A hepatocyte-specific transcriptional program driven by Rela and Stat3 exacerbates experimental colitis in mice by modulating bile synthesis

  1. Jyotsna
  2. Binayak Sarkar
  3. Mohit Yadav
  4. Alvina Deka
  5. Manasvini Markandey
  6. Priyadarshini Sanyal
  7. Perumal Nagarajan
  8. Nilesh Gaikward
  9. Vineet Ahuja
  10. Debasisa Mohanty
  11. Soumen Basak
  12. Rajesh S Gokhale  Is a corresponding author
  1. Immunometabolism Laboratory, National Institute of Immunology, India
  2. System Immunology Laboratory, National Institute of Immunology, India
  3. Department of GastroEnterology, All India Institute of Medical Sciences, India
  4. Center for Cellular and Molecular Biology, India
  5. Gaikwad Steroidomics Lab LLC, United States
  6. Department of Biology, Indian Institute of Science Education and Research, India
15 figures, 2 tables and 1 additional file

Figures

Figure 1 with 1 supplement
Initiation of colitis in mice leads to hepatic Rela and Stat3 activation.

(A) GO pathway enrichment analysis was done for DEGs with adjusted p-value <0.05 on day 6 post DSS treatment. Bubble plot depicts the enrichment of pathways on day 6 for different genotypes, where the coordinate on the x-axis represents gene ratio, size of bubble represents the gene count and color represents the p-value. (B) Heatmap represents the normalized transcript count of the Rela and Stat3 pathway obtained from the RNA-seq experiment of three biological replicates. Scale is normalized across each row and color from blue to red represents minimum and the maximum values, respectively. (C) Representative confocal microscopy images show Rela and Stat3 activation in untreated and 6 days DSS-treated liver tissue of C57BL/6 mice. Images were taken at 40 X. Scale is 20 μm. (D) Western blot revealing the abundance of total Rela and total Stat3, and their phosphorylated functionally active forms, in the liver extracts prepared from wild-type C57BL/6 mice either left untreated or subjected to dextran sodium sulfate (DSS) treatment for 2, 4, and 6 days, respectively. (E) The signal intensity of bands corresponding to the indicated phosphorylated proteins was quantified from western blots, normalized against beta-actin, and presented as a bar plot. The data represent the means of three biological replicates ± SEM.

Figure 1—figure supplement 1
Biochemical, histological and molecular characterization of mice liver upon colitis induction.

(a) Violin plot comparing the clinical parameters in serum of untreated and dextran sodium sulfate (DSS)-treated wild-type mice (n=3) (b) Liver sections from untreated and DSS-treated wild-type mice were examined for histological features involving hematoxylin and eosin (H&E) staining [upper panel] and Sirius red staining [lower panel]. Data were obtained in 10 X magnification and represented three experimental replicates; two fields per section and a total of three sections from each set were examined. (c) Principal component analysis (PCA) plot illustrating the hepatic transcriptome, identified through global RNA-seq analyses, of untreated or DSS-treated-wild-type mice (n=3). (d) Schematic description of an experimental setup for gut sterilization by antibiotic treatment. (e) Western blot revealing the abundance of total Stat3 and it’s phosphorylated functionally active forms p-Stat3 (Ser727), in the liver extracts prepared from DSS-treated wild-type C57BL/6 mice with and without a prior antibiotic treatment for 4 weeks.

Figure 2 with 1 supplement
Rela and Stat3 deficiency in hepatocytes ameliorates dextran sodium sulfate (DSS)-induced acute colitis in mice.

