Fig 2 - uploaded by Michael Downes
Content may be subject to copyright.
Effects of BDL and GW4064 on serum bilirubin concentrations and FXR-regulated genes. ( A ) Total bilirubin was measured in serum from WT and FXR-KO mice ( n ϭ 5–9) subjected to sham operation and vehicle (Veh) treatment, BDL and vehicle treatment, or BDL and GW4064 (GW) treatment. The presence of different lowercase letters indicates statistical significance ( P Ͻ 0.01) within each strain. ( B – H ) Total RNA was prepared from ileum of WT and FXR-KO mice ( n ϭ 5–9) subjected to sham operation and vehicle (Veh) treatment, BDL and vehicle treatment, or BDL and GW4064 (GW) treatment. Expression of the indicated genes was measured by RTQ-PCR by using cyclophilin as the internal control. Data represent the mean Ϯ SEM and are plotted as fold change relative to mRNA levels in sham-operated mice treated with vehicle. The presence of different lowercase letters indicates statistical significance ( P Ͻ 0.05) within each strain. 

Effects of BDL and GW4064 on serum bilirubin concentrations and FXR-regulated genes. ( A ) Total bilirubin was measured in serum from WT and FXR-KO mice ( n ϭ 5–9) subjected to sham operation and vehicle (Veh) treatment, BDL and vehicle treatment, or BDL and GW4064 (GW) treatment. The presence of different lowercase letters indicates statistical significance ( P Ͻ 0.01) within each strain. ( B – H ) Total RNA was prepared from ileum of WT and FXR-KO mice ( n ϭ 5–9) subjected to sham operation and vehicle (Veh) treatment, BDL and vehicle treatment, or BDL and GW4064 (GW) treatment. Expression of the indicated genes was measured by RTQ-PCR by using cyclophilin as the internal control. Data represent the mean Ϯ SEM and are plotted as fold change relative to mRNA levels in sham-operated mice treated with vehicle. The presence of different lowercase letters indicates statistical significance ( P Ͻ 0.05) within each strain. 

Source publication
Article
Full-text available
Obstruction of bile flow results in bacterial proliferation and mucosal injury in the small intestine that can lead to the translocation of bacteria across the epithelial barrier and systemic infection. These adverse effects of biliary obstruction can be inhibited by administration of bile acids. Here we show that the farnesoid X receptor (FXR), a...

Contexts in source publication

Context 1
... the ileum and cecum. In WT mice, BDL resulted in an increase in bacterial number in the mesenteric lymph node complex, and GW4064 treatment completely inhibited this effect. The effect was particularly pronounced for aerobic bacteria (Fig. 3 E ). FXR-KO mice had Ϸ 10-fold higher levels of aerobic bacteria in mesenteric lymph nodes as com- pared with WT animals (Fig. 3 E ; note differences in scales for WT and FXR-KO mice), and BDL caused a trend toward increased bacterial counts (Fig. 3 E and F ). This trend was not reversed by GW4064 administration (Fig. 3 E and F ). The increase in in vivo bacterial translocation suggested that BDL causes a disruption of the intestinal barrier function that is inhibited by FXR. To examine barrier integrity, immunostaining was done for occludin, a component of the epithelial tight junctions. In the villus epithelium of WT mice, occludin staining was continuous except where interrupted by the normal presence of mucus-secreting goblet cells (Fig. 4 A ). In contrast, many enterocytes in the BDL mice expressed little or no occludin (Fig. 4 A ). Consistent with these data, transmission electron microscopy revealed large ruptures in the mucosal surface of BDL WT mice, with bacteria penetrating deep into the epithelium (Fig. 4 C and D ). Hematoxylin and eosin staining revealed that the breakdown in barrier function was accompanied by dilated lymphatics and a pronounced interstitial edema, as previously described (Fig. 4 B ) (3, 25, 26, 34). In many villi, the severity of the edema caused separation of the villus epithelium from the underlying lamina propria (Fig. 4 B ). Bacteria were seen within edematous areas by electron microscopy (Fig. 4 D ). Consistent with the edema resulting from an inflammatory response, there was a marked increase in the number of neutrophils infiltrating the villi of BDL mice (Fig. 4 G ). Neutrophils are known to contribute to epithelial permeability in inflammatory diseases of the intestine (35). GW4064 prevented the pathological effects caused by BDL. Ilea from BDL mice administered GW4064 showed a more continuous pattern of occludin immunostaining (Fig. 4 A ), little or no edema (Fig. 4 B ), and a reduction in the number of neutrophils to the level seen in sham-operated mice (Fig. 4 G ). Virtually no mucosal damage was detected in electron microscopy studies done by using tissue from GW4064-treated mice (data not shown). Thus, activation of FXR prevents deterioration of the epithelial barrier and the accompanying inflamma- tion caused by biliary obstruction. No changes were seen in ileal rates of cell proliferation (assessed by immunostaining with Ki67 antibody) or apoptosis (assessed by TUNEL assays) in the different treatment groups (data not shown). Thus, FXR blocks the changes caused by BDL in WT mice without affecting cell proliferation or programmed cell death. In marked contrast to the continuous occludin immunostaining seen in sham-operated WT mice, occludin staining was discontinuous and weak in FXR-KO mice, indicating a deterioration in the epithelial barrier even without BDL (Fig. 4 A ). Consistent with this result and the higher incidence of bacterial translocation in FXR-KO mice (Fig. 3 E ), bacteria and edema were present in the junctions between epithelial cells in FXR-KO animals (Fig. 4 E ). Bacteria were also present in the dilated lymphatic vessels (Fig. 4 F ). These data demonstrate that the lack of FXR leads to mucosal injury and bacterial translocation even in the absence of additional insult. BDL did not exacerbate the edema (data not shown) or further increase the elevated neu- trophil numbers (Fig. 4 G ) in the FXR-KO mice. Moreover, GW4064 administration had no effect on any of these parame- ters in FXR-KO mice (Fig. 4 G and data not shown). Biliary obstruction causes bacterial overgrowth and translocation in the small intestine that can be reversed by administration of bile acids (3, 4, 25–30). It has been proposed that the detergent properties of bile acids are responsible for their enteroprotective actions. Although bile acids were not assessed for direct antibacterial actions in this study, our data suggest a more sophis- ticated mechanism wherein activation of the bile acid receptor FXR protects against bacterial proliferation and its detrimental effects in the distal small intestine. The products of several genes regulated by FXR in ileum, including Ang1 , iNos , and Il18 , have established antimicrobial actions. It is not possible to tell from our data whether these genes are regulated by FXR in enterocytes or other cell types, including resident immune cells. The expression profile of Ang1 as well as Fgf15 , Shp , Car12 , and Ibabp correlate well with FXR-mediated enteroprotection in the BDL model (Fig. 2), suggesting that the protective actions of FXR are likely to involve multiple downstream genes. Expression of iNos and Il18 is more complex in that their expression is increased by BDL in both WT and FXR-KO mice (Fig. 2), which likely reflects their regulation by proinflammatory signaling cascades that are activated by BDL. Nevertheless, iNOS and IL18 may contribute to FXR-induced enteroprotection under physiologic or other pathophysiologic conditions. Notably, elevated concentrations of iNOS and IL18 are associated with mucosal damage and inflammatory bowel disease (21, 36). Regulation of these genes by bile acids, which are released from the gallbladder into the small intestine during feeding, may ensure an adequate level of enteroprotection during periods of increased microbial expo- sure while preventing the overproduction of proteins that can cause inflammation and intestinal disease. In summary, our results demonstrate that FXR plays a crucial role in protecting the distal small intestine against bacterial overgrowth and the resulting disruption of the epithelial barrier. These findings raise the intriguing possibility that potent, synthetic FXR agonists may have therapeutic utility in patients with obstructed or reduced bile flow, including those on total par- enteral nutrition, who are susceptible to bacterial overgrowth and translocation (37, 38). Transcriptional Profiling Analysis. Total RNA was extracted from tissues by using RNA STAT-60 (Tel-Test, Friendswood, TX), and cDNA was prepared. Preparation of in vitro transcription products, oligonucleotide array hybridization, and scanning were performed by using Affymetrix high-density oligonucleotide array mouse genome 430A 2.0 chips according to Affymetrix protocols. To minimize discrepancies due to vari- ables, the raw expression data were scaled by using Affymetrix MICROARRAY SUITE 5.0 software, and pairwise comparisons were performed. The trimmed mean signal of all probe sets was adjusted to a user-specified target signal value (200) for each array for global scaling. No specific exclusion criteria were applied. Additional analysis was performed by using the freeware program BULLFROG 7 ...
