

Download

HSCs regulate liver regeneration and injury
a, Lrat-cre⁺TdTom⁺ mice expressing iDTR (iDTRhet) or not (iDTRWT) were injected with diphtheria toxin (DT). The TdTom⁺ area (n = 6 (iDTRWT) and n = 5 (iDTRhet)), Lrat and Ccnd1 mRNA (by qPCR, n = 8 (iDTRWT) and n = 11 (iDTRhet)) and the liver–body weight ratio (n = 8 (iDTRWT) and n = 8 (iDTRhet)) were determined 7 days later. b,c, iDTRWT and iDTRhet mice were treated with DT and, 1 week later, were subjected to 70% PHx (n = 7 per group) (b) or treatment with constitutive androstane receptor agonist (c), followed by Ki-67 IHC and quantification per high-power field (HPF) (n = 8 (iDTRWT) and n = 11 (iDTRhet)) as well as qPCR analysis of Mki67 (n = 8 (iDTRWT) and n = 10 (iDTRhet)). d, iDTRWT and iDTRhet mice were treated with DT. Then, 1 week later, the mice were subjected to treatment with a sublethal dose of APAP to determine the serum ALT and necrosis area in haematoxylin and eosin (H&E) sections (n = 6 (iDTRWT) and n = 4 (iDTRhet)), or with a lethal APAP dose to determine survival (n = 4 (iDTRWT) and n = 7 (iDTRhet)). e,f, iDTRWT and iDTRhet mice (n = 5 per group) were treated with DT and 1 week later were then treated with CCl4 (e; 0.5 mg per kg, n = 5 per group) or allyl alcohol (f; 60 mg per kg) to determine the serum ALT and necrosis area in H&E sections (n = 8 (iDTRWT) and n = 7 (iDTRhet)) or a lethal dose of allyl alcohol (f; 75 mg per kg; n = 10 (iDTRWT) and n = 12 (iDTRhet)) to determine survival. g,h, Primary mouse hepatocytes (Hep) were co-cultured with or without primary mouse HSCs in a contact-dependent (g; EdU, n = 6 per group; Mki67 mRNA, n = 4 per group) or contact-independent (h; EdU, n = 6 per group; qPCR, n = 3 (hepatocytes), n = 4 (hepatocytes + HSCs)) manner to determine proliferation based on EdU staining and qPCR analysis of Mki67 mRNA. Data are mean ± s.e.m. For a–h, each dot represents one biological replicate. Scale bars, 100 µm (a–h). P values were calculated using unpaired two-tailed t-tests (a–c, e, g and h, and d and f (middle and left)) or log-rank test (d and f (right)).
Source data
HSCs regulate metabolic zonation and zone-specific injury and proliferation in the liver
a,b, The top 40 genes downregulated in RNA-seq data from HSC-depleted mice versus controls (ctrl) in the iDTR × Lrat-cre and the JEDI models versus controls after subtraction of HSC-enriched genes (a), and qPCR confirmation in iDTRWT (n = 8) and iDTRhet (n = 11) mice (b) of select genes in livers from the iDTR × Lrat-cre model. c, CYP2E1, CYP1A2 and CYP2F2 IHC and quantification in iDTRWT (n = 8) and iDTRhet (n = 8) mice 7 days after treatment with diphtheria toxin. c, central vein; p, portal vein. d, Multiplex IHC analysis showing significantly altered expression of zonal genes in iDTRWT (n = 5) and iDTRhet (n = 5) mice. e, Zonal quantification of the indicated zone 1 (Zo1), zones 2–3 and strictly zone 3 markers from IHC performed in Fig. 2c and Extended Data Fig. 4g in iDTRWT and iDTRhet mice (n = 8 per group). f, 100-plex spatial transcriptomics for WNT-regulatory, WNT-target and cell marker genes shows differences in zonation patterns and WNT-target genes between iDTRWT (n = 1) versus iDTRhet (n = 1) mice. g,h, Zonal quantification of Ki-67⁺ cells after 70% PHx and TCPOBOP treatment (g) or of necrosis after APAP, CCl4 or allyl alcohol treatment (h) in iDTRWT (n = 5–8) and iDTRhet (n = 4–11) mice. Data are mean ± s.e.m. For b and c, each dot represents one biological replicate. Scale bars, 100 µm (c and d) and 1 mm (f). P values were calculated using unpaired two-tailed t-tests (b, c, e, g and h). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. AU, arbitrary units.
