1. Introduction
Farnesoid X receptor (FXR) is a regulator of bile acid (BA) and lipid, that meticulously regulates bile acid production and circulation. The role of FXR activation has been shown to be important in colitis [
1] and nonalcoholic steatohepatitis (NASH) [
2] in mouse models. FXR knockout (KO) mice exhibit increased BA pool concentration, which leads to spontaneous tumor development in liver, intestinal epithelial cell proliferation, and increased colon cancer susceptibility [
3,
4,
5]. Bile acids and FXR play an important role in the modulation of a range of inflammatory responses, barrier function and the prevention of bacterial translocation in the intestinal tract. Although it has been determined that FXR plays a variety of functions, further research is still needed to determine how it assists in intestinal epithelial barrier function and colon tumorigenesis [
3,
4,
6].
The three subfamilies of Klotho are αKlotho, βKlotho, and γKlotho [
7]. βKlotho is a co-receptor for fibroblast growth factor 19 (FGF19) and FGF21, two hormones that play important roles in regulating energy and bile acid metabolism. In response to bile acid binding, FXR induces the expression of FGF19 in the ileum, which then binds to beta-klotho and activates downstream signaling pathways, leading to inhibition of bile acid synthesis in the liver. βklotho protein levels were substantially decreased in FXR KO mice; on the other hand, overexpression of βklotho in FXR-lacking hepatocytes partially restored FGF19 signaling and inhibition by FGF19 of Cyp7a1, which encodes the rate-limiting BA biosynthetic enzyme [
8]. Overall, the interaction between FXR, βklotho, and FGF19/21 signaling plays an important role in regulating bile acid synthesis and metabolism. In the gut, the decreased βklotho expression caused by gene variation is associated with increased intestinal permeability in patients with irritable bowel syndrome with diarrhea [
9] Furthermore, βklotho activation enhanced tight junction (TJ) proteins, thereby protecting against alcohol-induced TJ proteins endocytosis and degradation as well as intestinal barrier impairment [
10].
The gene αKlotho has been associated with anti-aging effects and regulates several pathways involved in aging, such as phosphate homeostasis, insulin signaling, and Wnt/β-catenin signaling [
11,
12]. It also affects intracellular signaling pathways, including p53/p21, cyclic adenosine monophosphate (cAMP), protein kinase C (PKC) and transforming growth factor β (TGFβ) [
13,
14]. Studies have shown that αKlotho is expressed in the intestinal epithelium, which is the layer of cells that forms the intestinal barrier. αKlotho has been shown to promote the formation of tight junctions, which are the specialized structures that hold intestinal epithelial cells together. αKlotho levels are epigenetically downregulated in cancer, and overexpression or treatment with soluble klotho or the αKlotho domain slows growth of cancer cells in vitro and in vivo. αKlotho is also downregulated in colorectal cancer and αklotho overexpression inhibits growth of colorectal cancer cells [
15,
16]. αKlotho has been identified as a tumor suppressor and inhibits the insulin-like growth factor 1 (IGF1), FGF, and Wnt/β-catenin pathways; yet, the mode of action of αKlotho in cancer is a matter of confirm. To determine how much this interaction between FXR and Klotho affects the regulation of bile acid metabolism and other biological processes, more research into the molecular mechanisms underpinning it is necessary.
There is a strong interplay between intestinal flora and bile acid (BA) metabolism, which plays a crucial role in digestion and shaping the gut microbial community. Recent research has shown that gut microbiota-mediated deconjugation of bile acids influences de novo bile acid synthesis in the liver in an FXR-FGF15/19 axis-dependent manner in mice [
17]. Gut microbiome performs essential bile acids and also influences host metabolism via the modulation of metabolites [
18]. Gut microbiota also influences host metabolism via the modulation of metabolites, including the endotoxin LPS, bile acids, and short chain fatty acids. Therefore, they partially mediate the interaction between the gastrointestinal system and other organs [
19]. Prevalent genera identified in the guts include both beneficial and harmful microorganisms, such as
Bacteroides,
Eubacterium,
Bifidobacterium,
Ruminococcus,
Clostridium,
Lactobacillus,
Escherichia,
Streptococcus [
20], and naturally acquired enterohepatic
Helicobacter spp. infection [
21]. While most infected mice develop minimal pathologic changes, susceptible strains exhibit typhlocolitis and hepatitis, which can further progress to colon cancer and hepatocellular carcinoma [
22]. Intestinal bacterial dysbiosis and
Helicobacter hepaticus infection act synergistically during inflammation and neoplastic progression [
23].