Context 2
... in the regulatory regions of target genes. FXR is much studied in liver, where it regulates a program of genes involved in maintaining bile acid homeostasis (8). FXR is also expressed in the intestine, where it regulates several genes, including the ileal bile acid binding protein and fibroblast growth factor 15 (9–12). However, little is known about the overarching function of FXR in the gut. In this report we have examined the role of FXR in the ileum. We demonstrate that it plays a crucial role in preventing bacterial overgrowth and maintaining the integrity of the intestinal epithelium. Real-time quantitative PCR (RTQ-PCR) was used to measure FXR mRNA concentrations in duodenum, jejunum, ileum, colon, and liver. FXR was expressed in each of these tissues, with highest mRNA levels in ileum (Fig. 1 A ). A previous study showed that FXR is expressed in the villus epithelium in late-stage mouse embryos (13). Consistent with this finding, in situ hybridization analysis with ileum sections revealed that FXR is expressed in the villus epithelium in adult mouse ileum, with highest expression in the intervillus regions (Fig. 1 B ). Little or no FXR mRNA was detected in the crypts of Lieberk ̈hn, lamina propria, and tunica muscularis. To gain insight into the function of FXR in the small intestine, transcriptional profiling experiments were done by using mice lacking the gene encoding sterol 27-hydroxylase (CYP27), a key enzyme for bile acid synthesis. The CYP27-knockout (KO) mice produce only low levels of bile acids and thus are essentially devoid of endogenous FXR agonists (14). RNA was prepared from ileal mucosa of CYP27-KO mice administered either the potent, synthetic FXR agonist GW4064 (15) or vehicle for 14 h. Microarray analysis yielded a list of 15 genes whose expression was changed Ն 2-fold by GW4064 administration (Table 1). Among these were the established FXR target genes small heterodimer partner ( Shp ), fibroblast growth factor 15 ( Fgf15 ), and ileal bile acid binding protein ( Ibabp ), which encode proteins involved in bile acid homeostasis (9–12, 16, 17). Interestingly, several of the other genes in Table 1 have established roles in mucosal defense in the intestine. NO, the product of inducible NO synthase (iNOS), has direct antimicrobial effects and regulates many different aspects of the innate immune response, including mucus secretion, vascular tone, and epithelial barrier function (18, 19). The proinflammatory cytokine IL18 stimulates resistance to an array of pathogens, including intracellular and extracellular bacteria and mycobacteria, and appears to have a protective role during the early, acute phase of mucosal immune response (20, 21). Angiogenin (ANG1) and RNase A family member 4 (RNASE4) are closely related proteins that arise by differential splicing from a common gene locus (22). Although the function of RNASE4 is not established, ANG1 is part of the acute phase response to infection and has potent antibacterial and antimycotic actions (23). Carbonic anhydrase 12 (CAR12) is a member of a family of proteins involved in the maintenance of pH and ion balance (24). The regulation of all of these genes by FXR was confirmed by RTQ-PCR by using WT, CYP27-KO, and FXR-KO mice administered either GW4064 or vehicle (Fig. 5, which is published as supporting information on the PNAS web site). Notably, FXR had different effects on different genes. For example, activation of FXR had little or no effect on Ibabp or iNos expression in WT mice, but its elimination caused a marked decrease in the basal expression of these genes (Fig. 5). In contrast, FXR activation induced Il18 but did not contribute to its basal expression (Fig. 5). Based on these data, the possibility that Il18 induction by FXR is pharmacological rather than physiological cannot be excluded. Decreased bile secretion in rodents by either ligation of the common bile duct or induction of cirrhosis causes changes in the small intestine, including bacterial overgrowth and translocation across the mucosal barrier (3, 4, 25–30). Oral administration of bile acids inhibits these effects (3, 4). The genes regulated by FXR in ileum suggested that it might contribute to the enteroprotective actions of bile acids. To test this hypothesis, groups of WT and FXR-KO mice (31) were administered either GW4064 or vehicle for 2 days and then subjected to bile duct ligation (BDL) or sham operation. After 5 days, during which GW4064 or vehicle treatment was continued, the mice were killed and their intestines were analyzed for FXR target gene expression and bacterial content. As expected, BDL caused jaundice manifested by increased total serum bilirubin concentrations in both WT and FXR-KO mice (Fig. 2 A ). GW4064 administration had no effect on total serum bilirubin concentrations in BDL mice. BDL also had marked effects on genes regulated by FXR in ileum. In WT mice, expression of Fgf15 , Shp , Ang1 , Car12 , and Ibabp was reduced by BDL (Fig. 2). GW4064 administration increased expression of these genes in BDL mice to levels above those seen in sham- operated animals. None of these genes was induced by GW4064 in FXR-KO mice. In contrast, the iNos and Il18 genes showed a more complex expression pattern (Fig. 2 D and G ). Although both genes are induced by FXR (Fig. 5), there was a trend toward increased expression in BDL mice that occurred in an FXR- independent fashion (Fig. 2 D and G ). Because both iNos and Il18 are induced by proinflammatory stimuli, including lipopoly- saccharide and various cytokines (32, 33), this likely reflects regulation of these genes by additional signaling pathways that are activated by BDL. The effect of BDL and GW4064 treatment on the bacterial content of ileum and cecum was assessed. As expected, BDL increased the number of aerobic and anaerobic bacteria in the ileum and cecum of WT mice (Fig. 3 A – D ). The increase was particularly pronounced for aerobic bacteria (Fig. 3 A and C ). Notably, bacterial overgrowth in the lumen of both tissues was completely blocked by administration of GW4064 to BDL mice (Fig. 3 A – D ). To determine whether FXR is required for the antibacterial actions of GW4064, the same set of experiments was performed in FXR-KO mice. Two important differences were seen between WT and FXR-KO mice. First, GW4064 did not decrease aerobic or anaerobic bacteria counts in the ileum or cecum of FXR-KO mice (Fig. 3 A – D ). The reason for the marked variability in the bacterial counts in BDL FXR-KO mice is not known. Nevertheless, these data demonstrate that the antibacterial actions of GW4064 require FXR. Second, sham-operated FXR-KO mice had significantly higher aerobic bacteria counts than sham- operated, WT mice (Fig. 3 A and C ). These data support a role for FXR in suppressing bacterial proliferation under normal physiologic conditions. Because FXR-KO mice have reduced hepatic bile salt export pump (ABCB11) expression (31), de- creased bile acid concentrations in the intestinal lumen of these animals may contribute to the bacterial overgrowth. In comple- mentary in vitro experiments, GW4064 had no bacteriostatic effects in aerobic cultures of ileal contents even at 10 ␮ M concentrations (data not shown), demonstrating that GW4064 itself does not have direct antibacterial activity. As a measure of bacterial translocation across the mucosal barrier, bacteria were also quantified in the mesenteric lymph node complex for each of the treatment groups (Fig. 3 E and F ). The pattern for the bacterial counts was similar to that seen for bacterial overgrowth in the ileum and cecum. In WT mice, BDL resulted in an increase in bacterial number in the mesenteric lymph node complex, and GW4064 treatment completely inhibited this effect. The effect was particularly pronounced for aerobic bacteria (Fig. 3 E ). FXR-KO mice had Ϸ 10-fold higher levels of aerobic bacteria in mesenteric lymph nodes as com- pared with WT animals (Fig. 3 E ; note differences in scales for WT and FXR-KO mice), and BDL caused a trend toward increased bacterial counts (Fig. 3 E and F ). This trend was not reversed by GW4064 administration (Fig. 3 E and F ). The increase in in vivo bacterial translocation suggested that BDL causes a disruption of the intestinal barrier function that is inhibited by FXR. To examine barrier integrity, immunostaining was done for occludin, a component of the epithelial tight junctions. In the villus epithelium of WT mice, occludin staining was continuous except where interrupted by the normal presence of mucus-secreting goblet cells (Fig. 4 A ). In contrast, many enterocytes in the BDL mice expressed little or no occludin (Fig. 4 A ). Consistent with these data, transmission electron microscopy revealed large ruptures in the mucosal surface of BDL WT mice, with bacteria penetrating deep into the epithelium (Fig. 4 C and D ). Hematoxylin and eosin staining revealed that the breakdown in barrier function was accompanied by dilated lymphatics and a pronounced interstitial edema, as previously described (Fig. 4 B ) (3, 25, 26, 34). In many villi, the severity of the edema caused separation of the villus epithelium from the underlying lamina propria (Fig. 4 B ). Bacteria were seen within edematous areas by electron microscopy (Fig. 4 D ). Consistent with the edema resulting from an inflammatory response, there was a marked increase in the number of neutrophils infiltrating the villi of BDL mice (Fig. 4 G ). Neutrophils are known to contribute to epithelial permeability in inflammatory diseases of the intestine (35). GW4064 prevented the pathological effects caused by BDL. Ilea from BDL mice administered GW4064 showed a more continuous pattern of occludin immunostaining (Fig. 4 A ), little or no edema (Fig. 4 B ), and a reduction in the number of neutrophils to the level seen in sham-operated mice (Fig. 4 G ). Virtually no mucosal damage was detected in electron microscopy studies done by using tissue ...
