ABSTRACT
Toll-like receptor 4 (TLR4) is an innate immune receptor responsive to lipopolysaccharide (LPS). Single nucleotide polymorphisms (SNPs) in human TLR4 that encode an A896G transition at SNP rs4986790 (D299G) and a C1196T transition at SNP rs4986791 (T399I) render individuals hyporesponsive to LPS. In humans, these SNPs are also associated with increased susceptibility to inflammatory bowel diseases (IBDs). Using knock-in mice engineered to express the murine homologs of these human TLR4 mutations (“TLR4-SNP” mice), we have shown that TLR4-SNP mice develop significantly more severe colitis induced by dextran sodium sulfate (DSS) than wild-type (WT) mice, similar to IBD in humans expressing these SNPs. Previous studies have provided indirect evidence for “tissue repair” M2 macrophages (Mφ) in the resolution of colitis. Signaling through the IL-4/IL-13 receptor, IL-4Rα, and the transcription factor, peroxisome proliferator-activated receptor (PPARγ), have been shown to be required for induction of M2a Mφ, and our data provide direct evidence for the involvement of both in the repair of DSS-induced colonic damage. In response to DSS, colons of TLR4-SNP mice produced reduced levels of M2a Mφ marker mRNA and protein, including PPARγ, and therapeutic administration of the PPARγ agonist ligand, rosiglitazone, resolved colitis in TLR4-SNP mice, and increased expression of the M2a protein, Ym1. Together, these data indicate that the failure of TLR4-SNP mice to resolve DSS-induced colitis may be secondary to their failure to induce “tissue repair” M2a Mφ.
Importance
Inflammatory bowel disease (IBD), including Crohn’s disease and ulcerative colitis, impacts millions of individuals worldwide and severely impairs the quality of life for patients. Dysregulation of innate immune signaling pathways reduces barrier function and exacerbates disease progression. Macrophage (Mφ) signaling pathways are potential targets for IBD therapies. While multiple treatments are available for IBD, (i) not all patients respond, (ii) responses may diminish over time, and (iii) treatments often have undesirable side effects. Genetic studies have shown that the inheritance of two co-segregating SNPs expressed in the innate immune receptor, TLR4, is associated with human IBD. Mice expressing homologous SNPs (“TLR4-SNP” mice) exhibited more severe colitis than WT mice in a DSS-induced colonic inflammation/repair model. We identified a critical role for M2a “tissue repair” Mφ in the resolution of colitis. Our findings provide insight into potential development of novel therapies targeting Mφ signaling pathways that aim to alleviate the debilitating symptoms experienced by individuals with IBD.
KEYWORDS: TLR4 SNPs, DSS-induced colitis, M2a macrophages, IL-4Rα, PPARγ, rosiglitazone
INTRODUCTION
Toll-like receptor 4 (TLR4) is a pattern recognition receptor (PRR) predominantly expressed on innate immune cells, such as macrophages (Mφ) and dendritic cells, and responds to the prototype ligand, Gram-negative lipopolysaccharide (LPS) (1, 2), as well as to other microbial “pathogen-associated molecular patterns” (PAMPs) and host-derived “danger-associated molecular patterns” (DAMPs) (3 – 5). TLR4 is unique in that (i) it requires a co-receptor, MD-2, for ligand binding and dimerization, and (ii) in response to ligand, TLR4 activates two distinct intracellular signaling pathways, the MyD88 and TRIF signaling pathways (6, 7). Activation of these two pathways results in transcriptional responses leading to production of cytokines, chemokines, and interferons. While TLR4-mediated inflammatory responses to infection or trauma are intended to protect the host, an overexuberant response may lead to tissue damage.
Single nucleotide polymorphisms (SNPs) in human TLR4, an A896G transition at SNP rs4986790 (D299G) and a C1196T transition at SNP rs4986791 (T399I), were originally reported to cause TLR4 hyporesponsiveness to LPS in humans (8). Human TLR4 proteins containing the D299G and the co-segregating D299G/T399I mutations in the extracellular domain have since been associated with increased susceptibility to many diseases, e.g., Gram negative sepsis (9), inflammatory bowel disease (IBD) (10), respiratory syncytial virus (RSV) (11 – 14), and others (15 – 21). In other disease models, expression of these same SNPs is known to be protective, including in influenza virus infection (14), graft-versus-host disease (22), atherogenesis (23), acute coronary syndrome (24), rheumatoid arthritis (25), and leprosy (26).