In this study, we demonstrated that downregulating FXR levels by applying LPS-treated and FXR-KO resulted in a decline in levels of α/βKlotho, FGF19, FGF21, and FGF23. GW4064 was found to improve the αKloth/βKlotho/FGFs pathway and decrease intestinal inflammation and β-catenin formation induced by LPS-induced intestinal barrier failure and colon carcinogenesis. The study also revealed that GW4064 prevented LPS-induced intestinal tight-junction damage, regulated bile acids, and improved microbiota dysbiosis in an FXR-dependent mechanism. Furthermore, LPS treatment induced severe intestinal barrier damage, higher bile acid levels, microbiota dysbiosis, and inflammatory response in FXR-deficient mice. The findings suggest that activating FXR and the αKloth/βKlotho/FGFs pathway might be a new strategy for preventing intestinal epithelial barrier dysfunction and colon tumorigenesis.
3. Discussion
FXR activation can decrease inflammation in the gut and improve the integrity of the intestinal epithelial barrier. Similar to our research, Watanabe M et al. investigated at which FXR activation affected affects the function of the intestinal barrier in a mouse model of intestinal injury [
24]. Our preliminary research indicates that GW4064 treatment prevented the expression of ZO-1 and claudin-1 from declining only in WT mice, indicating that FXR activation may be important for maintaining TJPs expression in the gut [
25].
FXR is a transcription factor that regulates bile acid (BA) homeostasis in the liver and gastrointestinal system. In FXR KO mice, BA production is impaired, resulting in increased BA concentrations [
26]. Abnormalities in BA metabolism (such as TCA) are linked to hepatocellular carcinoma [
3] and colon cancer [
27]. Moreover, bile acid is absorbed via the ASBT and it is exported via the basolateral heteromeric OSTα-OSTβ in intestinal epithelial barrier [
28]. In the present study, OSTβ protein levels were significantly lower in both LPS-treated mice. Specifically, the OSTβ protein levels were significantly lower in FXR KO mice than in WT mice. OST levels were severely diminished in intestinal injury tissue and markedly reduced in adjacent non-tumorous tissue [
29]. In LPS-induced intestinal injury, FXR KO mice had higher serum levels of taurine-conjugated bile acids, which are linked to sepsis. The FXR-specific agonist GW4064 improved Tauro-BA profile in plasma and liver and enhanced ASBT and OSTα/β levels in WT mice, but not in FXR KO mice, suggesting that the effect of GW4064 was dependent on FXR. In LPS-induced intestinal injury, FXR KO mice showed increased expression of
Asbt,
Ibabp, and
Mdr2 and decreased protein levels of OSTβ, leading to intracellular accumulation of BAs, increased cell apoptosis, and inflammatory response in enterocytes of the colon.