Context 3
... barrier, bacteria were also quantified in the mesenteric lymph node complex for each of the treatment groups (Fig. 3 E and F ). The pattern for the bacterial counts was similar to that seen for bacterial overgrowth in the ileum and cecum. In WT mice, BDL resulted in an increase in bacterial number in the mesenteric lymph node complex, and GW4064 treatment completely inhibited this effect. The effect was particularly pronounced for aerobic bacteria (Fig. 3 E ). FXR-KO mice had Ϸ 10-fold higher levels of aerobic bacteria in mesenteric lymph nodes as com- pared with WT animals (Fig. 3 E ; note differences in scales for WT and FXR-KO mice), and BDL caused a trend toward increased bacterial counts (Fig. 3 E and F ). This trend was not reversed by GW4064 administration (Fig. 3 E and F ). The increase in in vivo bacterial translocation suggested that BDL causes a disruption of the intestinal barrier function that is inhibited by FXR. To examine barrier integrity, immunostaining was done for occludin, a component of the epithelial tight junctions. In the villus epithelium of WT mice, occludin staining was continuous except where interrupted by the normal presence of mucus-secreting goblet cells (Fig. 4 A ). In contrast, many enterocytes in the BDL mice expressed little or no occludin (Fig. 4 A ). Consistent with these data, transmission electron microscopy revealed large ruptures in the mucosal surface of BDL WT mice, with bacteria penetrating deep into the epithelium (Fig. 4 C and D ). Hematoxylin and eosin staining revealed that the breakdown in barrier function was accompanied by dilated lymphatics and a pronounced interstitial edema, as previously described (Fig. 4 B ) (3, 25, 26, 34). In many villi, the severity of the edema caused separation of the villus epithelium from the underlying lamina propria (Fig. 4 B ). Bacteria were seen within edematous areas by electron microscopy (Fig. 4 D ). Consistent with the edema resulting from an inflammatory response, there was a marked increase in the number of neutrophils infiltrating the villi of BDL mice (Fig. 4 G ). Neutrophils are known to contribute to epithelial permeability in inflammatory diseases of the intestine (35). GW4064 prevented the pathological effects caused by BDL. Ilea from BDL mice administered GW4064 showed a more continuous pattern of occludin immunostaining (Fig. 4 A ), little or no edema (Fig. 4 B ), and a reduction in the number of neutrophils to the level seen in sham-operated mice (Fig. 4 G ). Virtually no mucosal damage was detected in electron microscopy studies done by using tissue from GW4064-treated mice (data not shown). Thus, activation of FXR prevents deterioration of the epithelial barrier and the accompanying inflamma- tion caused by biliary obstruction. No changes were seen in ileal rates of cell proliferation (assessed by immunostaining with Ki67 antibody) or apoptosis (assessed by TUNEL assays) in the different treatment groups (data not shown). Thus, FXR blocks the changes caused by BDL in WT mice without affecting cell proliferation or programmed cell death. In marked contrast to the continuous occludin immunostaining seen in sham-operated WT mice, occludin staining was discontinuous and weak in FXR-KO mice, indicating a deterioration in the epithelial barrier even without BDL (Fig. 4 A ). Consistent with this result and the higher incidence of bacterial translocation in FXR-KO mice (Fig. 3 E ), bacteria and edema were present in the junctions between epithelial cells in FXR-KO animals (Fig. 4 E ). Bacteria were also present in the dilated lymphatic vessels (Fig. 4 F ). These data demonstrate that the lack of FXR leads to mucosal injury and bacterial translocation even in the absence of additional insult. BDL did not exacerbate the edema (data not shown) or further increase the elevated neu- trophil numbers (Fig. 4 G ) in the FXR-KO mice. Moreover, GW4064 administration had no effect on any of these parame- ters in FXR-KO mice (Fig. 4 G and data not shown). Biliary obstruction causes bacterial overgrowth and translocation in the small intestine that can be reversed by administration of bile acids (3, 4, 25–30). It has been proposed that the detergent properties of bile acids are responsible for their enteroprotective actions. Although bile acids were not assessed for direct antibacterial actions in this study, our data suggest a more sophis- ticated mechanism wherein activation of the bile acid receptor FXR protects against bacterial proliferation and its detrimental effects in the distal small intestine. The products of several genes regulated by FXR in ileum, including Ang1 , iNos , and Il18 , have established antimicrobial actions. It is not possible to tell from our data whether these genes are regulated by FXR in enterocytes or other cell types, including resident immune cells. The expression profile of Ang1 as well as Fgf15 , Shp , Car12 , and Ibabp correlate well with FXR-mediated enteroprotection in the BDL model (Fig. 2), suggesting that the protective actions of FXR are likely to involve multiple downstream genes. Expression of iNos and Il18 is more complex in that their expression is increased by BDL in both WT and FXR-KO mice (Fig. 2), which likely reflects their regulation by proinflammatory signaling cascades that are activated by BDL. Nevertheless, iNOS and IL18 may contribute to FXR-induced enteroprotection under physiologic or other pathophysiologic conditions. Notably, elevated concentrations of iNOS and IL18 are associated with mucosal damage and inflammatory bowel disease (21, 36). Regulation of these genes by bile acids, which are released from the gallbladder into the small intestine during feeding, may ensure an adequate level of enteroprotection during periods of increased microbial expo- sure while preventing the overproduction of proteins that can cause inflammation and intestinal disease. In summary, our results demonstrate that FXR plays a crucial role in protecting the distal small intestine against bacterial overgrowth and the resulting disruption of the epithelial barrier. These findings raise the intriguing possibility that potent, synthetic FXR agonists may have therapeutic utility in patients with obstructed or reduced bile flow, including those on total par- enteral nutrition, who are susceptible to bacterial overgrowth and translocation (37, 38). Transcriptional Profiling Analysis. Total RNA was extracted from tissues by using RNA STAT-60 (Tel-Test, Friendswood, TX), and cDNA was prepared. Preparation of in vitro transcription products, oligonucleotide array hybridization, and scanning were performed by using Affymetrix high-density oligonucleotide array mouse genome 430A 2.0 chips according to Affymetrix protocols. To minimize discrepancies due to vari- ables, the raw expression data were scaled by using Affymetrix MICROARRAY SUITE 5.0 software, and pairwise comparisons were performed. The trimmed mean signal of all probe sets was adjusted to a user-specified target signal value (200) for each array for global scaling. No specific exclusion criteria were applied. Additional analysis was performed by using the freeware program BULLFROG 7 ...
Context 4
... receptor family of ligand-activated transcription factors that is activated by bile acids including cholic acid and chenodeoxycholic acid (8). FXR modulates gene expression by binding as a heterodimer with the retinoid X receptors to DNA response elements in the regulatory regions of target genes. FXR is much studied in liver, where it regulates a program of genes involved in maintaining bile acid homeostasis (8). FXR is also expressed in the intestine, where it regulates several genes, including the ileal bile acid binding protein and fibroblast growth factor 15 (9–12). However, little is known about the overarching function of FXR in the gut. In this report we have examined the role of FXR in the ileum. We demonstrate that it plays a crucial role in preventing bacterial overgrowth and maintaining the integrity of the intestinal epithelium. Real-time quantitative PCR (RTQ-PCR) was used to measure FXR mRNA concentrations in duodenum, jejunum, ileum, colon, and liver. FXR was expressed in each of these tissues, with highest mRNA levels in ileum (Fig. 1 A ). A previous study showed that FXR is expressed in the villus epithelium in late-stage mouse embryos (13). Consistent with this finding, in situ hybridization analysis with ileum sections revealed that FXR is expressed in the villus epithelium in adult mouse ileum, with highest expression in the intervillus regions (Fig. 1 B ). Little or no FXR mRNA was detected in the crypts of Lieberk ̈hn, lamina propria, and tunica muscularis. To gain insight into the function of FXR in the small intestine, transcriptional profiling experiments were done by using mice lacking the gene encoding sterol 27-hydroxylase (CYP27), a key enzyme for bile acid synthesis. The CYP27-knockout (KO) mice produce only low levels of bile acids and thus are essentially devoid of endogenous FXR agonists (14). RNA was prepared from ileal mucosa of CYP27-KO mice administered either the potent, synthetic FXR agonist GW4064 (15) or vehicle for 14 h. Microarray analysis yielded a list of 15 genes whose expression was changed Ն 2-fold by GW4064 administration (Table 1). Among these were the established FXR target genes small heterodimer partner ( Shp ), fibroblast growth factor 15 ( Fgf15 ), and ileal bile acid binding protein ( Ibabp ), which encode proteins involved in bile acid homeostasis (9–12, 16, 17). Interestingly, several of the other genes in Table 1 have established roles in mucosal defense in the intestine. NO, the product of inducible NO synthase (iNOS), has direct antimicrobial effects and regulates many different aspects of the innate immune response, including mucus secretion, vascular tone, and epithelial barrier function (18, 19). The proinflammatory cytokine IL18 stimulates resistance to an array of pathogens, including intracellular and extracellular bacteria and mycobacteria, and appears to have a