Expression of the D299G/T399I SNPs in TLR4 affects its functionality (8, 14, 27), possibly by altering TLR4 expression and/or by interfering with TLR4 dimerization and recruitment of adapter molecules (8, 14, 27 – 32). The D299G mutation is thought to have arisen in Africa because it offered protection from malaria, while the T399I mutation likely emerged in individuals who migrated from Africa to Europe under selective pressure exerted by the plague (33). Genetic analyses showed that the D299G/T399I SNPs, expressed in the ectodomain of human TLR4, often co-segregate with a carrier frequency of 10%–18% in Caucasians (33). Our laboratory engineered a knock-in mouse strain that homozygously expresses homologous SNPs (D298G/N397I) in murine TLR4, and this model displays similar LPS-hyporesponsiveness and altered susceptibility to infection as previously reported in human studies (14).
IBD, comprising Crohn’s disease (CD) and ulcerative colitis (UC), impacts millions globally (34) and presents with relapsing and remitting colonic inflammation that interferes with quality of life (35, 36). Sex differences exist in IBD prevalence, with early onset CD (<16 years) being slightly more common in males, and UC occurring more frequently in older males (>45 years) (37). Controlling inflammation is challenging and although a variety of treatments are available (38 – 40), not all patients respond to treatment and treatment efficacy can wane over time, highlighting the need for new, and possibly individualized, therapeutics to improve IBD management and patient outcomes.
Even prior to the identification of TLR4 as the LPS sensor (1, 2), inbred mouse strains that exhibited LPS unresponsiveness (e.g., C3H/HeJ and C57BL/10ScN) (41, 42) also differed in susceptibility to dextran sodium sulfate (DSS)-induced colitis (43, 44). It was later discovered that these two mouse strains had naturally occurring mutations in the Tlr4 gene (1, 2). Subsequently, LPS-unresponsive TLR4−/− mice (45) were shown to develop more severe DSS-induced colitis than WT mice (46, 47). TLR4 has been reported to be upregulated in Mφ and intestinal epithelial cells during intestinal inflammation (48 – 50). Although the connection between impaired TLR4 sensing and increased sensitivity to IBD was noted many years ago, the underlying mechanisms remain elusive.
Main contributors to the complexity of IBD pathogenesis include dysregulation of the mucosal immune system, alteration of the gut microflora, and impairment of the epithelial barrier function (38, 39, 51 – 53). IL-10-deficient mice develop spontaneous colitis associated with an exuberant Th1 response and uncontrolled cytokine production by Mφ (54). The inflammatory response associated with IBD has been attributed to the action of Th17 cells (38, 39, 55, 56). Functioning cellular programs for intestinal homeostasis, immune tolerance, and epithelial repair are key to maintaining gut health (53). Genome-wide association studies have identified IBD susceptibility loci that are related to epithelial barrier function and innate and adaptive immunity (38, 53, 57), emphasizing the importance of genetic predisposition and the likely polygenic control of IBD, including genetic variation in TLRs (58, 59). However, the precise mechanism(s) by which the TLR4 299/399 SNPs increase sensitivity to IBD in humans is unknown.
Herein, we show that, like humans with IBD, the TLR4-SNP mice exhibit much more severe colitis in response to DSS than WT mice. Using IL-4Rα−/− mice and mice with a conditional deletion of PPARγ in myeloid cells (PPARγcKO), that fail to respond normally to IL-4 or IL-13 to induce Mφ with an M2a “repair” phenotype, we show that both IL-4Rα- and PPARγ-dependent signaling are required for amelioration of DSS-induced colonic tissue injury. DSS-treated TLR4-SNP mice exhibited a reduced capacity to express M2a Mφ markers, and their colitic disease was ameliorated by therapeutic treatment with a PPARγ agonist ligand.
RESULTS
TLR4 SNPs exacerbate colonic inflammation in DSS-treated mice
Previous studies reported that TLR4−/− mice are more sensitive to DSS-induced colitis than WT mice (46, 47). In humans, the TLR4 SNPs encoding D299G and T399I lead to LPS hyporesponsiveness (8) and are associated with IBD (10, 60 – 73). To better understand the role of TLR4 SNPs in IBD, we utilized “TLR4-SNP” knock-in mice that express two homologous point mutations in Tlr4 (encoding D298G/N397I) and have been shown to result in LPS hyporesponsivness and related phenotypes (14). We hypothesized that mice expressing these homologous SNPs would exhibit a more severe disease phenotype upon DSS administration, a widely recognized model for chemically-induced colonic tissue inflammation, injury, and repair resembling human colitis (56, 74 – 78). Both WT C57BL/6J and TLR4-SNP mice (on a C57BL/6J background) received 3% DSS in their drinking water for 8 days ad libitum (“tissue injury phase,” Days 0–7), followed by 7 days of regular drinking water (“tissue recovery phase,” Days 8–14) (Fig. 1A). Control mice received regular drinking water only.