The βKlotho/FGF15/19/21 pathway activation via FXR results in the reduction of bile acid production through negative feedback control. The βKlotho/FGF19 signaling was hampered in FXR-KO mice hepatocytes, which is similar to our finding [
8]. βKlotho KO mice presented hepatic alterations, combining a proinflammatory status and initiation of fibrosis. These defects are associated with a massive shift in BA composition in the enterohepatic system and the circulation, which is characterized by a large excess of microbiota-derived deoxycholic acid (DCA) known for its genotoxicity in the gastrointestinal tract [
30]. Interestingly, despite increased synthesis and excretion of bile acids by the liver, ileal expression of ASBT was not reduced in β-Klotho-deficient mice [
15,
16]. FXR and βKlotho/FGF19/FGF15 plays an important role in modulation of the liver receptor homolog -1 (LRH-1) or pregnane X receptor (RAR)/retinoic acid receptor (RAR) pathway to regulate ASBT causing in alter bile acid translation. Our findings demonstrated that loss of FXR or treatment with GW4064, which enhances FXR activity, interferes with the response of β-Klotho to ASBT regulation. This reveals the mechanisms by which FXR influences β-Klotho expression and activity. Furthermore, βKlotho appears to regulate the endocytosis and degradation of Occludin and ZO-1. The endocytosis of TJ proteins from the plasma membrane is a key that mechanism regulates TJ plasticity and function in epithelial barrier tissues [
31]. In the present study, we have shown that FXR activation by GW4064 can regulate BA homeostasis and intestinal barrier function involved in the βKlotho/FGF19/FGF21 pathway.
Mechanistically, we showed that BAs that antagonize intestinal Farnesoid X receptor (FXR) function, including tauro-β-muricholic acid (T-βMCA) and deoxycholic acid (DCA), induce proliferation and DNA damage in Lgr5+ cells. Conversely, selective activation of intestinal FXR can restrict abnormal Lgr5
+ cell growth and curtail CRC progression. FXR silencing in chronic colitis mouse models of intestinal tumorigenesis results in early mortality and increased tumor progression [
32]. In colon cancer, low FXR expression was correlated with worse clinical outcome [
32]. The present study revealed that knockdown of FXR activated Wnt/β-catenin signaling [
5]. The mechanism by which FXR suppresses tumor growth remains unclear, but it may involve protecting the colonic epithelium from inflammation and ameliorating BA toxicity by upregulating intracellular BA binding proteins and efflux transporters and downregulating influx transporters and de novo BA synthesis [
33,
34]. Thus, the re-establishment of FXR signaling not only restricts aberrant Lgr5+ stem cell proliferation but also promotes gut health including restoring the intestinal barrier [
35,
36] and BA homeostasis [
37,
38]. Beyond its well-established role in regulating cytotoxicity of hydrophobic BAs, early study highlights the role of FXR in restricting the tumorigenesis of Lgr5+ cells, which mediate the key adenoma-to-adenocarcinoma transformation. FXR deficiency increases colon cancer susceptibility by increasing epithelial permeability to bacteria, promoting Wnt/β-catenin signaling and increasing intestinal inflammation [
29,
36]. In this study, we shown that GW4064 significantly reduced colon tumorigenesis markers, LGR5, CD44, CD34, Cyclin D1 in LPS-treat WT mice, but not in FXR KO mice.
The αklotho is a classical aging suppressor, which is a subfamily of βKlotho. Klotho has been shown to regulate intestinal barrier function and protect against gut inflammation, which may help to prevent the development of colon cancer. This decrease in αKlotho levels may be observed in patients with several aging-related diseases such as cancer [
39]. In colorectal cancer models, Klotho overexpression has been shown to reduce overall β-catenin expression, inhibiting transcriptional pathway activity by binding to the Wnt3a ligand, and thereby decreasing nuclear translocation of β-catenin [
40]. The anti-neoplastic effects of Klotho and describe the modulation of downstream oncogenic signaling pathways, including Wnt/β-catenin, FGF, IGF1, PIK3K/AKT, TGFβ, and the Unfolded Protein Response [
39]. In this study, we shown that GW4064 significantly reduced colon tumorigenesis may through FXR dependent in αKlotho/FGF23 pathway.