protective role during the early, acute phase of mucosal immune response (20, 21). Angiogenin (ANG1) and RNase A family member 4 (RNASE4) are closely related proteins that arise by differential splicing from a common gene locus (22). Although the function of RNASE4 is not established, ANG1 is part of the acute phase response to infection and has potent antibacterial and antimycotic actions (23). Carbonic anhydrase 12 (CAR12) is a member of a family of proteins involved in the maintenance of pH and ion balance (24). The regulation of all of these genes by FXR was confirmed by RTQ-PCR by using WT, CYP27-KO, and FXR-KO mice administered either GW4064 or vehicle (Fig. 5, which is published as supporting information on the PNAS web site). Notably, FXR had different effects on different genes. For example, activation of FXR had little or no effect on Ibabp or iNos expression in WT mice, but its elimination caused a marked decrease in the basal expression of these genes (Fig. 5). In contrast, FXR activation induced Il18 but did not contribute to its basal expression (Fig. 5). Based on these data, the possibility that Il18 induction by FXR is pharmacological rather than physiological cannot be excluded. Decreased bile secretion in rodents by either ligation of the common bile duct or induction of cirrhosis causes changes in the small intestine, including bacterial overgrowth and translocation across the mucosal barrier (3, 4, 25–30). Oral administration of bile acids inhibits these effects (3, 4). The genes regulated by FXR in ileum suggested that it might contribute to the enteroprotective actions of bile acids. To test this hypothesis, groups of WT and FXR-KO mice (31) were administered either GW4064 or vehicle for 2 days and then subjected to bile duct ligation (BDL) or sham operation. After 5 days, during which GW4064 or vehicle treatment was continued, the mice were killed and their intestines were analyzed for FXR target gene expression and bacterial content. As expected, BDL caused jaundice manifested by increased total serum bilirubin concentrations in both WT and FXR-KO mice (Fig. 2 A ). GW4064 administration had no effect on total serum bilirubin concentrations in BDL mice. BDL also had marked effects on genes regulated by FXR in ileum. In WT mice, expression of Fgf15 , Shp , Ang1 , Car12 , and Ibabp was reduced by BDL (Fig. 2). GW4064 administration increased expression of these genes in BDL mice to levels above those seen in sham- operated animals. None of these genes was induced by GW4064 in FXR-KO mice. In contrast, the iNos and Il18 genes showed a more complex expression pattern (Fig. 2 D and G ). Although both genes are induced by FXR (Fig. 5), there was a trend toward increased expression in BDL mice that occurred in an FXR- independent fashion (Fig. 2 D and G ). Because both iNos and Il18 are induced by proinflammatory stimuli, including lipopoly- saccharide and various cytokines (32, 33), this likely reflects regulation of these genes by additional signaling pathways that are activated by BDL. The effect of BDL and GW4064 treatment on the bacterial content of ileum and cecum was assessed. As expected, BDL increased the number of aerobic and anaerobic bacteria in the ileum and cecum of WT mice (Fig. 3 A – D ). The increase was particularly pronounced for aerobic bacteria (Fig. 3 A and C ). Notably, bacterial overgrowth in the lumen of both tissues was completely blocked by administration of GW4064 to BDL mice (Fig. 3 A – D ). To determine whether FXR is required for the antibacterial actions of GW4064, the same set of experiments was performed in FXR-KO mice. Two important differences were seen between WT and FXR-KO mice. First, GW4064 did not decrease aerobic or anaerobic bacteria counts in the ileum or cecum of FXR-KO mice (Fig. 3 A – D ). The reason for the marked variability in the bacterial counts in BDL FXR-KO mice is not known. Nevertheless, these data demonstrate that the antibacterial actions of GW4064 require FXR. Second, sham-operated FXR-KO mice had significantly higher aerobic bacteria counts than sham- operated, WT mice (Fig. 3 A and C ). These data support a role for FXR in suppressing bacterial proliferation under normal physiologic conditions. Because FXR-KO mice have reduced hepatic bile salt export pump (ABCB11) expression (31), de- creased bile acid concentrations in the intestinal lumen of these animals may contribute to the bacterial overgrowth. In comple- mentary in vitro experiments, GW4064 had no bacteriostatic effects in aerobic cultures of ileal contents even at 10 ␮ M concentrations (data not shown), demonstrating that GW4064 itself does not have direct antibacterial activity. As a measure of bacterial translocation across the mucosal barrier, bacteria were also quantified in the mesenteric lymph node complex for each of the treatment groups (Fig. 3 E and F ). The pattern for the bacterial counts was similar to that seen for bacterial overgrowth in the ileum and cecum. In WT mice, BDL resulted in an increase in bacterial number in the mesenteric lymph node complex, and GW4064 treatment completely inhibited this effect. The effect was particularly pronounced for aerobic bacteria (Fig. 3 E ). FXR-KO mice had Ϸ 10-fold higher levels of aerobic bacteria in mesenteric lymph nodes as com- pared with WT animals (Fig. 3 E ; note differences in scales for WT and FXR-KO mice), and BDL caused a trend toward increased bacterial counts (Fig. 3 E and F ). This trend was not reversed by GW4064 administration (Fig. 3 E and F ). The increase in in vivo bacterial translocation suggested that BDL causes a disruption of the intestinal barrier function that is inhibited by FXR. To examine barrier integrity, immunostaining was done for occludin, a component of the epithelial tight junctions. In the villus epithelium of WT mice, occludin staining was continuous except where interrupted by the normal presence of mucus-secreting goblet cells (Fig. 4 A ). In contrast, many enterocytes in the BDL mice expressed little or no occludin (Fig. 4 A ). Consistent with these data, transmission electron microscopy revealed large ruptures in the mucosal surface of BDL WT mice, with bacteria penetrating deep into the epithelium (Fig. 4 C and D ). Hematoxylin and eosin staining revealed that the breakdown in barrier function was accompanied by dilated lymphatics and a pronounced interstitial edema, as previously described (Fig. 4 B ) (3, 25, 26, 34). In many villi, the severity of the edema caused separation of the villus epithelium from the underlying lamina propria (Fig. 4 B ). Bacteria were seen within edematous areas by electron microscopy (Fig. 4 D ). Consistent with the edema resulting from an inflammatory response, there was a marked increase in the number of neutrophils infiltrating the villi of BDL mice (Fig. 4 G ). Neutrophils are known to contribute to epithelial permeability in inflammatory diseases of the intestine (35). GW4064 prevented the pathological effects caused by BDL. Ilea from BDL mice administered GW4064 showed a more continuous pattern of occludin ...
Context 5
... binding protein and fibroblast growth factor 15 (9–12). However, little is known about the overarching function of FXR in the gut. In this report we have examined the role of FXR in the ileum. We demonstrate that it plays a crucial role in preventing bacterial overgrowth and maintaining the integrity of the intestinal epithelium. Real-time quantitative PCR (RTQ-PCR) was used to measure FXR mRNA concentrations in duodenum, jejunum, ileum, colon, and liver. FXR was expressed in each of these tissues, with highest mRNA levels in ileum (Fig. 1 A ). A previous study showed that FXR is expressed in the villus epithelium in late-stage mouse embryos (13). Consistent with this finding, in situ hybridization analysis with ileum sections revealed that FXR is expressed in the villus epithelium in adult mouse ileum, with highest expression in the intervillus regions (Fig. 1 B ). Little or no FXR mRNA was detected in the crypts of Lieberk ̈hn, lamina propria, and tunica muscularis. To gain insight into the function of FXR in the small intestine, transcriptional profiling experiments were done by using mice lacking the gene encoding sterol 27-hydroxylase (CYP27), a key enzyme for bile acid synthesis. The CYP27-knockout (KO) mice produce only low levels of bile acids and thus are essentially devoid of endogenous FXR agonists (14). RNA was prepared from ileal mucosa of CYP27-KO mice administered either the potent, synthetic FXR agonist GW4064 (15) or vehicle for 14 h. Microarray analysis yielded a list of 15 genes whose expression was changed Ն 2-fold by GW4064 administration (Table 1). Among these were the established FXR target genes small heterodimer partner ( Shp ), fibroblast growth factor 15 ( Fgf15 ), and ileal bile acid binding protein ( Ibabp ), which encode proteins involved in bile acid homeostasis (9–12, 16, 17). Interestingly, several of the other genes in Table 1 have established roles in mucosal defense in the intestine. NO, the product of inducible NO synthase (iNOS), has direct antimicrobial effects and regulates many different aspects of the innate immune response, including mucus secretion, vascular tone, and epithelial barrier function (18, 19). The proinflammatory cytokine IL18 stimulates resistance to an array of pathogens, including intracellular and extracellular bacteria and mycobacteria, and appears to have a protective role during the early, acute phase of mucosal immune response (20, 21). Angiogenin (ANG1) and RNase A family member 4 (RNASE4) are closely related proteins that arise by differential splicing from a common gene locus (22). Although the function of RNASE4 is not established, ANG1 is part of the acute phase response to infection and has potent antibacterial and antimycotic actions (23). Carbonic anhydrase 12 (CAR12) is a member of a family of proteins involved in the maintenance of pH and ion balance (24). The regulation of all of these genes by FXR was confirmed by RTQ-PCR by using WT, CYP27-KO, and