Fig 1.
TLR4-SNP mice are more sensitive to DSS-induced colonic inflammation than C57BL/6J mice. (A) Schematic of the DSS experimental set-up. Experimental mice were given 3% DSS in their drinking water beginning on Day 0 and were switched to regular drinking water on Day 8, with fresh DSS water replaced on Days 2, 4, and 6. Control mice received regular drinking water throughout the experiment. On alternating days, stool consistency and fecal occult blood were assessed. Mice were euthanized and colons were resected on Day 7 or on Day 14. (B) Mean ± SEM colonic symptom scores (stool consistency score + fecal occult blood score) over time in control and DSS-treated WT (C57BL/6J) and TLR4-SNP mice (males and females combined). (C) Comparison of male only (left panel, blue graph) and female only (right panel, pink graph), control and DSS-treated, WT vs TLR4-SNP mouse responses. Two-way ANOVA with Sidak post-hoc multiple comparisons test. **P < 0.01, ***P < 0.001, ****P < 0.0001. On the female graph, # indicates days in which 3% DSS-treated female TLR4-SNP mice were determined to have significantly lower colonic symptom scores compared to 3% DSS-treated male TLR4-SNP mice (Student’s t-test). Results are derived from five independent experiments in which, together, control WT (n = 14; 6 males); control TLR4-SNP (n = 18; 11 males); 3% DSS WT Days 0–7 (n = 57; 30 males), Days 7–14 (n = 31; 17 males); 3% DSS TLR4-SNP Days 0–7 (n = 58; males), Days 7–14 (n = 32; 18 males). (D) Colonic permeability in WT vs TLR4-SNP male mice measured by the concentration of 4 kDa fluorescein isothiocyanate (FITC)-dextran in sera 4 h after administration of FITC-dextran. One-tailed Student’s t-test at each time point. Results are derived from four independent experiments in which, together, control WT (n = 10); control TLR4-SNP (n = 12); 3% DSS WT, Day 7 (n = 18), Day 14 (n = 15); 3% DSS TLR4-SNP Day 7 (n = 16), Day 14 (n = 17). (E) Representative colons from DSS-treated WT and TLR4-SNP mice at Day 14 (left panel). Mean colon lengths (showing individual responses) at Days 7 and 14 in control and DSS-treated WT and TLR4-SNP mice: males and females combined (center panel) and males vs females (right panel). Two-way ANOVA with Sidak (males and females combined) or Tukey (males vs females) post-hoc multiple comparisons test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Results are derived from five independent experiments in which, together, control WT (n = 14; 6 males); control TLR4-SNP (n = 18; 11 males); 3% DSS WT, Day 7 (n = 26; 13 males), Day 14 (n = 31; 17 males); 3% DSS TLR4-SNP, Day 7 (n = 26; 13 males), Day 14 (n = 32; 18 males).
TLR4-SNP mice developed more severe colitis than WT mice throughout the experimental period as measured by “colonic symptom score” (Fig. 1B). When the responses of male and female TLR4-SNP mice were directly compared, male TLR4-SNP mice exhibited both enhanced symptom scores from Days 0 to 7 and a reduced capacity for recovery from Days 8 to 14 (Fig. 1C, left panel) compared to female TLR4-SNP mice (Fig. 1C, right panel; significant differences between males and females are indicated by #). Human IBD and DSS-induced colitis are associated with increased colonic permeability (55, 76, 79 – 86). Relative colonic permeability, assessed by detecting serum fluorescence in mice 4 h after oral administration of 4 kDa fluorescein isothiocyanate (FITC)-dextran, revealed no differences in permeability between control WT and TLR4-SNP mice (Fig. 1D). By Day 7, both strains exhibited increased serum FITC-dextran levels, with TLR4-SNP mice trending toward significantly greater colonic permeability compared to WT mice (Fig. 1D). By Day 14, WT and TLR4-SNP mice exhibited reduced serum levels of FITC-dextran compared to Day 7 levels; however, TLR4-SNP mice had significantly higher serum fluorescence than WT mice (Fig. 1D), suggesting that TLR4-SNP mice have an impaired ability to repair colonic damage. In addition, at Days 7 and 14, TLR4-SNP mice exhibited significantly shorter colon lengths, indicative of an increased inflammatory state, compared to WT mice (Fig. 1E, left and center panels); however, no sex differences in colon length were observed (Fig. 1E, right panel).