In the present study,
Bacteroidetes, and
Bacteroides genera were the predominant gut anaerobes, which increased as a result of FXR deactivation in both WT mice and FXR KO mice treated with LPS; these data are consistent with those of Jena et al. [
41]
Bacteroides species are bile acid-resistant pathogens, which are found in most anaerobic infections with a mortality of >19% [
42]. In this study, we focused on the TCA, whose components, taurine and cholic acid, support the growth of microbial groups of low abundance. These less less-abundant microbes are mechanistically implicated in DNA-damage and tumor-promotion mediated by their metabolic by-products [
27]. Our data revealed that
Bacteroides thetaiotaomicron, and
Bacteroides acidifaciens were eliminated by GW4064 in LPS-treated WT mice, but not in FXR KO mice. However, the persistent
Bacteroides thetaiotaomicron in FXR KO mice might lead to maintaining chronic intestinal inflammation after GW4064 treatment. Furthermore, the commensal gut bacterium
Bacteroides thetaiotaomicron has a robust ability to degrade dietary polysaccharides and host mucin glycans. This, in turn, leads to a notable reduction in the thickness of the colonic mucus layer and exacerbates enteric infections by altering the metabolic environment [
43]. In the present study, GW4064 reversed LPS-induced relative abundance of
Bacteroides acidifaciens in WT mice. Compared to WT mice,
Bacteroides thetaiotaomicron was increased in FXR KO mice, and neither LPS nor GW4064 treatment showed any significant change in it. While FXR deficiency strongly affects the expression of genes related to immunity and bile acid metabolism, as well as the composition of the microbiome; its deficiency was not able to produce significant histopathological changes in the absence of
Helicobacter hepaticus infection. This data confirms those reported by Swennes et al. [
23]
Bacteroides acidifaciens, a colitis-associated species and an abundant member of the
Bacteroidaceae family [
44], was also elevated during inflammation, but decreased rapidly after colitis. Specifically, LPS decreased FXR levels and induced microbiota dysbiosis in the colon, which elevated TCA, TCDCA, T-ursodeoxycholic acid (TUDCA), and TDCA in the plasma and liver, leading to the impairment of intestinal tight junctions and inflammation. Conversely, direct activation of FXR restores bile acid levels, microbiota profiles, intestinal tight junctions, and reduces inflammation in LPS-treated WT.
The contribution of SCFAs to maintenance of gut homeostasis has been investigated extensively, there is an increasing body of evidence that commensal bacteria and bacterial metabolites have opposing roles in inflammatory responses and carcinogenesis depending on the cell type and the environment. Based on our findings from the gut microbiota and SCFAs results, we can highlight three important aspects: (1) The genus
Clostridium has been associated with the production of several SCFAs, including butyrate acid, valerate acid, isobutyrate acid, and 4-methylvalerate acid. The trends in
Clostridium populations appear to correspond with the levels of these SCFAs [
45]. (2) The genus
Bacteroides has been linked with the production of propionate, another type of SCFA that has important roles in gut health and metabolism. The trends in
Bacteroides populations appear to correspond with the levels of propionate.
Bacteroides and
Clostridium may be responsible for the increase in SCFA levels and bile acid metabolism in FXR KO treatments. (3) Isovalerate (or isovaleric acid) is a branched-chain fatty acid produced by the gut microbiota. The trends in isovalerate levels appear to correspond with the populations of
Clostridium saccharogumia. However, research on the relationship between
Clostridium saccharogumia and SCFAs is currently lacking. In a steady-state situation, butyrate is present in the mM range in the gut lumen and serves as the primary energy source for colonocytes [
46]. However, in the context of cancerous colonocytes, butyrate was shown to act paradoxically. In addition to that, butyrate is capable of promoting carcinogenesis in a genetic mouse model based on mutations in the Apc and the mismatch repair gene Msh2 (Apc
Min/+; Msh2
–/–) [
47]. And superior inhibitory efficacy of butyrate over propionate and acetate against human colon cancer cell proliferation via cell cycle arrest and apoptosis [
48]. Our study shows that GW4064 positively affects SCFA and BCFA levels in normal mice. However, in FXR KO mice, the regulatory effect of GW4064 on SCFA is different from that in normal mice, possibly due to an increase in total bile acids and pathogenic intestinal tract bacteria, including
Clostridium and
Bacteroidetes spp.