FXR-KO mice administered either GW4064 or vehicle (Fig. 5, which is published as supporting information on the PNAS web site). Notably, FXR had different effects on different genes. For example, activation of FXR had little or no effect on Ibabp or iNos expression in WT mice, but its elimination caused a marked decrease in the basal expression of these genes (Fig. 5). In contrast, FXR activation induced Il18 but did not contribute to its basal expression (Fig. 5). Based on these data, the possibility that Il18 induction by FXR is pharmacological rather than physiological cannot be excluded. Decreased bile secretion in rodents by either ligation of the common bile duct or induction of cirrhosis causes changes in the small intestine, including bacterial overgrowth and translocation across the mucosal barrier (3, 4, 25–30). Oral administration of bile acids inhibits these effects (3, 4). The genes regulated by FXR in ileum suggested that it might contribute to the enteroprotective actions of bile acids. To test this hypothesis, groups of WT and FXR-KO mice (31) were administered either GW4064 or vehicle for 2 days and then subjected to bile duct ligation (BDL) or sham operation. After 5 days, during which GW4064 or vehicle treatment was continued, the mice were killed and their intestines were analyzed for FXR target gene expression and bacterial content. As expected, BDL caused jaundice manifested by increased total serum bilirubin concentrations in both WT and FXR-KO mice (Fig. 2 A ). GW4064 administration had no effect on total serum bilirubin concentrations in BDL mice. BDL also had marked effects on genes regulated by FXR in ileum. In WT mice, expression of Fgf15 , Shp , Ang1 , Car12 , and Ibabp was reduced by BDL (Fig. 2). GW4064 administration increased expression of these genes in BDL mice to levels above those seen in sham- operated animals. None of these genes was induced by GW4064 in FXR-KO mice. In contrast, the iNos and Il18 genes showed a more complex expression pattern (Fig. 2 D and G ). Although both genes are induced by FXR (Fig. 5), there was a trend toward increased expression in BDL mice that occurred in an FXR- independent fashion (Fig. 2 D and G ). Because both iNos and Il18 are induced by proinflammatory stimuli, including lipopoly- saccharide and various cytokines (32, 33), this likely reflects regulation of these genes by additional signaling pathways that are activated by BDL. The effect of BDL and GW4064 treatment on the bacterial content of ileum and cecum was assessed. As expected, BDL increased the number of aerobic and anaerobic bacteria in the ileum and cecum of WT mice (Fig. 3 A – D ). The increase was particularly pronounced for aerobic bacteria (Fig. 3 A and C ). Notably, bacterial overgrowth in the lumen of both tissues was completely blocked by administration of GW4064 to BDL mice (Fig. 3 A – D ). To determine whether FXR is required for the antibacterial actions of GW4064, the same set of experiments was performed in FXR-KO mice. Two important differences were seen between WT and FXR-KO mice. First, GW4064 did not decrease aerobic or anaerobic bacteria counts in the ileum or cecum of FXR-KO mice (Fig. 3 A – D ). The reason for the marked variability in the bacterial counts in BDL FXR-KO mice is not known. Nevertheless, these data demonstrate that the antibacterial actions of GW4064 require FXR. Second, sham-operated FXR-KO mice had significantly higher aerobic bacteria counts than sham- operated, WT mice (Fig. 3 A and C ). These data support a role for FXR in suppressing bacterial proliferation under normal physiologic conditions. Because FXR-KO mice have reduced hepatic bile salt export pump (ABCB11) expression (31), de- creased bile acid concentrations in the intestinal lumen of these animals may contribute to the bacterial overgrowth. In comple- mentary in vitro experiments, GW4064 had no bacteriostatic effects in aerobic cultures of ileal contents even at 10 ␮ M concentrations (data not shown), demonstrating that GW4064 itself does not have direct antibacterial activity. As a measure of bacterial translocation across the mucosal barrier, bacteria were also quantified in the mesenteric lymph node complex for each of the treatment groups (Fig. 3 E and F ). The pattern for the bacterial counts was similar to that seen for bacterial overgrowth in the ileum and cecum. In WT mice, BDL resulted in an increase in bacterial number in the mesenteric lymph node complex, and GW4064 treatment completely inhibited this effect. The effect was particularly pronounced for aerobic bacteria (Fig. 3 E ). FXR-KO mice had Ϸ 10-fold higher levels of aerobic bacteria in mesenteric lymph nodes as com- pared with WT animals (Fig. 3 E ; note differences in scales for WT and FXR-KO mice), and BDL caused a trend toward increased bacterial counts (Fig. 3 E and F ). This trend was not reversed by GW4064 administration (Fig. 3 E and F ). The increase in in vivo bacterial translocation suggested that BDL causes a disruption of the intestinal barrier function that is inhibited by FXR. To examine barrier integrity, immunostaining was done for occludin, a component of the epithelial tight junctions. In the villus epithelium of WT mice, occludin staining was continuous except where interrupted by the normal presence of mucus-secreting goblet cells (Fig. 4 A ). In contrast, many enterocytes in the BDL mice expressed little or no occludin (Fig. 4 A ). Consistent with these data, transmission electron microscopy revealed large ruptures in the mucosal surface of BDL WT mice, with bacteria penetrating deep into the epithelium (Fig. 4 C and D ). Hematoxylin and eosin staining revealed that the breakdown in barrier function was accompanied by dilated lymphatics and a pronounced interstitial edema, as previously described (Fig. 4 B ) (3, 25, 26, 34). In many villi, the severity of the edema caused separation of the villus epithelium from the underlying lamina propria (Fig. 4 B ). Bacteria were seen within edematous areas by electron microscopy (Fig. 4 D ). Consistent with the edema resulting from an inflammatory response, there was a marked increase in the number of neutrophils infiltrating the villi of BDL mice (Fig. 4 G ). Neutrophils are known to contribute to epithelial permeability in inflammatory diseases of the intestine (35). GW4064 prevented the pathological effects caused by BDL. Ilea from BDL mice administered GW4064 showed a more continuous pattern of occludin immunostaining (Fig. 4 A ), little or no edema (Fig. 4 B ), and a reduction in the number of neutrophils to the level seen in sham-operated mice (Fig. 4 G ). Virtually no mucosal damage was detected in electron microscopy studies done by using tissue from GW4064-treated mice (data not shown). Thus, activation of FXR prevents deterioration of the epithelial barrier and the accompanying inflamma- tion caused by biliary obstruction. No changes were seen in ileal rates of cell proliferation (assessed by immunostaining ...
Context 6
... of FXR in the ileum. We demonstrate that it plays a crucial role in preventing bacterial overgrowth and maintaining the integrity of the intestinal epithelium. Real-time quantitative PCR (RTQ-PCR) was used to measure FXR mRNA concentrations in duodenum, jejunum, ileum, colon, and liver. FXR was expressed in each of these tissues, with highest mRNA levels in ileum (Fig. 1 A ). A previous study showed that FXR is expressed in the villus epithelium in late-stage mouse embryos (13). Consistent with this finding, in situ hybridization analysis with ileum sections revealed that FXR is expressed in the villus epithelium in adult mouse ileum, with highest expression in the intervillus regions (Fig. 1 B ). Little or no FXR mRNA was detected in the crypts of Lieberk ̈hn, lamina propria, and tunica muscularis. To gain insight into the function of FXR in the small intestine, transcriptional profiling experiments were done by using mice lacking the gene encoding sterol 27-hydroxylase (CYP27), a key enzyme for bile acid synthesis. The CYP27-knockout (KO) mice produce only low levels of bile acids and thus are essentially devoid of endogenous FXR agonists (14). RNA was prepared from ileal mucosa of CYP27-KO mice administered either the potent, synthetic FXR agonist GW4064 (15) or vehicle for 14 h. Microarray analysis yielded a list of 15 genes whose expression was changed Ն 2-fold by GW4064 administration (Table 1). Among these were the established FXR target genes small heterodimer partner ( Shp ), fibroblast growth factor 15 ( Fgf15 ), and ileal bile acid binding protein ( Ibabp ), which encode proteins involved in bile acid homeostasis (9–12, 16, 17). Interestingly, several of the other genes in Table 1 have established roles in mucosal defense in the intestine. NO, the product of inducible NO synthase (iNOS), has direct antimicrobial effects and regulates many different aspects of the innate immune response, including mucus secretion, vascular tone, and epithelial barrier function (18, 19). The proinflammatory cytokine IL18 stimulates resistance to an array of pathogens, including intracellular and extracellular bacteria and mycobacteria, and appears to have a protective role during the early, acute phase of mucosal immune response (20, 21). Angiogenin (ANG1) and RNase A family member 4 (RNASE4) are closely related proteins that arise by differential splicing from a common gene locus (22). Although the function of RNASE4 is not established, ANG1 is part of the acute phase response to infection and has potent antibacterial and antimycotic actions (23). Carbonic anhydrase 12 (CAR12) is a member of a family of proteins involved in the maintenance of pH and ion balance (24). The regulation of all of these genes by FXR was confirmed by RTQ-PCR by using WT, CYP27-KO, and FXR-KO mice administered either GW4064 or vehicle (Fig. 5, which is published as supporting information on the PNAS web site). Notably, FXR had different effects on different genes. For example, activation of FXR had little or no effect on Ibabp or iNos expression in WT