Disease severity was also assessed in H&E-stained micrographs of the distal 1 cm colon (DC) of WT and TLR4-SNP mice. No significant differences were observed between untreated WT and TLR4-SNP mice (Fig. 2A, top row). On Day 7, DSS-treated WT mice exhibited few inflammatory infiltrates in the mucosa and submucosal regions compared to non-DSS-treated control mice and the crypt structure remained largely intact (Fig. 2A, middle row). In contrast, by Day 7, DSS-treated TLR4-SNP mice displayed disrupted crypt structures with inflammatory infiltrates in the mucosa and submucosal regions, accompanied by epithelial damage (Fig. 2A, middle row). By Day 14, the crypt structures of DSS-treated TLR4-SNP mice were severely disrupted by the extensive inflammatory infiltrates in the mucosa (Fig. 2A, bottom row; red line) and submucosal (Fig. 2A, bottom row; red arrowhead) regions in contrast to WT mice. These histopathological differences between WT and TLR4-SNP mice were more pronounced in male mice (Fig. 2A, left panels) and confirmed by histologic scoring (Fig. 2B) and measurements of the muscularis externa, submucosal height, and mucosal depth (Fig. 2C). The notable colonic shortening in DSS-treated TLR4-SNP mice that was observed grossly at Day 14 (Fig. 1E) correlated with broadening of the muscularis mucosa (Fig. 2C, left panel). Moreover, inflammatory infiltrates accumulated and contributed to edema in the submucosa (Fig. 2A, bottom row) which correlated with increased submucosal height (Fig. 2C, center panel), while observed crypt regeneration in response to damage was evidenced by increased mucosal depth (Fig. 2C, right panel). At higher magnification, Day 14 DSS-treated TLR4-SNP mice exhibited substantially greater cellular infiltration into the colonic mucosa and submucosa than WT mice (Fig. 2D), with infiltrating cells that were predominantly monocytic (Fig. 2E). Collectively, our observations of worse colonic symptoms, reduced colon lengths, and more severe histopathology in TLR4-SNP mice indicate an impaired ability to recover from DSS-induced colonic injury. These findings prompted us to investigate possible mechanism(s) underlying the lack of recovery in the TLR4-SNP mice.
Fig 2.
TLR4-SNP mice develop exacerbated colonic histopathology by Day 14. (A) Representative H&E-stained images of control and DSS-treated WT and TLR4-SNP distal 1 cm colon (DC) sections at Days 7 and 14 (males and females). In the TLR4-SNP Male Day 14 image, the red line indicates the region of extensive inflammatory cells in the mucosa; red arrowhead indicates inflammatory cells infiltrating into the submucosa. (B) Histology scores of H&E-stained DC sections from control and DSS-treated WT vs TLR4-SNP mice at Days 7 and 14 (showing individual responses): males and females combined (top panel) and males vs females (bottom panel). Two-way ANOVA with Sidak post-hoc multiple comparisons test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Results were derived from five independent experiments in which, together, control WT (n = 14; 6 males); control TLR4-SNP (n = 18; 11 males); 3% DSS WT, Day 7 (n = 26; 13 males), Day 14 (n = 31; 17 males); 3% DSS TLR4-SNP, Day 7 (n = 26; 13 males), Day 14 (n = 32; 18 males). (C) Measurements of histologic region sizes (mucosa depth, submucosal height, muscularis externa height) from control and experimental H&E-stained DC sections at Days 7 and 14. Two-way ANOVA with Tukey’s post-hoc multiple comparisons test. *P < 0.05; **P < 0.01; ****P < 0.0001. Results are derived from five independent experiments in which, together, control WT (n = 14; 6 males); control TLR4-SNP (n = 18; 11 males); 3% DSS WT Day 7 (n = 26; 13 males), Day 14 (n = 27; 13 males); 3% DSS TLR4-SNP Day 7 (n = 26; 13 males), Day 14 (n = 27; 13 males). (D) Additional microscopic images were taken of the identical tissue sections shown in Fig. 2A (DSS, male WT vs TLR4-SNP, Day 14) at both 10× and 100× magnification to better illustrate the observed inflammatory differences between the two mouse strains and to allow for quantification of the infiltrating cell types as shown in Fig. 2E. (E) Number of granulocytes (left panel) and monocytes (right panel) in 100× high power H&E-stained DC sections of WT and TLR4-SNP mice. Two-way ANOVA with Sidak’s post-hoc multiple comparisons test. ***P < 0.001; ****P < 0.0001. Data are derived from two independent experiments, n = 5 mice/strain for each time point.