Ultimately, the study search elucidates that the activation of the FXR/αKlotho/βKlotho/FGFs pathway may hold promise as a potential strategy for averting intestinal barrier dysfunction and colon tumorigenesis. Lastly, our research highlights those elevated levels of bile acids and the high abundance of certain microbiota, such as Clostridium, Bacteroidetes, and Helicobacter hepaticus, Bacteroides thetaiotaomicron, in FXR KO mice are crucial factors leading to the reduction of colonic tight junction proteins and consequent damage to the intestinal barrier. Additionally, LPS treatment exacerbates structural damage to the intestine, rendering the administration of GW4064 less effective in mitigating these effects. As such, this study emphasizes two key aspects: the paramount importance of FXR in the context of intestinal injury and the secondary significance of the gut microbiota-bile acid-FXR axis as a therapeutic target for intestinal diseases.
Figure 1.
GW4064-dependent FXR activation ameliorates histological characteristics of tight junctions. (A) Representative hematoxylin and eosin staining and immunohistochemistry of FXR, Claudin-1, and ZO-1 staining for colon sections are shown. Scale bar: 50 μm. The (B, C) FXR, (D, E) Claudin-1, and ZO-1 protein were determined by Western blot. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 1.
GW4064-dependent FXR activation ameliorates histological characteristics of tight junctions. (A) Representative hematoxylin and eosin staining and immunohistochemistry of FXR, Claudin-1, and ZO-1 staining for colon sections are shown. Scale bar: 50 μm. The (B, C) FXR, (D, E) Claudin-1, and ZO-1 protein were determined by Western blot. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 2.
GW4064 affects bile acids profile. (A) Schematic diagram of the experiment. The concentration of (B) plasma, (C) liver, (D) colon bile acids, and (E-H) individual bile acids in LPS-treated wild-type (WT) and FXR knockout (KO) mice with and without GW4064. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 2.
GW4064 affects bile acids profile. (A) Schematic diagram of the experiment. The concentration of (B) plasma, (C) liver, (D) colon bile acids, and (E-H) individual bile acids in LPS-treated wild-type (WT) and FXR knockout (KO) mice with and without GW4064. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 3.
The effects of GW4064 on bile acid receptors and transporters expression. Immunohistochemistry of colon sections for (A) MRP3, OATP1, (B) OSTβ, and ASBT staining. Scale bar: 100 μm. (C, D) The MRP2, MRP3, OSTβ, ASBT, and OATP1 proteins were determined by Western blot. qRT-PCR analyses of colonic mRNA expression of (E) Fxr, Pxr, Car, Cyp3a11, Sult2a1, and (F) Oatp2b1, Asbt, Ibabp, Ostα, Ostβ, and (G) Mrp2, Mrp3, Mdr1b, Mdr2, and Bcrp. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 3.
The effects of GW4064 on bile acid receptors and transporters expression. Immunohistochemistry of colon sections for (A) MRP3, OATP1, (B) OSTβ, and ASBT staining. Scale bar: 100 μm. (C, D) The MRP2, MRP3, OSTβ, ASBT, and OATP1 proteins were determined by Western blot. qRT-PCR analyses of colonic mRNA expression of (E) Fxr, Pxr, Car, Cyp3a11, Sult2a1, and (F) Oatp2b1, Asbt, Ibabp, Ostα, Ostβ, and (G) Mrp2, Mrp3, Mdr1b, Mdr2, and Bcrp. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 4.
GW4064 regulates αKlotho/βKlotho/FGF19/FGF21/FGF23 pathway. Immunohistochemistry of colon sections for (A) αKlotho, βKlotho, and (B) FGF19, FGF21, FGF23 staining. Scale bar: 50 μm. The (C) αKlotho, βKlotho, and (D) FGF19, FGF21, FGF23 protein levels were determined by Western blot. *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 4.
GW4064 regulates αKlotho/βKlotho/FGF19/FGF21/FGF23 pathway. Immunohistochemistry of colon sections for (A) αKlotho, βKlotho, and (B) FGF19, FGF21, FGF23 staining. Scale bar: 50 μm. The (C) αKlotho, βKlotho, and (D) FGF19, FGF21, FGF23 protein levels were determined by Western blot. *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 5.