Source data
HSC-derived RSPO3 regulates hepatocyte gene expression and liver zonation
a, GSEA of CTNNB1-regulated genes from RNA-seq data of HSC-depleted (JEDI) versus control livers; and a heat map showing the expression (Exp) of the top 15 downregulated genes from Ctnnb1ΔHep versus Ctnnb1fl/fl livers in HSC-depleted versus control livers. b, CellPhoneDB analysis showing the top HSC–hepatocyte ligand–receptor interactions in healthy mouse liver snRNA-seq data. n = 2 livers. c, snRNA-seq analysis of Rspo3 expression in healthy mouse liver. n = 2. d, RNAscope analysis of Rspo3 colocalization with TdTom⁺ HSCs in Lrat-cre × TdTom livers. A representative image of two technical replicates is shown. e, RSPO3 ELISA in the supernatants from primary mouse HSCs, ECs, Kupffer cells (KCs) and hepatocytes. n = 3 per group. f, Analysis of Rspo3 expression in HSCs across mouse liver zones using 100-plex spatial transcriptomics data. g, The liver–body weight ratio and qPCR analysis of Rspo3 mRNA (n = 4 per group) in HSCs from Rspo3fl/fl (n = 8) and Rspo3ΔHSC (n = 8, 7 male, 1 female) mice. h,i, The indicated WNT-target genes determined by qPCR (h) or IHC with morphometric and zone-specific quantification (i) in Rspo3fl/fl (n = 8) and Rspo3ΔHSC (n = 8, 7 male, 1 female) livers. j, Analysis of Rspo3 expression in ECs across mouse liver zones using 100-plex spatial transcriptomics data. k, Rspo3 mRNA in isolated ECs (n = 4 per group), and the liver–body weight ratio in Rspo3fl/fl (n = 6) and Rspo3ΔEC (n = 8) mice. l,m, WNT-target genes determined by qPCR (l), and IHC analysis with morphometric and zone-specific quantification (m) in Rspo3fl/fl (n = 6) and Rspo3ΔEC (n = 8) livers. Data are mean ± s.e.m. Each dot represents one cell (c) or one biological replicate (g–i and k–m). For d, i and m, scale bars, 100 µm. For the violin plots in f and j, the box plots show the interquartile range (IQR; Q1–Q3) (box limits), the median (centre line), and the minimum (Q1 − 1.5 × IQR) and maximum (Q3 + 1.5 × IQR) values (whiskers). P values were calculated using unpaired two-tailed t-tests (e, g–i and k–m) or Wilcoxon rank-sum tests (f and j). UMAP, uniform manifold approximation and projection.
Source data
HSC-derived RSPO3 regulates hepatocyte injury, liver regeneration and steatosis
a, Ki-67 and cyclin D1 IHC from Rspo3fl/fl (n = 7) and Rspo3ΔHSC (n = 6) mice subjected to 70% PHx. b–d, The necrosis area (n = 5 (Rspo3fl/fl), n = 6 (Rspo3ΔHSC)), ALT levels (n = 5 (Rspo3fl/fl), n = 6 (Rspo3ΔHSC)) and survival (n = 12 (Rspo3fl/fl), n = 10 (Rspo3ΔHSC)) in Rspo3fl/fl and Rspo3ΔHSC mice treated with APAP (b; 300 mg per kg or 750 mg per kg lethal dose); the necrosis area (n = 3 per group) and ALT levels (n = 7 per group) in Rspo3fl/fl and Rspo3ΔHSC mice treated with CCl4 (c, 0.5 ml kg⁻¹); and the necrosis area (n = 8 per group), ALT levels (n = 8 per group) and survival (n = 14 (Rspo3fl/fl), n = 12 (Rspo3ΔHSC)) in Rspo3fl/fl and Rspo3ΔHSC mice treated with allyl alcohol (d; 60 mg per kg or 75 mg per kg lethal dose). e, Zonal quantification of Ki-67⁺ cells (n = 7 (Rspo3fl/fl), n = 6 (Rspo3ΔHSC)) and necrosis in APAP (n = 5 (Rspo3fl/fl), n = 6 (Rspo3ΔHSC)), CCl4 (n = 3 per group) and allyl alcohol (n = 8 per group) models in Rspo3fl/fl and Rspo3ΔHSC