IL-4Rα-dependent signaling is necessary for tissue recovery from DSS-induced colonic damage
There are many studies that indicate that the microbiome is important in DSS-induced colitis (87 – 92). However, we initially carried out co-housing experiments in which WT and TLR4-SNP mice were co-housed for 3 weeks prior to DSS treatment and throughout the entire experiment to normalize their microbiomes (93, 94). Nonetheless, the TLR4-SNP mice remained much more sensitive to DSS (data not shown), indicating that the increased sensitivity of TLR4-SNP mice to DSS was not likely attributable to dysbiosis.
There is also substantial indirect evidence in the literature that anti-inflammatory M2-like Mφ contribute to resolution of DSS-induced colitis (40, 95 – 110). For example, adoptive transfer of in vitro-derived IL-4/IL-13-stimulated M2a Mφ into mice depleted of resident Mφ resulted in reduced dinitrobenzene sulfonic acid (DNBS)-induced colitis symptoms in mice (110). While M2 Mφ subsets are defined by their various inducers and anti-inflammatory products produced (111 – 114), we next focused our studies on the potential role of M2a Mφ since the only known inducers of this subset are IL-4 and IL-13 via signaling through a shared IL-4Rα chain (115, 116). IL-4Rα−/− mice fail to generate significant M2a Mφ in response to IL-4 or IL-13 (117). For this reason, we sought direct evidence for a role for M2a Mφ in recovery from DSS-induced colitis by comparing the sensitivity of WT (BALB/c) and IL-4Rα−/− (on a BALB/c background) mice to DSS.
We first confirmed that IL-4Rα−/− mice do not express IL-4Rα (Fig. 3A) by measuring Il4ra mRNA in colons of WT vs IL-4Rα−/− mice. WT mice displayed unaltered Il4ra mRNA levels in response to DSS treatment, while Il4ra mRNA levels in IL-4Rα−/− colons were at or below the limit of detection. Following the tissue injury phase, IL-4Rα−/− mice exhibited an impaired ability to recover from DSS-induced colonic injury compared to WT mice evidenced by statistically increased colonic symptom scores beginning at Day 10 (Fig. 3B, left panel) with males being more sensitive than females (Fig. 3B, right panel). By Day 14, the IL-4Rα−/− mice exhibited significantly shorter colons than WT mice (Fig. 3C) and increased pathology (Fig. 3D), which was confirmed by histologic scoring (Fig. 3E). Few inflammatory infiltrates were seen in the mucosal and submucosal regions in DSS-treated WT mice at Day 14 compared to non-DSS treated controls and the crypt structure remained intact in contrast to the IL-4Rα−/− mice (Fig. 3D). At Day 14, DSS-treated IL-4Rα−/− mice exhibited visibly disrupted crypts, with abundant inflammatory infiltrates in the mucosal and submucosal regions and extensive epithelial damage (Fig. 3D). No sex differences were observed for colon length or histology scores (Fig. 3C and E). These findings are the first to directly support the conclusion that IL-4- and/or IL-13-induced signaling via IL-4Rα is necessary for tissue recovery from DSS-induced damage.
Fig 3.
IL-4Rα−/− mice are more sensitive to DSS-induced colitis than WT BALB/cByJ WT mice. (A) qRT-PCR was carried out on MC sections for Il4ra gene expression in control and DSS-treated WT and IL-4Rα−/− mice to confirm genotype. (B) Colonic symptom scores (mean ± SEM) over time in control and DSS-treated WT vs IL-4Rα−/− mice (left panel, responses of males and females mice combined; right panel, comparison of male vs female responses to DSS). Two-way ANOVA with Sidak’s post-hoc multiple comparisons test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0001. (C) Colon lengths at Day 14 in control and experimental WT vs IL-4Rα−/− mice. Far left panel, representative DSS-treated WT and IL-4Rα−/− colons at Day 14. Middle panel, male and female responses combined. Two-way ANOVA with Sidak post-hoc multiple comparisons test. Far right panel, DSS-treated male vs female IL-4Rα−/− mice. Unpaired two-tailed Student’s t-test. (D) Representative H&E-stained images of DC sections from control and DSS-treated WT and IL-4Rα−/− mice at Day 14. (E) Histology scores of H&E-stained DC sections from control and DSS-treated WT and IL-4Rα−/− mice at Day 14 (left panel, males and females combined; right panel, male vs female, DSS-treated IL-4Rα−/− mice). Two-way ANOVA with Sidak post-hoc multiple comparisons test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Right panel: Histology scores at Day 14 in male vs female DSS-treated IL-4Rα−/− mice. Unpaired two-tailed Student’s t-test. Results are derived from three independent experiments in which, together, control WT (n = 10; 5 males), control IL-4Rα−/− (n = 13; 5 males); 3% DSS WT Day 14 (n = 10; 5 males); 3% DSS IL-4Rα−/− Day 14 (n = 23; 13 males).