FXR deletion exhibits signs of TLR4-mediated inflammatory response, apoptosis of IECs, and leads to ER stress. qRT-PCR analyses of colonic mRNA levels of (A) Tnfα, Ifnγ, Il-1β, pro-caspase-1, Asc, Nlrp3, Pannexin-1 and (B) Atf4, Atf6, Grp78, Chop and Xbp1s. (C) Immunohistochemistry of colon sections for TLR4 and Caspase 3 staining. Scale bar: 100 μm. (D, E) The TLR4, MyD88 and NF-κB protein were determined by Western blot. (F, G) The Caspase 3 protein levels were determined by Western blot. (H) Immunohistochemistry of colon sections for β-catenin and c-Myc staining. Scale bar: 100 μm. (I, J) The β-catenin, c-Myc, (K, L) TGFβRII, ICAM, VCAM, VEGF1, VEGFR1, and MMP9 protein levels were determined by Western blot. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 5.
FXR deletion exhibits signs of TLR4-mediated inflammatory response, apoptosis of IECs, and leads to ER stress. qRT-PCR analyses of colonic mRNA levels of (A) Tnfα, Ifnγ, Il-1β, pro-caspase-1, Asc, Nlrp3, Pannexin-1 and (B) Atf4, Atf6, Grp78, Chop and Xbp1s. (C) Immunohistochemistry of colon sections for TLR4 and Caspase 3 staining. Scale bar: 100 μm. (D, E) The TLR4, MyD88 and NF-κB protein were determined by Western blot. (F, G) The Caspase 3 protein levels were determined by Western blot. (H) Immunohistochemistry of colon sections for β-catenin and c-Myc staining. Scale bar: 100 μm. (I, J) The β-catenin, c-Myc, (K, L) TGFβRII, ICAM, VCAM, VEGF1, VEGFR1, and MMP9 protein levels were determined by Western blot. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 6.
GW4064 alters colonic stem cell proliferation. (A) Double immunofluorescence staining of LGR5 (red) and CD44 (green); (B) PCNA (red) and CD44 (green); BrdU (red) and (C) CD34 (green); (D) BrdU (red) and CD133 (green); (E) Cyclin D1 (red) and GCSF (green) with DAPI (blue) in colon. Scale bar = 50 μm. (F) qRT-PCR analyses of colonic mRNA level of Lgr5, Olfm4, and Cyclin D1. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 6.
GW4064 alters colonic stem cell proliferation. (A) Double immunofluorescence staining of LGR5 (red) and CD44 (green); (B) PCNA (red) and CD44 (green); BrdU (red) and (C) CD34 (green); (D) BrdU (red) and CD133 (green); (E) Cyclin D1 (red) and GCSF (green) with DAPI (blue) in colon. Scale bar = 50 μm. (F) qRT-PCR analyses of colonic mRNA level of Lgr5, Olfm4, and Cyclin D1. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 7.
GW4064 alters intestinal microbial composition in mice. Alpha diversity measurements of the microbiota across locations. (A) Chao1 (B) and Shannon’s diversity index. PCA plot based on the abundance of bacterial gene sequences in fecal content. (C) Axes correspond to principal component 1 (x axis) and 2 (y axis). (D) Microbial community bar plot by phylum relative abundance (%). (E-I) Phylum levels of Proteobacteria, Verrucomicrobia, Firmicutes, Bacteroidetes, and Firmicutes/Bacteroidetes (F/B) ratio. (J) Microbial community bar plot by class relative abundance (%). (K-N) Class levels of Betaproteobacteria, Gammaproteobacteria, Clostridia and Verrucomicrobiae. (O) Microbial community bar plot by order relative abundance (%). (P-R) Order levels of Enterobacteriales, Verrucomicrobiales and Burkholderiales. (S) Genus levels of Bacteroides, Clostridium, and Escherichia. (T) Microbial community bar plot by species relative abundance (%). (U-Z) Species levels of Bacteroides thetaiotaomicron, Escherichia coli, Bacteroides acidifaciens, Akkermansia muciniphila, Clostridium saccharogumia, and Helicobacter hepaticus. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Figure 7.