mice. f, Oil Red O staining and quantification, serum ALT and AST, and qPCR analysis of Aldh2 mRNA in Rspo3fl/fl (n = 11) and Rspo3ΔHSC (n = 9) mice treated with the Lieber–DeCarli diet. g, Oil Red O staining and quantification, the serum ALT and AST (n = 10 (Rspo3fl/fl), n = 11 (Rspo3ΔHSC)), representative images and the tumour number and tumour size in Rspo3fl/fl (n = 8) and Rspo3ΔHSC mice (n = 7) that were treated with CDAA-HFD diet for the indicated times. h, DESI–MS imaging showing triglycerides (TG; 52:3, red) and phosphatidylcholine (PC; 36:5, blue) in Rspo3fl/fl and Rspo3ΔHSC (n = 4 per group) mice as well as a representative for localization of TG 52:3 around pericentral zones marked by GS (green) and quantification of TG 52:3, TG 52:4 and TG 55:8 species. Data are mean ± s.e.m. Each dot represents one biological replicate (a–d and f–h). Scale bars, 100 µm (a–d, f and g), 1 cm (h, left), 1 mm (h, right). P values were calculated using unpaired two-tailed t-tests (a, c and e–h, and b and d (top)) or log-rank tests (b and d (bottom)).
Source data
Dynamic regulation of RSPO3 in liver disease
a,b, scRNA-seq analysis of Rspo3 mRNA of HSCs from CCl4-treated (a) or high-fat high-fructose diet (HF-HFD)-treated (b) mice. c, scRNA-seq analysis of Rspo3 mRNA from quiescent (qHSC), intermediate-active (imHSC), activated (actHSC) and deactivated (deactHSC) mouse HSCs (from ref. ⁶¹). d, qPCR analysis of Rspo3 in quiescent mouse HSCs treated with the indicated cytokines. n = 4 per group. e, snRNA-seq analysis of RSPO3 mRNA expression (Exp) in human liver. n = 6. f, CellPhoneDB analysis showing the top HSC–hepatocyte interactions in snRNA-seq data from human livers. g, snRNA-seq analysis of RSPO3 mRNA in human cyHSCs and myHSCs. h, qPCR analysis of RSPO3 in PDGF- and TGFβ-treated LX-2 human HSCs. n = 3 per group (control and PDGF) and n = 4 (TGFβ). i, snRNA-seq analysis of RSPO3 mRNA in HSCs from healthy control individuals (Ctrl) and patients with MASLD, alcoholic cirrhosis (Alc. cirrh.) or alcoholic hepatitis (Alc. hep.). Data are mean ± 95% confidence intervals. j, RSPO3 mRNA in different stages of MASLD and correlation with the WNT-target genes CYP2E1, CYP1A2 and TBX3 in the GSE135251 cohort. k, The correlation between HSC RSPO3 and hepatocyte CYP1A2 and CYP2E1 expression in snRNA-seq data of healthy individuals and patients with MASLD and ALD (n = 25). l, RSPO3 mRNA in different stages of ALD and survival stratified by RSPO3 expression in the dbGaP phs001807.v1.p1 ALD cohort. Non.-sev., non-severe; TPM, transcripts per million. m, Survival by RSPO3 expression in the SteatoSITE MASLD cohort. Data are mean ± s.e.m. Each dot represents one cell (a–c) or one biological replicate (d and h). ****P < 0.0001 versus control or normal; ###P < 0.001 versus F0–1. In the violin plots, the box plots show the IQR (box limits), the median (centre line), the minimum (Q1 − 1.5 × IQR) and maximum (Q3 + 1.5 × IQR) values (whiskers), and outliers (individual dots). P values were calculated using two-way analysis of variance (ANOVA) with Tukey’s multiple-comparison test (a–c, i and l (left)), one-way ANOVA with Dunnett’s multiple-comparison test (d and h), Wilcoxon rank-sum tests (j) or log-rank tests (l (right) and m). Correlations were evaluated by the Pearson correlation coefficient (j and k). MASH, metabolic dysfunction-associated steatohepatitis.
Source data
Hepatic stellate cells control liver zonation, size and functions via R-spondin 3