mice, but its elimination caused a marked decrease in the basal expression of these genes (Fig. 5). In contrast, FXR activation induced Il18 but did not contribute to its basal expression (Fig. 5). Based on these data, the possibility that Il18 induction by FXR is pharmacological rather than physiological cannot be excluded. Decreased bile secretion in rodents by either ligation of the common bile duct or induction of cirrhosis causes changes in the small intestine, including bacterial overgrowth and translocation across the mucosal barrier (3, 4, 25–30). Oral administration of bile acids inhibits these effects (3, 4). The genes regulated by FXR in ileum suggested that it might contribute to the enteroprotective actions of bile acids. To test this hypothesis, groups of WT and FXR-KO mice (31) were administered either GW4064 or vehicle for 2 days and then subjected to bile duct ligation (BDL) or sham operation. After 5 days, during which GW4064 or vehicle treatment was continued, the mice were killed and their intestines were analyzed for FXR target gene expression and bacterial content. As expected, BDL caused jaundice manifested by increased total serum bilirubin concentrations in both WT and FXR-KO mice (Fig. 2 A ). GW4064 administration had no effect on total serum bilirubin concentrations in BDL mice. BDL also had marked effects on genes regulated by FXR in ileum. In WT mice, expression of Fgf15 , Shp , Ang1 , Car12 , and Ibabp was reduced by BDL (Fig. 2). GW4064 administration increased expression of these genes in BDL mice to levels above those seen in sham- operated animals. None of these genes was induced by GW4064 in FXR-KO mice. In contrast, the iNos and Il18 genes showed a more complex expression pattern (Fig. 2 D and G ). Although both genes are induced by FXR (Fig. 5), there was a trend toward increased expression in BDL mice that occurred in an FXR- independent fashion (Fig. 2 D and G ). Because both iNos and Il18 are induced by proinflammatory stimuli, including lipopoly- saccharide and various cytokines (32, 33), this likely reflects regulation of these genes by additional signaling pathways that are activated by BDL. The effect of BDL and GW4064 treatment on the bacterial content of ileum and cecum was assessed. As expected, BDL increased the number of aerobic and anaerobic bacteria in the ileum and cecum of WT mice (Fig. 3 A – D ). The increase was particularly pronounced for aerobic bacteria (Fig. 3 A and C ). Notably, bacterial overgrowth in the lumen of both tissues was completely blocked by administration of GW4064 to BDL mice (Fig. 3 A – D ). To determine whether FXR is required for the antibacterial actions of GW4064, the same set of experiments was performed in FXR-KO mice. Two important differences were seen between WT and FXR-KO mice. First, GW4064 did not decrease aerobic or anaerobic bacteria counts in the ileum or cecum of FXR-KO mice (Fig. 3 A – D ). The reason for the marked variability in the bacterial counts in BDL FXR-KO mice is not known. Nevertheless, these data demonstrate that the antibacterial actions of GW4064 require FXR. Second, sham-operated FXR-KO mice had significantly higher aerobic bacteria counts than sham- operated, WT mice (Fig. 3 A and C ). These data support a role for FXR in suppressing bacterial proliferation under normal physiologic conditions. Because FXR-KO mice have reduced hepatic bile salt export pump (ABCB11) expression (31), de- creased bile acid concentrations in the intestinal lumen of these animals may contribute to the bacterial overgrowth. In comple- mentary in vitro experiments, GW4064 had no bacteriostatic effects in aerobic cultures of ileal contents even at 10 ␮ M concentrations (data not shown), demonstrating that GW4064 itself does not have direct antibacterial activity. As a measure of bacterial translocation across the mucosal barrier, bacteria were also quantified in the mesenteric lymph node complex for each of the treatment groups (Fig. 3 E and F ). The pattern for the bacterial counts was similar to that seen for bacterial overgrowth in the ileum and cecum. In WT mice, BDL resulted in an increase in bacterial number in the mesenteric lymph node complex, and GW4064 treatment completely inhibited this effect. The effect was particularly pronounced for aerobic bacteria (Fig. 3 E ). FXR-KO mice had Ϸ 10-fold higher levels of aerobic bacteria in mesenteric lymph nodes as com- pared with WT animals (Fig. 3 E ; note differences in scales for WT and FXR-KO mice), and BDL caused a trend toward increased bacterial counts (Fig. 3 E and F ). This trend was not reversed by GW4064 administration (Fig. 3 E and F ). The increase in in vivo bacterial translocation suggested that BDL causes a disruption of the intestinal barrier function that is inhibited by FXR. To examine barrier integrity, immunostaining was done for occludin, a component of the epithelial tight junctions. In the villus epithelium of WT mice, occludin staining was continuous except where interrupted by the normal presence of mucus-secreting goblet cells (Fig. 4 A ). In contrast, many enterocytes in the BDL mice expressed little or no occludin (Fig. 4 A ). Consistent with these data, transmission electron microscopy revealed large ruptures in the mucosal surface of BDL WT mice, with bacteria penetrating deep into the epithelium (Fig. 4 C and D ). Hematoxylin and eosin staining revealed that the breakdown in barrier function was accompanied by dilated lymphatics and a pronounced interstitial edema, as previously described (Fig. 4 B ) (3, 25, 26, 34). In many villi, the severity of the edema caused separation of the villus epithelium from the underlying lamina propria (Fig. 4 B ). Bacteria were seen within edematous areas by electron microscopy (Fig. 4 D ). Consistent with the edema resulting from an inflammatory response, there was a marked increase in the number of neutrophils infiltrating the villi of BDL mice (Fig. 4 G ). Neutrophils are known to contribute to epithelial permeability in inflammatory diseases of the intestine (35). GW4064 prevented the pathological effects caused by BDL. Ilea from BDL mice administered GW4064 showed a more continuous pattern of occludin immunostaining (Fig. 4 A ), little or no edema (Fig. 4 B ), and a reduction in the number of neutrophils to the level seen in sham-operated mice (Fig. 4 G ). Virtually no mucosal damage was detected in electron microscopy studies done by using tissue from GW4064-treated mice (data not shown). Thus, activation of FXR prevents deterioration of the epithelial barrier and the accompanying inflamma- tion caused by biliary obstruction. No changes were seen in ileal rates of cell proliferation (assessed by immunostaining with Ki67 antibody) or apoptosis (assessed by TUNEL assays) in the different treatment groups (data not shown). Thus, FXR blocks the changes caused by BDL in WT mice without ...

Similar publications

Article
Full-text available
While the ontogeny and recruitment of the intestinal monocyte/macrophage lineage has been studied extensively, their precise localization and function has been overlooked. Here we show by imaging the murine small and large intestines in steady-state that intestinal CX3CR1+ macrophages form an interdigitated network intimately adherent to the entire...

Citations

... For patients with MAFLD, the activation of bile acid receptors appears to play a beneficial role in reducing disease severity. FXR activation has been shown to decrease steatosis by inhibiting lipogenesis, reduce chemically induced hepatic inflammation and fibrosis, and maintain intestinal barrier integrity, thereby protecting the liver from inflammatory signals originating from gut bacteria [52,53]. Similarly, TGR5 activation has been demonstrated to mitigate high-fat diet-induced glucose intolerance, insulin resistance, and inflammation, while also protecting against lipopolysaccharide-induced inflammation [54,55]. ...
... Additionally, BAs can suppress intestinal dendritic cell (DC) differentiation and activation via FXR [62,63], and regulate inflammatory responses through TGR5 activation, leading to decreased IFN-γ, IL-1β, IL-6, and TNF-α, and increased IL-10 [42]. FXR and TGR5 activation can also improve metabolic health by reducing steatosis (fatty liver) via inhibiting lipogenesis and decreasing hepatic inflammation [52], as well as by mitigating high-fat diet (HFD)-induced glucose intolerance and insulin resistance [53]. ...
Article
Full-text available
Metabolic-associated fatty liver disease (MAFLD) is a risk factor for severe COVID-19. This study explores the potential influence of gut hormone receptor and immune response gene expression on COVID-19 outcomes in MAFLD patients. Methods: We investigated gene expression levels of AHR, FFAR2, FXR, and TGR5 in patients with MAFLD and COVID-19 compared to controls. We examined associations between gene expression and clinical outcomes. Results: COVID-19 patients displayed altered AHR expression, potentially impacting immune response and recovery. Downregulated AHR in patients with MAFLD correlated with increased coagulation parameters. Elevated FFAR2 expression in patients with MAFLD was linked to specific immune cell populations and hospital stay duration. A significantly lower FXR expression was observed in both MAFLD and severe COVID-19. Conclusion: Our findings suggest potential modulatory roles for AHR, FFAR2, and FXR in COVID-19 and MAFLD.
... For patients with MAFLD, activation of bile acid receptors appears to play a beneficial role in reducing disease severity. FXR activation has been shown to decrease steatosis by inhibiting lipogenesis, reduce chemically induced hepatic inflammation and fibrosis, and maintain intestinal barrier integrity, thereby protecting the liver from inflammatory signals originating from gut bacteria [45,46]. Similarly, TGR5 activation has been demonstrated to mitigate high-fat diet-induced glucose intolerance, insulin resistance, and inflammation, while also protecting against lipopolysaccharideinduced inflammation [47,48]. ...