GW4064 alters intestinal microbial composition in mice. Alpha diversity measurements of the microbiota across locations. (A) Chao1 (B) and Shannon’s diversity index. PCA plot based on the abundance of bacterial gene sequences in fecal content. (C) Axes correspond to principal component 1 (x axis) and 2 (y axis). (D) Microbial community bar plot by phylum relative abundance (%). (E-I) Phylum levels of Proteobacteria, Verrucomicrobia, Firmicutes, Bacteroidetes, and Firmicutes/Bacteroidetes (F/B) ratio. (J) Microbial community bar plot by class relative abundance (%). (K-N) Class levels of Betaproteobacteria, Gammaproteobacteria, Clostridia and Verrucomicrobiae. (O) Microbial community bar plot by order relative abundance (%). (P-R) Order levels of Enterobacteriales, Verrucomicrobiales and Burkholderiales. (S) Genus levels of Bacteroides, Clostridium, and Escherichia. (T) Microbial community bar plot by species relative abundance (%). (U-Z) Species levels of Bacteroides thetaiotaomicron, Escherichia coli, Bacteroides acidifaciens, Akkermansia muciniphila, Clostridium saccharogumia, and Helicobacter hepaticus. Values are expressed as the mean ± standard error of the mean (n = 5 mice per group). *p < 0.05, WT vs WT+ LPS; #p < 0.05, WT + LPS vs WT + LPS + GW4064; £p < 0.05, WT vs FXR KO; ¤p < 0.05, FXR KO vs FXR KO + LPS; ¥p < 0.05, FXR KO + LPS vs FXR KO + LPS + GW4064.
Table 1.
The antibodies used for IF, IHC, and Western blot.
Table 1.
The antibodies used for IF, IHC, and Western blot.
Antibodies |
Company/State/ Country |
Cat number |
ASBT/SLC10A2 |
Bioss Antibidies, Woburn, MA, USA |
Cat# bs-4189R |
αKlotho |
Abcam, Trumpington, Cambridge, UK |
Cat# ab181373 |
β-actin |
Proteintech, Rosemont, IL, USA |
Cat# 60008-1-lg |
β-catenin |
Abcam, Trumpington, Cambridge, UK |
Cat# ab6302 |
βKlotho |
GeneTex, Alton Pkwy Irvine, CA, USA |
Cat# GTX122197 |
Caspase 3 |
Proteintech, Rosemont, IL, USA |
Cat# 25546-1-AP |
CD133 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab19898 |
CD34 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab8158 |
Claudin-1 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab15098 |
C-Myc |
Abcam, Trumpington, Cambridge, UK |
Cat# ab32 |
FGF19 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab225942 |
FGF21 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab171941 |
FGF23 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab307421 |
FXR |
Abcam, Trumpington, Cambridge, UK |
Cat# ab129089 |
Histone |
Santa Cruz Biotechnology, Santa Cruz, California, USA |
Cat# sc-56695 |
ICAM |
Thermo Fisher Scientific, Rockford, IL, USA |
Cat# MA5407 |
LGR5 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab75850 |
MMP9 |
Merck Millipore, Burlington, Massachusetts, USA |
Cat# NP_004985 |
MRP2 |
Thermo Fisher Scientific, Rockford, IL, USA |
Cat# MA5-15700 |
MRP3 |
Biocompare, South San Francisco, CA, USA |
Cat# L1010 |
MyD88 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab28763 |
NF-κB |
Santa Cruz Biotechnology, Santa Cruz, California, USA |
Cat# sc-8008 |
OATP/SLCO1A2 |
Merck Millipore, Burlington, Massachusetts, USA |
Cat# SAB4502814 |
OSTβ |
Biorbyt, Cambridge, UK |
Cat# orb1964 |
PCNA |
Abcam, Trumpington, Cambridge, UK |
Cat# ab18197 |
TGFβRII |
Thermo Fisher Scientific, Rockford, IL, USA |
Cat# PA5-36115 |
TLR4 |
Thermo Fisher Scientific, Rockford, IL, USA |
Cat# MA5-16216 |
VCAM |
Abcam, Trumpington, Cambridge, UK |
Cat# ab78712 |
VEGF |
Boster Biological Technology, Pleasanton, CA, USA |
Cat# PB9071 |
VEGFR1 |
Abcam, Trumpington, Cambridge, UK |
Cat# ab32152 |
ZO-1 |
Abcam, Trumpington, Cambridge, UK, |
Cat# ab59720 |
Goat anti-mouse IgG |
MilliporeSigma, Burlington, Massachusetts |
Cat# Ap124P |
Goat anti-rabbit IgG |
Jackson ImmunoResearch Laboratories, West Grove, PA, USA |
Cat# 111-065-003 |
Goat anti-rat IgG |
Jackson ImmunoResearch Laboratories, West Grove, PA, USA |
Cat# 112-065-003 |
Table 2.