March 2025

·

266 Reads

·

7 Citations

Nature

Hepatic stellate cells (HSCs) have a central pathogenetic role in the development of liver fibrosis. However, their fibrosis-independent and homeostatic functions remain poorly understood1, 2, 3, 4–5. Here we demonstrate that genetic depletion of HSCs changes WNT activity and zonation of hepatocytes, leading to marked alterations in liver regeneration, cytochrome P450 metabolism and injury. We identify R-spondin 3 (RSPO3), an HSC-enriched modulator of WNT signalling, as responsible for these hepatocyte-regulatory effects of HSCs. HSC-selective deletion of Rspo3 phenocopies the effects of HSC depletion on hepatocyte gene expression, zonation, liver size, regeneration and cytochrome P450-mediated detoxification, and exacerbates alcohol-associated and metabolic dysfunction-associated steatotic liver disease. RSPO3 expression decreases with HSC activation and is inversely associated with outcomes in patients with alcohol-associated and metabolic dysfunction-associated steatotic liver disease. These protective and hepatocyte-regulating functions of HSCs via RSPO3 resemble the R-spondin-expressing stromal niche in other organs and should be integrated into current therapeutic concepts.


Microscopic changes in mouse livers and primary mouse hepatocytes
(A) Liver specimens under light and electron microscopy. Liver tissues in the first and second rows were stained with hematoxylin-eosin and Sirius red, respectively (×100 magnification). Liver tissues in the third row show hepatocyte mitochondrial changes under transmission electron microscopy (×20,000 magnification, scale bar 200 nm) (n = 6 for each group). (B) Isolated primary hepatocytes under transmission electron microscopy (First row: ×4,000 magnification, scale bar 1 µm; second row: ×20,000 magnification, scale bar 200 nm) (n = 6 for each group). CO, control; KO, knockout; NS, normal saline; TAA, thioacetamide; DMSO, dimethyl sulfoxide.
Iron metabolism in mouse livers and primary mouse hepatocytes
(A, B) Ferrous ion and hepcidin concentrations in the liver (n = 6 for each group). (C, D) The mRNA expression levels of Tfr and Fpn in primary mouse hepatocytes (n = 6 for each group). (A, B) p < 0.05: *vs. CO NS group; &vs. KO NS group; #vs. CO TAA group. (C, D) p < 0.05: *vs. DMSO group; #vs. erastin + DMSO group. Tfr, transferrin; Fpn, ferroportin; CO, control; NS, normal saline; KO, knockout; TAA, thioacetamide; DMSO, dimethyl sulfoxide.
Lipid metabolomics of mouse livers
(A–D) Heatmaps of FFAs (A), PE (B), PC (C), and PS (D) (n = 6 for each group). (E–H) Concentrations of AA (E), AdA (F), Lin (G), and FFA (C22:6) (H) (n = 6 for each group). p < 0.05: *vs. CO NS group; #vs. CO TAA group. FFA, free fatty acid; PE, phosphatidylethanolamine; PC, phosphatidylcholine; PS, phosphatidylserine; AA, arachidonic acid; AdA, adrenic acid; Lin, linoleic acid; CO, control; NS, normal saline; KO, knockout; TAA, thioacetamide.
Oxidative stress in mouse livers and primary mouse hepatocytes