... Additionally, BAs can suppress intestinal dendritic cell (DC) differentiation and activation via FXR [53,54], and regulate inflammatory responses through TGR5 activation, leading to decreased IFN-γ, IL-1β, IL-6, and TNFα, and increased IL-10 [35]. FXR and TGR5 activation can also improve metabolic health by reducing steatosis (fatty liver) via inhibiting lipogenesis and decreasing hepatic inflammation [45], as well as by mitigating high-fat diet (HFD)-induced glucose intolerance and insulin resistance [46]. ...
Preprint
Full-text available
Metabolic-associated fatty liver disease (MAFLD) is a risk factor for severe COVID-19. This study explores the potential influence of gut hormone receptor and immune response gene expression on COVID-19 outcomes in MAFLD patients. Methods: We investigated gene expression levels of AHR, FFAR2, FXR, and TGR5 in patients with MAFLD and COVID-19 compared to controls. We examined associations between gene expression and clinical outcomes. Results: COVID-19 patients displayed altered AHR expression, potentially impacting immune response and recovery. Downregulated AHR in MAFLD patients correlated with increased coagulation parameters. Elevated FFAR2 expression in MAFLD linked to specific immune cell populations and hospital stay duration. Significantly lower FXR expression was observed in both MAFLD and severe COVID-19. Conclusion: Our findings suggest potential modulatory roles for AHR, FFAR2, and FXR in COVID-19 and MAFLD. Keywords: COVID-19; MAFLD; AHR; FXR; Gene expression
... showed Proteobacteria-derived lipopolysaccharide suppressed FXR activity and expression in mouse liver [47]. Porphyromonadaceae abundance negatively correlated with FXR signaling in patients with NAFLD [21]. ...
Article
Full-text available
Obesity is associated with alterations in lipid metabolism and gut microbiota dysbiosis. This study investigated the effects of puerarin, a bioactive isoflavone, on lipid metabolism disorders and gut microbiota in high-fat diet (HFD)-induced obese mice. Supplementation with puerarin reduced plasma alanine aminotransferase, liver triglyceride, liver free fatty acid (FFA), and improved gut microbiota dysbiosis in obese mice. Puerarin’s beneficial metabolic effects were attenuated when farnesoid X receptor (FXR) was antagonized, suggesting FXR-mediated mechanisms. In hepatocytes, puerarin ameliorated high FFA-induced sterol regulatory element-binding protein (SREBP) 1 signaling, inflammation, and mitochondrial dysfunction in an FXR-dependent manner. In obese mice, puerarin reduced liver damage, regulated hepatic lipogenesis, decreased inflammation, improved mitochondrial function, and modulated mitophagy and ubiquitin-proteasome pathways, but was less effective in FXR knockout mice. Puerarin upregulated hepatic expression of FXR, bile salt export pump (BSEP), and downregulated cytochrome P450 7A1 (CYP7A1) and sodium taurocholate transporter (NTCP), indicating modulation of bile acid synthesis and transport. Puerarin also restored gut microbial diversity, the Firmicutes/Bacteroidetes ratio, and the abundance of Clostridium celatum and Akkermansia muciniphila. This study demonstrates that puerarin effectively ameliorates metabolic disturbances and gut microbiota dysbiosis in obese mice, predominantly through FXR-dependent pathways. These findings underscore puerarin’s potential as a therapeutic agent for managing obesity and enhancing gut health, highlighting its dual role in improving metabolic functions and modulating microbial communities.
... These bile salts play an essential role in fat and fat-soluble vitamin absorption, along with protecting against bacterial overgrowth, immunomodulation, enhancement of the epithelial layer integrity, and indirectly inhibiting production of pro-inflammatory cytokines. 21 When a dysbiotic state occurs, the SCFA and bile salt milieu is changed, resulting in a profound metabolic change. Recurrent C difficile infection (CDI) is also associated with reduced inferred microbiota bile salt hydrolases functionality/recoverability of bile salt hydrolases after antibiotic Q7 s. 15 Therefore, by restoring the microbiota via FMT and correcting the dysbiotic state, we are able to correct the problem by directly supplementing SCFAs and bile salts with the contents administered and/or indirectly via changes in metabolic output from the newly supplemented microbial species. ...
... FXR is activated by both primary and secondary bile acids, with the most potent agonist being the primary bile acid chenodeoxycholic acid (CDCA) [44]. The upregulated expression of FXR in the ileum, when compared to the colon in IBD patients and HIs, is consistent with the findings of Inagaki et al., who reported the greater mRNA expression of this receptor in the ileum of mice when compared to other intestinal sites [45]. Our results also indicate that FXR expression is downregulated by inflammation, and indeed that TNF-α and IL-1α treatments have been reported to repress the expression of FXR in Hep3B cells in vitro [46]; however, the fact that the observed FXR downregulation in UC was greater when comparing the non-inflamed colonic mucosa of UC patients with HIs suggests that the dysregulation of this gene may not just be an effect of inflammatory stimulation. ...
Article
Full-text available
Metabolites produced by dysbiotic intestinal microbiota can influence disease pathophysiology by participating in ligand–receptor interactions. Our aim was to investigate the differential expression of metabolite receptor (MR) genes between inflammatory bowel disease (IBD), healthy individuals (HIs), and disease controls in order to identify possible interactions with inflammatory and fibrotic pathways in the intestine. RNA-sequencing datasets containing 643 Crohn’s disease (CD) patients, 467 ulcerative colitis (UC) patients and 295 HIs, and 4 Campylobacter jejuni-infected individuals were retrieved from the Sequence Read Archive, and differential expression was performed using the RaNA-seq online platform. The identified differentially expressed MR genes were used for correlation analysis with up- and downregulated genes in IBD, as well as functional enrichment analysis using a R based pipeline. Overall, 15 MR genes exhibited dysregulated expression in IBD. In inflamed CD, the hydroxycarboxylic acid receptors 2 and 3 (HCAR2, HCAR3) were upregulated and were associated with the recruitment of innate immune cells, while, in the non-inflamed CD ileum, the cannabinoid receptor 1 (CNR1) and the sphingosine-1-phospate receptor 4 (S1PR4) were downregulated and were involved in the regulation of B-cell activation. In inflamed UC, the upregulated receptors HCAR2 and HCAR3 were more closely associated with the process of TH-17 cell differentiation, while the pregnane X receptor (NR1I2) and the transient receptor potential vanilloid 1 (TRPV1) were downregulated and were involved in epithelial barrier maintenance. Our results elucidate the landscape of metabolite receptor expression in IBD, highlighting associations with disease-related functions that could guide the development of new targeted therapies.
... Furthermore, obeticholic acid and other FXR agonists restore microbiota composition, enhance intestinal innate defenses, mitigate intestinal inflammation, and lower bacterial translocation and endotoxemia in experimental cirrhosis [55,90,91]. Reduced ileal FXR signaling is a probable result of a decrease in primary bile acids and a rise in secondary bile acids in the gut, in addition to intestinal inflammation [55,92,93]. The aforementioned irregularities in intestinal barrier function in cirrhosis have been associated with alterations in the intestinal structure, encompassing submucosal edema, modest immune cell infiltration, and the disorganization of interepithelial tight junction proteins in humans and experimental models of cirrhosis [86,87,[93][94][95][96]. ...
Article
Full-text available
Portal hypertension (PH) is a complex clinical challenge with severe complications, including variceal bleeding, ascites, hepatic encephalopathy, and hepatorenal syndrome. The gut microbiota (GM) and its interconnectedness with human health have emerged as a captivating field of research. This review explores the intricate connections between the gut and the liver, aiming to elucidate how alterations in GM, intestinal barrier function, and gut-derived molecules impact the development and progression of PH. A systematic literature search, following PRISMA guidelines, identified 12 original articles that suggest a relationship between GM, the gut–liver axis, and PH. Mechanisms such as dysbiosis, bacterial translocation, altered microbial structure, and inflammation appear to orchestrate this relationship. One notable study highlights the pivotal role of the farnesoid X receptor axis in regulating the interplay between the gut and liver and proposes it as a promising therapeutic target. Fecal transplantation experiments further emphasize the pathogenic significance of the GM in modulating liver maladies, including PH. Recent advancements in metagenomics and metabolomics have expanded our understanding of the GM’s role in human ailments. The review suggests that addressing the unmet need of identifying gut–liver axis-related metabolic and molecular pathways holds potential for elucidating pathogenesis and directing novel therapeutic interventions.
... Conversely, bile acids can also modulate the composition of the gut microbiota through FXR. Studies revealed that FXR inhibited bacterial overgrowth and mucosal injury in the ileum of mice subjected to bile duct ligation, thereby decreasing bacterial translocation, which suggests that FXR agonists may prevent epithelial injury and bacterial translocation in patients with impaired bile flow [101,102]. This demonstrates the protective role of FXR in the gut-liver axis. ...