(A) Line plot charting disease activity index of wild-type, RelaΔhep, Stat3Δhep, and RelaΔhepStat3Δhep littermate mice subjected to treatment with 2% DSS for 6 days. (B) Bar plot depicting colon length measured on day 6 post-onset of DSS treatment of mice of the indicated genotypes. Untreated wild-type littermates of corresponding genotypes were used as controls. (C) Colon sections from untreated and DSS-treated mice of the indicated genotypes were examined for histological features involving hematoxylin and eosin (H&E) staining [left panel] and alcian blue staining [right panel]. Data were obtained in 10 X magnification and represented three experimental replicates; two fields per section and a total of three sections from each set were examined. (D) Bar plot quantifying gut permeability of untreated and DSS-treated wild-type and RelaΔhepStat3Δhep mice. Briefly, the serum concentration of fluorescein isothiocyanate (FITC) was measured 6 hr after oral gavaging of the mice with FITC-dextran. (E) RT-qPCR reveals the relative abundance of the indicated mRNAs encoding broadly enterocyte-specific (above panel) or goblet cell-specific (below panel) markers in untreated or DSS-treated mice of the indicated genotypes.

Figure 2—figure supplement 1
Methods for characterization of mice strain and analysis of colon pathology of colitogenic mice.

Characterization of mice strain by (a) genotyping and (b) western blotting; WTE - whole tissue extract. (c) Representative images of the colon from untreated and dextran sodium sulfate (DSS)-treated wild-type and RelaΔhepStat3Δhep mice. (d) Dot plot representing the histological score for hematoxylin and eosin (H&E) stained colon sections in Figure 2C, each dot represents the score for each field analyzed.

Figure 3 with 1 supplement
Charting hepatic gene expressions in colitogenic wild-type and RelaΔhepStat3Δhep mice.

(A) Principal component analysis (PCA) plot illustrating the hepatic transcriptome, identified through global RNA-seq analyses, of untreated or dextran sodium sulfate (DSS)-treated wild-type and RelaΔhepStat3Δhep mice (n=3). DSS treatment was carried out for 6 days. (B) Bubble plot depicting the relative enrichment of GO biological terms for differentially expressed genes in wild-type or RelaΔhepStat3Δhep mice. The gene ratio for a given term and the adjusted p-value associated with the enrichment score has been presented for the individual genetic backgrounds. (C) Dot plot of dinor-cholic acid and dinor-chenodeoxycholic acid as detected in an untargeted LC-MS-based quantification of bile acids in the mucosal biopsy samples from inflammatory bowel disease (IBD) and Non-IBD patients. (D) Schematic presentation of classical and alternate pathways of bile synthesis in mice liver tissue. CA, CDCA, MCA, and UDCA represent cholic, chenodeoxycholic, muricholic, and ursodeoxycholic acids, respectively. (E) RT-qPCR analyses comparing the hepatic abundance of indicated mRNAs encoding enzymes involved in bile metabolism in DSS-treated wild-type and RelaΔhepStat3Δhep mice (n=3). Fold change is relative to their corresponding wild-type littermates.

Figure 3—source data 1

Table containing all the significantly regulated GO terms of which few are plotted in Figure 3B.

https://cdn.elifesciences.org/articles/93273/elife-93273-fig3-data1-v1.docx
Figure 3—source data 2

Data used for generating graph in Figure 3C.

https://cdn.elifesciences.org/articles/93273/elife-93273-fig3-data2-v1.docx
Figure 3—source data 3

Data used for generating graph in Figure 3E.

https://cdn.elifesciences.org/articles/93273/elife-93273-fig3-data3-v1.docx
Figure 3—source data 4

Table containing the demography of the control and UC patient for which the bile acids have been quantitated.

https://cdn.elifesciences.org/articles/93273/elife-93273-fig3-data4-v1.docx
Figure 3—figure supplement 1
Analysis of biochemical and molecular parameters of colitogenic wildtype and RelaΔhepStat3Δhep mice.

(a) Violin plot comparing the clinical parameters in serum of untreated and dextran sodium sulfate (DSS)-treated RelaΔhepStat3Δhep mice (n=3) (b) Liver sections from untreated and DSS-treated RelaΔhepStat3Δhep mice were examined for histological features involving hematoxylin and eosin (H&E) staining [upper panel] and Sirius red staining [lower panel]. Data were obtained in 10 X magnification and represent three experimental replicates; two fields per section and a total of three sections from each set were examined. Heatmap represents relative transcript abundance of (c) biosynthesis pathway (d) regulators of bile acid metabolic pathways and (e) modification enzymes of bile acid metabolic pathway of indicated genotypes obtained from the RNA-seq experiment of three biological replicates.