The sequences of primers used for qRT-PCR.
Table 2.
The sequences of primers used for qRT-PCR.
Gene |
Forward Primer sequences (5’ - 3’) |
Reverse Primer sequences (5’ - 3’) |
Nlrp3 |
agccttccaggatcctcttc |
cttgggcagcagtttctttc |
Asc |
gaagctgctgacagtgcaac |
gccacagctccagactcttc |
Panx-1 |
ggccacggagtatgtgttct |
tacagcagcccagcagtatg |
Pro-casp 1 |
agatggcacatttccaggac |
gatcctccagcagcaacttc |
Tnf-α |
ttgacctcagcgctgagttg |
cctgtagcccacgtcgtagc |
Il-1β |
gcaactgttcctgaactca |
ctcggagcctgtagtgcag |
Ifn-γ |
tggcatagatgtggaagaaaagag |
tgcaggattttcatgtcaccat |
Mrp2 |
gcttcccatggtgatctctt |
atcatcgcttcccaggtact |
Mrp3 |
ctgggtcccctgcatctac |
gccgtcttgagcctggataac |
Mdr1b |
gtgttaaaggggcgatgggcg |
aggcttggccagacaacagctt |
Mdr2 |
atcctatgcactggccttctggt |
gaaagcatcaatacagggggcag |
Asbt |
ggaactggctccaatatcctg |
gttcccgagtcaacccacat |
Ibabp |
acaggacttcacctggtc |
gcgctcataggtcacatc |
Gapdh |
tcaccaccatggagaaggc |
gctaagcagttggtggtgca |
Car |
ggaggaccagatctcccttc |
atttcattgccactcccaag |
Cyp3a11 |
gtgctcctagcaatcagctt |
cagtgcctaaaaatggcagagg |
Sult2a1 |
ggaaggaccacgactcataac |
gattcttcacaaggtttgtgttacc |
Bcrp |
tcgcagaaggagatgtgttgag |
ccagaatagcattaaggccagg |
Oatp2b1 |
attgcaggcatcacacaaga |
tagaagacctggcttttgcc |
Ostα |
gccaggcaggactcatatcaaa |
ggcaactgagccagtggtaaga |
Ostβ |
caggaactgctggaagaaatgc |
gcaggtcttctggtgtttctttgt |
Grp78/Bip |
acatggacctgttccgctcta |
tggctccttgccattgaaga |
Chop |
tatctcatccccaggaaacg |
gggcactgaccactctgttt |
Xbp1s |
ctgagtccgaatcaggtgcag |
gtccatgggaagatgttctgg |
Atf4 |
atggccggctatggatgat |
cgaagtcaaactctttcagatccatt |
Lgr5 |
caagccatgaccttggccctg |
tttcccagggagtggattctatt |
Atf6 |
tgggcaggactatgaagtaatg |
aggcttggccagacaacagctt |
Cyclin D1 |
ggggacaactcttaagtctcac |
ccaataaaagaccaatctctc |
Olfm4 |
cagccactttccaatttcactg |
gctggacatactccttcacctta |