TLR4-SNP mice exhibit a reduced capacity to induce M2a Mφ in response to DSS
Since IL-4Rα signaling was required for the repair of DSS-induced colitis, we next sought to determine if the worsened colitis of DSS-treated TLR4-SNP mice was attributable to a decreased capacity for differentiation of M2a Mφ. Since the repair phase occurs between Days 8 and 14, Days 9 and 11 were selected to measure the induction of M2a Mφ markers in the colons of WT and TLR4-SNP mice. We compared the induction of M2a genes in colon sections of control and DSS-treated mice. M2a Mφ markers, Arg1, and Chil3 mRNA levels were significantly reduced in TLR4-SNP colons at Days 9 and 11, while Mrc1 mRNA showed a trend toward reduced M2a gene expression in TLR4-SNP colon sections compared to WT mice, particularly at Day 9 (Fig. 4A). These observations were confirmed by Western analysis showing reduced Arginase 1 (Arg1), Ym1 (Chil3) (Fig. 4B), and Mrc1 (Fig. 4C) protein levels in TLR4-SNP colon homogenates at Day 11. These findings indicate that TLR4-SNP mice have an impaired ability to induce M2a Mφ markers in response to DSS.
Fig 4.
DSS-treated TLR4-SNP mice exhibit reduced M2a markers in the colon and reduced β-hydroxybutyrate (β-HB) levels in the sera. (A) M2a gene expression measured in the MC of male WT vs TLR4-SNP mice, comparing control, Day 9, and Day 11 samples by qRT-PCR. One-way ANOVA with Sidak multiple comparisons test. *P < 0.05; **P < 0.01; ****P < 0.0001. Data are derived from two independent experiments. In total, control WT n = 11; control TLR4-SNP n = 15; WT Day 9 n = 13, Day 11 n = 13; TLR4-SNP Day 9 n = 15, Day 11 n = 15. (B) M2a (Ym1, Arg1) protein production measured in DC samples in WT vs TLR4-SNP mice, comparing control and Day 11 samples. Data are representative of results from two independent experiments totaling control WT n = 5; control TLR4-SNP n = 5; WT Day 11 n = 9; TLR4-SNP Day 11 n = 9. (C) M2a (Mrc1, PPARγ) protein production measured in DC samples in WT vs TLR4-SNP mice, comparing control and Day 11 samples. Data representative of results from two independent experiments totaling control WT n = 5; control TLR4-SNP n = 5; WT Day 11 n = 9; TLR4-SNP Day 11 n = 9. (D) β-HB concentration measured in the sera of control and DSS-treated, WT vs TLR4-SNP male mice at Days 9 and 11. Two-way ANOVA with Sidak multiple comparisons. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Data are derived from three independent experiments in which, together, control WT n = 12; control TLR4-SNP n = 13; WT Day 11 n = 10; TLR4-SNP Day 11 n = 10.
Reduced circulating levels of β-hydroxybutyrate in TLR4-SNP mice
β-Hydroxybutyrate (β-HB) is a metabolite produced by both animals and bacteria (118 – 120) and a hallmark of Gram-negative bacteria (121 – 124). Numerous studies have implicated the microbiome in IBD pathogenesis (87 – 92), noting significantly reduced β-HB levels in the colonic mucosa (103) and sera (125, 126) of patients with IBD. Introduction of bacteria engineered to produce β-HB (127) or intrarectal administration of β-HB (103) improved DSS-induced symptoms in mice. In vitro, Huang et al. showed that β-HB synergistically enhanced IL-4-induced signaling and, in vivo, exogenous administration of β-HB to WT mice induced the M2a genes Arg1 and Chil3 (103). Therefore, we hypothesized that the observed defect in repair in the TLR4-SNP mice could be attributable, in part, to impaired M2a Mφ differentiation driven by β-HB. In sera of DSS-treated TLR4-SNP mice, β-HB levels were significantly reduced at Day 11 compared to WT mice (Fig. 4D).