(A, B) Concentrations of GSH and MDA in mouse livers (colorimetric assay, n = 6 for each group). (C) GPX4 expression in mouse livers (Western blot, n = 6 for each group). (D) Ptgs2 mRNA expression level in primary mouse hepatocytes (n = 6 for each group). (E–G) Concentrations of PGE2, GSH, and MDA in primary mouse hepatocytes (colorimetric assay, n = 6 for each group). (H) Slc7a11 mRNA expression level in primary mouse hepatocytes (n = 6 for each group). (I–K) GPX4 expression in primary mouse hepatocytes (I: qRT-PCR, n = 6 for each group; J: immunofluorescence, scale bars 50 µm, n = 6 for each group; K: Western blot, n = 4 for each group). (A–C) p < 0.05: *vs. CO NS group; &vs. KO NS group; #vs. CO TAA group. (D–K) p < 0.05: *vs. DMSO group; #p < 0.05 vs. erastin + DMSO group. GSH, glutathione; MDA, malondialdehyde; GPX4, glutathione peroxidase 4; CO, control; NS, normal saline; KO, knockout; TAA, thioacetamide; DMSO, dimethyl sulfoxide; +: with the reagent; -: without the reagent.
Nrf2 signaling pathway of primary mouse hepatocytes
(A, B) Nrf2 and GPX4 expressions were measured in primary mouse hepatocytes by Western blot (n = 3 for each group). p < 0.05: *vs. DMSO group; #p < 0.05 vs. erastin + DMSO group. Nrf2, nuclear factor erythroid 2-related factor 2; GPX4, glutathione peroxidase 4; DMSO, dimethyl sulfoxide; +: with the reagent; -: without the reagent.
Inhibition of Cyclooxygenase-2 Upregulates the Nuclear Factor Erythroid 2-related Factor 2 Signaling Pathway to Mitigate Hepatocyte Ferroptosis in Chronic Liver Injury

March 2025

·

5 Reads

Background and Aims Ferroptosis plays an essential role in chronic liver diseases, and cyclooxygenase-2 (COX-2) affects liver fibrosis through multiple mechanisms. However, research on COX-2 regulation of ferroptosis in chronic liver injury remains limited. This study aimed to investigate whether and how COX-2 regulates ferroptosis in chronic liver injury. Methods In vivo, a thioacetamide (TAA)-induced chronic liver injury model, characterized by significant liver lipid peroxidation and oxidative stress, was used. COX-2+/+ and COX-2–/– mice were treated with TAA or normal saline. In vitro, primary mouse hepatocytes were isolated and treated with dimethyl sulfoxide (DMSO), erastin+DMSO, etoricoxib+erastin+DMSO, and tBHQ+erastin+DMSO. Mitochondrial morphology, iron metabolism, lipid peroxidation, and oxidative stress were assessed to verify ferroptosis. The nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway was measured to investigate the relationship between COX-2 and ferroptosis. Results TAA-treated COX-2–/– mice presented milder liver fibrosis, whereas TAA-treated COX-2–/– mice livers and etoricoxib+erastin+DMSO-treated primary hepatocytes exhibited alleviated mitochondrial damage compared with TAA-treated COX-2+/+ littermates and erastin+DMSO-treated primary hepatocytes, respectively. The knockout of COX-2 decreased ferrous ion concentration (p < 0.01) and mitigated lipid peroxidation in TAA-treated livers (p < 0.05). Furthermore, both COX-2 knockout and etoricoxib restored reduced glutathione (p < 0.05) and glutathione peroxidase 4 (p < 0.05), while decreasing malondialdehyde levels (p < 0.05). Additionally, COX-2 inhibition upregulated Nrf2, which helped alleviate erastin+DMSO-induced ferroptosis (p < 0.01). Conclusions Ferroptosis contributes to the progression of chronic liver injury. Inhibition of COX-2 upregulates Nrf2, mitigating hepatocyte ferroptosis in chronic liver injury.