Figure 4 with 1 supplement
Altered accumulation of primary bile acids in RelaΔhepStat3Δhep mice accompanies a less severe inflammatory signature in the colitogenic gut.

Targeted LC-MS-based quantification of primary bile acid in the liver (A) and the colon (B) of dextran sodium sulfate (DSS)-treated wild-type and RelaΔhepStat3Δhep mice (n=5). (C) RT-qPCR analyses comparing the colonic abundance of indicated mRNAs encoding pro-inflammatory cytokines (n=3) for DSS-treated wild-type and RelaΔhepStat3Δhep mice. Fold change is relative to their corresponding wild-type littermates. (D) Dot-plot representing the frequency of F4/80+, CD11c+, and Ly6G+ cells among total DAPI-stained cells in the colon sections derived from DSS-treated wild-type and RelaΔhepStat3Δhep mice.

Figure 4—figure supplement 1
Microscopic analysis of immune cell infilteration in the colon of colitogenic mice.

(a) Representative images of colon tissue stained with indicated immune cell markers, DAPI, and merged section in the dextran sodium sulfate (DSS)-treated wild-type and RelaΔhepStat3Δhep mice. Scale represents 50 μm.

Figure 5 with 1 supplement
Supplementing chenodeoxycholic acid (CDCA) restores the colitogenic sensitivity in RelaΔhepStat3Δhep mice.

(A) Line plot charting the disease activity in a time course of wild-type and RelaΔhepStat3Δhep mice subjected to dextran sodium sulfate (DSS) treatment while being supplemented with 10 mg/kg CDCA daily. Mice devoid of CDCA supplementation were treated with DMSO as controls. (B) Bar plot comparing the colon length of RelaΔhepStat3Δhepmice subjected to DSS treatment for 6 days in the absence or presence of CDCA supplementation. (C) Hematoxylin and eosin (H&E) stained colon sections from DSS-treated RelaΔhepStat3Δhep mice with and without CDCA supplementation. Data were obtained in 10 X magnification, this is a representative of three experimental replicates and a total of four sections from each set were examined. RT-qPCR analyses comparing the colonic abundance of indicated mRNAs encoding (D) IEC-specific markers and (E) pro-inflammatory cytokines in mice subjected to DSS treatment for 6 days in the absence or presence of CDCA supplementation (n=4). Untreated RelaΔhepStat3Δhep mice were used as controls.

Figure 5—figure supplement 1
Supplementation of Chenodeoxycholic acid (CDCA) to the mice and its effect on gut.

(a) Schematic description of the experimental design describing the course of dextran sodium sulfate (DSS) treatment and chenodeoxycholic acid (CDCA) supplementation. (b) Quantification of CDCA in the colon of mice, that received intraperitoneal injection of either CDCA or DMSO (control), through targeted LC-MS experiments. (c) Line plot charting the disease activity in a time course of RelaΔhepStat3Δhep mice subjected to daily supplementation of 10 mg/kg CDCA. Mice devoid of CDCA supplementation were treated with DMSO as controls. (d) Bar plot comparing the colon length of RelaΔhepStat3Δhepmice subjected to CDCA supplementation. (e) Colon sections from RelaΔhepStat3Δhepmice supplemented with CDCA were examined by hematoxylin and eosin (H&E) staining.

A model depicting the immuno-metabolic network linking the inflammation-induced hepatic signaling pathway to intestinal pathologies in mice.
Author response image 1
Author response image 2
Author response image 3
Author response image 4
Author response image 5
Author response image 6
Author response image 7
Author response image 8
Author response image 9