TLR4-SNP mice produce reduced levels of PPARγ in response to DSS
Previous studies reported that differentiation of M2a Mφ is dependent on the ligand-activated transcription factor, PPARγ (128 – 138). PPARγ is a nuclear hormone receptor (NHR) that forms a heterodimer complex with another NHR, retinoid X receptor (RXR), to regulate the expression of various adipocyte-specific and M2a Mφ differentiation genes (128 – 130, 139, 140). PPARγ is highly expressed in adipose tissue and the colon (141), particularly in epithelial cells (142) and Mφ (143). Rectal biopsies from patients with active UC show reduced PPARγ mRNA and protein levels in the colonic mucosa (144, 145). Animal models of IBD revealed that PPARγ influences disease timing and severity (146). Consistent with reduced levels of Ym1 (Chil3), Arg1, and Mrc1 proteins in colon homogenates (Fig. 4B and C), DSS-treated TLR4-SNP mice also produced lower levels of PPARγ protein when compared to WT mice (Fig. 4C).
The PPARγ agonist ligand, rosiglitazone, improved DSS-induced colitis in TLR4-SNP mice therapeutically
To confirm the role of PPARγ in DSS-induced colitis, we utilized PPARγ conditional knockout (PPARγcKO) mice originally derived by crossing homozygous floxed PPARγ mice with a transgenic mouse containing the Cre recombinase gene under the control of the murine Lysozyme M (Lyz2) promoter to delete the Pparg gene in lysozyme-producing cells (i.e., predominantly Mφ and neutrophils) (133, 147 – 149). A lack of PPARγ protein expression in medium- or IL-4-stimulated, PPARγcKO vs WT thioglycollate-elicited peritoneal Mφ was confirmed by Western blot (Fig. 5A). Although DSS-treated PPARγcKO mice presented on Days 2 and 4 with milder colonic symptoms compared to DSS-treated WT mice, their disease severity significantly increased between Days 10 and 14, suggesting an impaired ability to recover (Fig. 5B). By Day 14, PPARγcKO mice exhibited significantly shorter colon lengths (Fig. 5C) and increased histopathology (Fig. 5D) than WT mice, phenotypes also observed in DSS-treated TLR4-SNP and IL-4Rα−/− mice. No sex differences were observed for any of the parameters measured.
Fig 5.
PPARγcKO mice are more sensitive to DSS-induced colitis than WT mice. (A) PPARγ protein expression in thioglycollate-elicited macrophages from WT and PPARγcKO mice in the absence or presence of IL-4 (24 h) to confirm genotype. (B) Colonic symptom scores (Mean ± SEM) in control and DSS-treated WT vs PPARγcKO mice (responses of male and female mice combined). Two-way ANOVA with Sidak’s post-hoc multiple comparisons test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0001. (C) Representative colon lengths at Day 14 in control and DSS-treated WT vs PPARγcKO mice. Left panel, gross image of representative DSS-treated WT and PPARγcKO colons on Day 14. Right panel, male and female responses combined. Two-way ANOVA with Sidak’s post-hoc multiple comparisons test. ****P < 0001. (D) Left panel: representative H&E-stained images of DC sections. Right panel: histology scores of H&E-stained DC sections from control and DSS-treated WT and PPARγcKO mice on Day 14, males and females combined. Two-way ANOVA with Sidak post-hoc multiple comparisons test. ****P < 0001. Results were derived from three independent experiments in which in total control WT (n = 5), control PPARγcKO (n = 5), 3% DSS WT (n = 16 males), 3% DSS PPARγcKO (n = 12; 8 males). Although the early kinetics of DSS-induced disease were delayed in PPARγcKO compared to WT mice, we observed deaths in four males and one female, suggesting that myeloid PPARγ plays a key role in tissue repair.
Thiazolidinedione (TZD) agents, such as rosiglitazone, are PPARγ agonist ligands shown previously to ameliorate DSS-induced colitis in mice (139, 150 – 154) and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in rats (155). TLR4-SNP mice were administered 3% DSS in their drinking water starting on Day 0 and then treated therapeutically with saline or rosiglitazone (once daily from Day 2 to Day 7). On Days 8 to 14, all mice were given regular drinking water. Compared to DSS + saline-treated TLR4-SNP mice, TLR4-SNP mice treated with DSS + rosiglitazone displayed reduced clinical symptoms (Fig. 6A), increased colon lengths at Day 14 (Fig. 6B), repaired crypt structure, and reduced numbers of infiltrating inflammatory cells (Fig. 6C). Under high power, the infiltrating cells in the DSS + saline-treated TLR4-SNP mice were predominantly mononuclear with a few granulocytic cells (indicated with red arrows; Fig. 6D). Histopathological changes were reflected in the histology scores (Fig. 6E). Consistent with a role for M2a Mϕ in the resolution of DSS-induced colitis, the rosiglitazone-treated TLR4-SNP mice exhibited increased expression of the M2a protein, Ym1 (Chil3), compared to the saline-treated group (Fig. 6F). These findings indicate that therapeutic treatment with the PPARγ agonist ligand improves DSS-induced colitis symptoms in the hypersensitive TLR4-SNP mice.