Epidemiology of liver diseases: global disease burden and forecasted research trends

October 2024

·

52 Reads

·

12 Citations

Science China. Life sciences

We assessed the global incidence, mortality, and disability-adjusted life years (DALYs) associated with various liver diseases, including alcohol-related liver disease (ALD), hepatitis B/C virus infections (HBV or HCV), liver cancer, metabolic dysfunction-associated steatotic liver disease (MASLD), and other chronic liver diseases, from the 2019 Global Burden of Disease study. Additionally, we analyzed the global trends in hepatology research and drug development. From 2000 to 2019, prevalence rates increased for ALD, MASLD and other liver diseases, while they decreased for HBV, HCV, and liver cancer. Countries with a high socio-demographic index (SDI) exhibited the lowest mortality rates and DALYs. The burden of liver diseases varied due to factors like sex and region. In nine representative countries, MASLD, along with hepatobiliary cancer, showed highest increase in funding in hepatology research. Globally, the major research categories in hepatology papers from 2000 to 2019 were cancer, pathobiology, and MASLD. The United States (U.S.) was at the forefront of hepatology research, with China gradually increasing its influence over time. Hepatologists worldwide are increasingly focusing on studying the communication between the liver and other organs, while underestimating the research on ALD. Cancer, HCV, and MASLD were the primary diseases targeted for therapeutic development in clinical trials. However, the proportion of new drugs approved for the treatment of liver diseases was relatively low among all newly approved drugs in the U.S., China, Japan, and the European Union. Notably, there were no approved drug for the treatment of ALD in the world.




Recent developments in the management of ascites in cirrhosis

February 2024

·

114 Reads

·

4 Citations

In recent years, advances have been made for treating ascites in patients with cirrhosis. Recent studies have indicated that several treatments that have been used for a long time in the management of portal hypertension may have beneficial effects that were not previously identified. Long‐term albumin infusion may improve survival in patients with cirrhosis and ascites while beta‐blockers may reduce ascites occurrence. Transjugular intrahepatic porto‐systemic shunt (TIPS) placement may also improve survival in selected patients in addition to the control with ascites. Low‐flow ascites pump insertion can be another option for some patients with intractable ascites. In this review, we summarize the latest data related to the management of ascites occurring in cirrhosis. There are still unanswered questions, such as the optimal use of albumin as a long‐term therapy, the place of beta‐blockers, and the best timing for TIPS placement to improve the natural history of ascites, as well as the optimal stent diameter to reduce the risk of shunt‐related side‐effects. These issued should be addressed in future studies.




Citations (18)


... Liver infiltrating macrophages (also termed monocyte-derived macrophages) and resident macrophages (also termed Kupffer cells) acquire diverse phenotypes according to microenvironmental cues during liver diseases (143,144). Hence, liver macrophages are broadly involved in regulating hepatic inflammation, lipid metabolism, injury-repair processes (e.g., hepatocyte regeneration and ductular reaction), fibrogenesis and carcinogenesis, thereby taking active roles in MASLD progression (143,(145)(146)(147). All of these bring a crucial value in targeting liver (macrophage) inflammation to protect against MASLD. ...

Reference:

Bariatric surgery for metabolic dysfunction-associated steatotic liver disease (MASLD): Current knowledge of mechanisms
Reactive cholangiocyte-derived ORM2 drives a pathogenic modulation of the injured biliary niche through macrophage reprogramming

Gut

... Upon injury, hepatic stellate cells become activated and differentiate into highly contractile myofibroblasts implicated in acute injury repair and can contribute to liver scarring and fibrosis [29,31,33,34]. Recently, hepatic stellate cells were shown to be essential drivers of liver zonation through the secretion of modulators of the Wnt signaling pathway [35]. ...