Tables

Key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Strain, strain background (Mus musculus)Alb-Cre Rela-/- Stat3-/-A gift from Dr. Lee Quinton’s lab at Boston UniversityC57BL/6
Strain, strain background (Mus musculus)Relafl/fl Stat3fl/flA gift from Dr. Lee Quinton’s lab at Boston UniversityC57BL/6
Strain, strain background (Mus musculus)Alb-Cre Rela-/-A gift from Dr. Lee Quinton’s lab at Boston UniversityC57BL/6
Strain, strain background (Mus musculus)Relafl/flA gift from Dr. Lee Quinton’s lab at Boston UniversityC57BL/6
Strain, strain background (Mus musculus)Alb-Cre Stat3-/-A gift from Dr. Lee Quinton’s lab at Boston UniversityC57BL/6
Strain, strain background (Mus musculus)Stat3fl/flA gift from Dr. Lee Quinton’s lab at Boston UniversityC57BL/6
Antibodyanti-mouse Ly-6G-PE
(Rat monoclonal)
 BD BiosciencesCat# 551461,
RRID: AB_394208
IF (1:500)
Antibodyanti-mouse F4/80-FITC (Mouse Polyclonal)BioLegendCat# 157309, RRID: AB_2876535IF (1:500)
Antibodyanti-mouse CD11c-PE (Hamster monoclonal)BioLegendCat# 117308, RRID: AB_313777IF (1:500)
Antibodyanti-mouse CD4-PE (Rat monoclonal)BioLegendCat# 100408, RRID: AB_312693IF (1:500)
Antibodyanti-mouse STAT3 (mouse monoclonal)ThermoFisher ScientificCat# MA1-13042, RRID: AB_10985240IF (1:100)
WB (1:1000)
Antibodyanti-mouse Rela (Rabbit polyclonal)Santa Cruz BiotechnologyCat# sc372, RRID: AB_632037IF (1:100)
WB (1:1000)
Antibodyanti-mouse Phospho-Stat3 (Ser727) (Rabbit monoclonal)Cell Signaling TechnologyCat# 34911, RRID: AB_2737598IF (1:100)
WB (1:1000)
Antibodyanti-mouse Phospho-Stat3 (Ser727) (Rabbit monoclonal)Cell Signaling TechnologyCat# 34911, RRID: AB_2737598IF (1:100)
WB (1:1000)
Antibodyanti-mouse Phospho-Stat3 (Tyr705) (Rabbit monoclonal)Cell Signaling TechnologyCat# 34911, RRID: AB_2737598IF (1:100)tjp
WB (1:1000)
Antibodyanti-mouse Phospho-NF-κB p65 (Ser536) (Rabbit polyclonal)Cell Signaling TechnologyCat# 3031,
RRID: AB_330559
WB (1:1000)
Antibodyanti-mouse GAPDH (Rabbit monoclonal)Cell Signaling TechnologyCat# 2118,
RRID: AB_561053
WB (1:1000)
Antibodyanti-mouse β-Actin (Rabbit polyclonal)Cell Signaling TechnologyCat# 4967,
AB_330288
WB (1:1000)
Antibodyanti-rabbit IgG (H+L) Secondary Antibody Alexa Fluor Plus 555 (Goat polyclonal)Thermo Fisher ScientificCat# A32732, AB_2633281IF (1:2000)
Sequence-based reagentTjp1_F This paperPCR primerGCTTTAGCGAACAGAAGGAGC
Sequence-based reagentTjp1_R This paperPCR primerTTCATTTTTCCGAGACTTCACCA
Sequence-based reagentOcln_F This paperPCR primerTGAAAGTCCACCTCCTTACAGA
Sequence-based reagentOcln_R This paperPCR primerCCGGATAAAAAGAGTACGCTGG
Sequence-based reagentMuc2_F This paperPCR primerAGGGCTCGGAACTCCAGAAA
Sequence-based reagentMuc2_R This paperPCR primerCCAGGGAATCGGTAGACATCG
Sequence-based reagentTff3_F This paperPCR primerTTGCTGGGTCCTCTGGGATAG
Sequence-based reagentTff3_R This paperPCR primerTACACTGCTCCGATGTGACAG
Sequence-based reagentIl1b_F This paperPCR primerCATCCCATGAGTCACAGAGGATG
Sequence-based reagentIl1b_R This paperPCR primerACCTTCCAGGATGAGGACATGAG