Preprint
Full-text available
Primary biliary cholangitis (PBC) is a cholestatic liver disease characterized by immune-mediated injury to small bile ducts. Although PBC is an autoimmune disease, the results of traditional immunosuppressive therapy are disappointing. Nearly 40% of PBC patients do not respond to the first-line drug UDCA. Without appropriate intervention, PBC patients would eventually progress to liver cirrhosis and even death. There is an urgent need to develop new therapies. The gut-liver axis emphasizes the interconnection between the gut and the liver, and there is increasing evidence that gut microbiota and bile acids play an important role in cholestatic diseases. Dysbiosis of gut microbiota, imbalance of bile acids, and immune-mediated bile duct injury constitute the triad of pathophysiology in PBC. Understanding the gut microbiota-bile acid network and corresponding immune functions in PBC provides a new perspective for its treatment strategies. Therefore, we summarize the latest advancements in research of gut microbiota and bile acids in PBC and, for the first time, explore the potential of immune factors related to them as novel immunotherapy targets. This article discusses potential therapeutic approaches focusing on regulating gut microbiota, maintaining bile acid homeostasis, their interactions, and related immune factors.
... Additionally, the conversion of primary bile acids to secondary bile acids occurs within the colon, a process confined to a specific subset of clostridial species and facilitated by 7α/β-dehydroxylation enzymes [63]. Bile acids exert a significant impact on the composition and density of the gut microbiota: activation of FXR in the small intestine hinders bacterial overgrowth and translocation [66,67]. Bile acids exhibit both direct antimicrobial effects, exemplified by cholic acid (CA) and deoxycholic acid (DCA) on Bifidobacterium breve and Lactobacillus salivarius [68], and indirect effects, including the stimulation of host production of antimicrobial peptides such as cathelicidin [69,70], angiogenin I [66], and inducible nitric oxide synthase [71]. ...
... Bile acids exert a significant impact on the composition and density of the gut microbiota: activation of FXR in the small intestine hinders bacterial overgrowth and translocation [66,67]. Bile acids exhibit both direct antimicrobial effects, exemplified by cholic acid (CA) and deoxycholic acid (DCA) on Bifidobacterium breve and Lactobacillus salivarius [68], and indirect effects, including the stimulation of host production of antimicrobial peptides such as cathelicidin [69,70], angiogenin I [66], and inducible nitric oxide synthase [71]. CA could induce an increase in Clostridia and subclass Erysipelotrichi while reducing members of the phyla Bacteroidetes and Actinobacteria [72]. ...
... Bile Acids in IBD: ↓ Secondary BA, ↑ Primary BA − Lipid digestion and absorption [55] − ↓ lipogenesis and hepatic gluconeogenesis [56,57] − Liver regeneration [59][60][61] − Production of antimicrobial peptides [59][60][61] − Intestinal barrel homeostasis and regeneration [58] − Intestinal wound healing [62] − Bacterial overgrowth and translocation [66,67] − Increasing inflammation (↓ IL-10, Treg, M2) and inability to produce a regulatory response during inflammation [78] ...
Article
Full-text available
In recent years, there has been a growing focus on the intricate interplay between the gut microbiota and host health, specifically in the context of inflammatory bowel diseases (IBDs). The gut microbiota produces a diverse array of metabolites, influencing the host’s immune response and tissue homeostasis. Noteworthy metabolites, such as short-chain fatty acids, bile acids, and indoles, exert significant effects on intestinal inflammation and fibrosis. This review integrates current research findings to clarify the mechanisms through which gut microbiota metabolites contribute to the progression of IBD and fibrosis, offering insights into potential therapeutic targets and strategies for managing these intricate gastrointestinal conditions. The unraveling of the complex relationship between gut microbiota metabolites and inflammatory processes holds promise for the development of targeted interventions that could lead to more effective and personalized treatment approaches for individuals affected by IBD and subsequent intestinal fibrosis.
... Bile acids modulate lipid and glucose/insulin metabolism and inflammation [82]. The gut microbiota further modifies primary bile acids to produce secondary bile acids, which interact with various receptors to influence cardiometabolic phenotypes and disease susceptibility [83][84][85][86]. Notably, alterations in levels of bile acids in plasma have been correlated with insulin resistance in type 2 diabetes mellitus [82]. ...
Article
Full-text available
Recent studies have shown that a pro-inflammatory diet and dysbiosis, especially a high level of trimethylamine-N-oxide (TMAO), are associated with various adverse health conditions. Cardiovascular diseases and pancreatic diseases are two major morbidities in the modern world. Through this narrative review, we aimed to summarize the association between a pro-inflammatory diet, gut microbiota, and cardiovascular and pancreatic diseases, along with their underlying mechanisms. Our review revealed that TMAO is associated with the development of cardiovascular diseases by promoting platelet aggregation, atherosclerotic plaque formation, and vascular inflammation. TMAO is also associated with the development of acute pancreatitis. The pro-inflammatory diet is associated with an increased risk of pancreatic cancer and cardiovascular diseases through mechanisms that include increasing TMAO levels, activating the lipopolysaccharides cascade, and the direct pro-inflammatory effect of certain nutrients. Meanwhile, an anti-inflammatory diet decreases the risk of cardiovascular diseases and pancreatic cancer.
... The ileum exhibits the highest expression levels of FXR along the gastrointestinal tract, and thus, is considered the most relevant intestinal segment involved in FXR-FGF19 signaling [35,36]. Reduced intestinal FXR activation was reported in rats with CCl 4 -induced cirrhosis (with ascites) and bile-duct ligated (BDL) mice after 5 days (i.e. a noncirrhotic stage) [4,35]. ...
... The ileum exhibits the highest expression levels of FXR along the gastrointestinal tract, and thus, is considered the most relevant intestinal segment involved in FXR-FGF19 signaling [35,36]. Reduced intestinal FXR activation was reported in rats with CCl 4 -induced cirrhosis (with ascites) and bile-duct ligated (BDL) mice after 5 days (i.e. a noncirrhotic stage) [4,35]. In our study cohort, FGF19 expression in the ileum decreased in patients with dACLD, as compared to controls, being linked to lower expression of FXR and SHP in ileum biopsies. ...
Article
Full-text available
Background and aims Experimental studies linked dysfunctional Farnesoid X receptor (FXR)-fibroblast growth factor 19 (FGF19) signaling to liver disease. This study investigated key intersections of the FXR-FGF19 pathway along the gut–liver axis and their link to disease severity in patients with cirrhosis. Methods Patients with cirrhosis undergoing hepatic venous pressure gradient measurement (cohort-I n = 107, including n = 53 with concomitant liver biopsy; n = 5 healthy controls) or colonoscopy with ileum biopsy (cohort-II n = 37; n = 6 controls) were included. Hepatic and intestinal gene expression reflecting FXR activation and intestinal barrier integrity was assessed. Systemic bile acid (BA) and FGF19 levels were measured. Results Systemic BA and FGF19 levels correlated significantly ( r = 0.461; p < 0.001) and increased with cirrhosis severity. Hepatic SHP expression decreased in patients with cirrhosis (vs. controls; p < 0.001), indicating reduced FXR activation in the liver. Systemic FGF19 ( r = −0.512, p < 0.001) and BA ( r = −0.487, p < 0.001) levels correlated negatively with hepatic CYP7A1, but not SHP or CYP8B1 expression, suggesting impaired feedback signaling in the liver. In the ileum, expression of FXR, SHP and FGF19 decreased in patients with cirrhosis, and interestingly, intestinal FGF19 expression was not linked to systemic FGF19 levels. Intestinal zonula occludens-1, occludin, and alpha-5-defensin expression in the ileum correlated with SHP and decreased in patients with decompensated cirrhosis as compared to controls. Conclusions FXR-FGF19 signaling is dysregulated at essential molecular intersections along the gut–liver axis in patients with cirrhosis. Decreased FXR activation in the ileum mucosa was linked to reduced expression of intestinal barrier proteins. These human data call for further mechanistic research on interventions targeting the FXR-FGF19 pathway in patients with cirrhosis. Clinical trial number: NCT03267615 Graphical abstract Physiology of enterohepatic FXR-FGF19 signaling and its regulation in patients with advanced chronic liver disease (ACLD). (FXR) farnesoid X receptor; (FGF19) fibroblast growth factor 19; (BA) bile acids; (c/dACLD) compensated/decompensated advanced chronic liver disease; (FXR) farnesoid X receptor; (SHP) small heterodimer partner; (OST-α/-β) organic solute transporter subunit alpha/beta; (CYP7A1) cholesterol 7 alpha-hydroxylase; (NTCP) Na+-taurocholate cotransporting polypeptide; (CYP8B1) sterol 12-alpha-hydroxylase; (HVPG) hepatic venous pressure gradient; (TJ) tight junctions; (AMP) antimicrobial peptides; (ASBT) Apical Sodium Dependent Bile Acid Transporter; (ZO 1) zonula occludens-1; (OCLN) occluding; (DEFA5) alpha-5-defensin.