Hepatic stellate cells control liver zonation, size and functions via R-spondin 3

Nature

... This paradox-declining rates but rising deaths-mirrors patterns also seen in countries with aging populations [25,26], where decades-long HCV progression to cirrhosis and HCC manifests as delayed mortality [26]. For instance, Japan observed similar mortality trends despite universal DAAs coverage [27], driven by late-stage diagnoses in older adults. China's success in reducing HBV incidence through vaccination contrasts with its slower progress against HCV, likely due to the absence of an HCV vaccine and historically fragmented screening programs-particularly in rural areas [10,28,29]. ...

Epidemiology of liver diseases: global disease burden and forecasted research trends
  • Citing Article
  • October 2024

Science China. Life sciences

... The liver contains diverse cell types, including hepatocytes, cholangiocytes, liver sinusoidal endothelial cells (LSECs), hepatic stellate cells (HSCs), Kupffer cells (KCs) and other immune cells. These cells interact to maintain hepatic sinusoidal homeostasis under healthy conditions, which can be disrupted in liver cirrhosis [48,49]. A recent study showed that mice lacking hepatocyte Bmal1 and Hif1α develop HPS [50], highlighting the crucial liver-lung communication. ...

Angiocrine signaling in sinusoidal homeostasis and liver diseases
  • Citing Article
  • May 2024

Journal of Hepatology

... Although many obese individuals with MASLD remain stable or have only mild liver disease, a proportion may progress to MASH, cirrhosis, end-stage liver failure, and MASH-HCC [138,139]. Therefore, early identification and monitoring of MASH patients at high risk of cirrhosis and HCC are of paramount importance. ...

Recent developments in the management of ascites in cirrhosis

... Ella Cre mice were crossed with COX-2 flox/flox mice to generate conventional COX-2 knockout mice (COX-2 -/-) and littermate controls (COX-2 +/+ ). 15 Age-matched male mice (six to eight weeks old, weighting 20-25 g) were selected for further experiments. Thioacetamide (TAA), which induces lipid peroxidation and oxidative stress-two components of ferroptosiswas employed to establish a chronic liver injury model in this study. ...

Atypical cholangiocytes derived from hepatocyte-cholangiocyte transdifferentiation mediated by COX-2: a kind of misguided liver regeneration

Inflammation and Regeneration

... Similarly, there are reports indicating that abnormal activation of the STING-IRF3 pathway facilitates hepatocyte apoptosis and disrupts glucose and lipid metabolism, resulting in the development of NAFLD (66). Interestingly, cGAS-STING signaling was shown to enhance hepatocyte pyroptosis and hepatic inflammation in liver fibrosis through activation of NLRP3 inflammasome (67). These evidences suggest that inhibiting the hyperactivation of the cGAS-STING pathway is a potential therapeutic strategy to inhibit hepatocyte death and liver injury. ...

STING mediates hepatocyte pyroptosis in liver fibrosis by Epigenetically activating the NLRP3 inflammasome
  • Citing Article
  • March 2023

Redox Biology

... These findings suggest that natural language processing approaches may be particularly valuable for complex patient populations with multifaceted risk factors. [61] Temporal validation procedures confirmed the robustness and generalizability of natural language processing-enhanced models across different time periods. Models trained on earlier time periods maintained strong performance when applied to later patient cohorts, with only minimal degradation in prediction accuracy. ...

“Open a Can of Worms”: Eosinophilic Liver Infiltration Due to Clonorchis sinensis Infection
  • Citing Article
  • February 2023

The American Journal of Gastroenterology

... In endotoxin-induced systemic inflammation in rats, the liver and lung produce more TNF than the spleen, with KCs in the liver being the primary sources of circulating TNF [68]. During cholestasis and liver diseases, macrophages in the liver produce pro-inflammatory mediators, including TNF [69,70], which may explain one of the mechanisms underlying HPS development. Monocytes are recruited to the hepatic sinusoid and pulmonary intravasculature following CBDL, accompanied by a systemic elevation in monocyte numbers [29]. ...

Role of Immune Cells in Biliary Repair