Sequence-based reagentTnf_F This paperPCR primerCTGAACTTCGGGGTGATCGG
Sequence-based reagentTnf_R This paperPCR primerGGCTTGTCACTCGAATTTTGAGA
Sequence-based reagentIl6_F This paperPCR primerCCCCAATTTCCAATGCTCTCC
Sequence-based reagentIl6_R This paperPCR primerGGATGGTGTTGGTCCTTAGCC
Sequence-based reagentGapdh_F This paperPCR primerAGGTCGGTGTGAACGGATT
Sequence-based reagentGapdh_R This paperPCR primerAATCTCCACTTTGCCACTGC
Sequence-based reagentCyp7a1_F This paperPCR primerGCTGTGGTAGTGAGCTGTTG
Sequence-based reagentCyp7a1_R This paperPCR primerGTTGTCCAAAGGAGGTTCACC
Sequence-based reagentCyp8b1_F This paperPCR primerCCTCTGGACAAGGGTTTTGTG
Sequence-based reagentCyp8b1_R This paperPCR primerGCACCGTGAAGACATCCCC
Sequence-based reagentCyp27a1_F This paperPCR primerAGGGCAAGTACCCAATAAGAGA
Sequence-based reagentCyp27a1_R This paperPCR primerTCGTTTAAGGCATCCGTGTAGA
Sequence-based reagentCyp7b1_F This paperPCR primerTCCTGGCTGAACTCTTCTGC
Sequence-based reagentCyp7b1_R This paperPCR primerCCAGACCATATTGGCCCGTA
Sequence-based reagentCre_F This paperPCR primerGGTGAACGTGCAAAACAGGCTC
Sequence-based reagentCre_R This paperPCR primerAAAACAGGTAGTTATTCGGATCATCAGC
Sequence-based reagentTcrd_F This paperPCR primerCAAATGTTGCTTGTCTGGTG
Sequence-based reagentTcrd_R This paperPCR primerGTCAGTCGAGTGCACAGTTT
Sequence-based reagentStat3flox_F This paperPCR primerCCTGAAGACCAAGTTCATCTGTGTTGAC
Sequence-based reagentStat3flox_R This paperPCR primerCACACAAGCCATCAAACTCTGGTCTCC
Sequence-based reagentRelaflox_F This paperPCR primerGAGCGCATGCCTAGCACCAG
Sequence-based reagentRelaflox_R This paperPCR primerGTGCACTGCATGCGTGCAG
Chemical compound, drugDextran sulphate sodium saltSigma-AldrichCat# 42867
Chemical compound, drugFluorescein isothiocyanate-dextranSigma-AldrichCat# 60842-46-8
Chemical compound, drugChenodeoxycholic acidSigma-AldrichCat# C9377
Chemical compound, drugDAPISigma-AldrichCat# D9542
Chemical compound, drugPowerUp SYBRThermo Fisher ScientificCat# A25742
Chemical compound, drugFluorosheildSigma-AldrichCat# F6182
Commercial assay or kitNucleoSpin RNAMacherey-NagelCat# 74106
Commercial assay or kitPrimescript 1st strand cDNA synthesis kitTakara BioCat# 6110 A
Software, algorithmPrism 9GraphPad9.0
Author response table 1
The total
number of
nuclei
analyzed
90th percentile of
signal intensity for
control samples
Nuclei of DSS-treated
sections with signal
intensity above the 90th
percentile
Percentage
overlap
For Rela
probed
samples
293.46527100
For Stat3
probed
samples
292.9352993

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Jyotsna
  2. Binayak Sarkar
  3. Mohit Yadav
  4. Alvina Deka
  5. Manasvini Markandey
  6. Priyadarshini Sanyal
  7. Perumal Nagarajan
  8. Nilesh Gaikward
  9. Vineet Ahuja
  10. Debasisa Mohanty
  11. Soumen Basak
  12. Rajesh S Gokhale
(2024)
A hepatocyte-specific transcriptional program driven by Rela and Stat3 exacerbates experimental colitis in mice by modulating bile synthesis
eLife 12:RP93273.
https://doi.org/10.7554/eLife.93273.3