Fig 6.
Administration of rosiglitazone, a PPARγ agonist ligand, ameliorates DSS-induced colonic damage in TLR4-SNP mice. Male TLR4-SNP mice were administered DSS starting on Day 0. On Days 2–7, mice were administered either saline or rosiglitazone (25 mg/kg) once daily i.p. (A) Average colonic symptom score over time in saline- and rosiglitazone-treated male TLR4-SNP mice. Two-way ANOVA with Sidak’s multiple comparisons; *P < 0.05. (B) Colon lengths at Day 14 in saline- and rosiglitazone-treated TLR4-SNP mice. Unpaired two-tailed Students t-test. ***P < 0.001. (C) Representative H&E-stained images of DC sections from saline- and rosiglitazone-treated TLR4-SNP mice at Day 14. (D) High-power image of submucosal infiltrating inflammatory cells in a representative DSS- and saline-treated TLR4-SNP mouse on Day 14. The infiltrating cells are predominantly mononuclear with occasional granulocytic cells (red arrows). (E) Right panel: histology scores of H&E-stained DC sections from saline- and rosiglitazone-treated TLR4-SNP mice at Day 14. Unpaired two-tailed Students t-test; **P < 0.01. Results were derived from three independent experiments. In total, TLR4-SNP saline n = 17; TLR4-SNP rosiglitazone n = 19. (F) Ym1 (Chil3) protein expression at Day 11 in DC of DSS-treated TLR4-SNP mice therapeutically administered rosiglitazone or saline. Data are representative of results from two independent experiments totaling TLR4-SNP rosiglitazone-treated n = 9; TLR4-SNP saline-treated n = 8.
DISCUSSION
The D299G and T399I substitutions in human TLR4 have been shown to exacerbate or ameliorate inflammation in different diseases and disease models, including infection, inflammation, and cancer (3, 58, 156). In IBD, the majority of independent association studies and meta-analyses reported an association between inheritance of human TLR4 SNPs encoding D299G/T399I and increased risk of IBD in humans (10, 60 – 70); however, some studies with smaller sample sizes reported no significant association (65 – 67, 71 – 73). Early studies showed that mice with natural or engineered deficiencies in TLR4 signaling (e.g., C3H/HeJ, C57BL/10ScN, TLR4−/−) exhibited increased susceptibility to chemically-induced colitis (43, 46, 47). Rakoff-Nahoum et al. attributed the increased mortality of MyD88−/− mice and TLR4−/− mice to increased colonic bleeding resulting from epithelial denudation and glandular mucodepletion (46). The mechanism(s) by which reduced TLR4 expression contributes to colitis sensitivity are clearly multifaceted and still not entirely understood. While commensal overgrowth and hyperinfiltrating leukocytes were ruled out as potential contributors to disease, dysregulated epithelial cell proliferation and differentiation and compromised expression of cytoprotective and repair factors like IL-6, TNF, KC-1, and heat shock proteins were identified as potential contributing factors (46). The presence of the microbiome was required for cytokine production by the colon and recognition of commensal microbial ligands by TLRs was essential for protection from DSS-induced epithelial injury and mortality (46). For this reason, it is surprising that targeting of TLR4 (157) early during DSS treatment by administration of a monoclonal antibody directed against the TLR4-MD-2 complex (158) or a synthetic TLR4 antagonist (159) reduced disease. In contrast, Fukata et al. reported increased bacterial translocation in DSS-treated TLR4−/− mice and reduced neutrophil recruitment due to impaired MIP-2 production by TLR4-deficient Mφ (47).
Since the human D299G/T399I mutations in TLR4 are co-inherited by a significant proportion of individuals (33), “TLR4-SNP” mice were engineered to enable mechanistic studies by which these TLR4 SNPs impact diseases. To date, these mice have been shown to reflect phenotypes in humans, including LPS hyporesponsiveness, increased sensitivity to Gram negative infection and RSV, and increased resistance to influenza infection (14). In addition, the TLR4-SNP mice exhibit reduced allergic inflammation with LPS and OVA but increased inflammation, eosinophilic infiltration, and Th2 cytokines following house dust mite allergen induction, reflecting a significant effect of the environment on their Th2-dependent responses (160). Further, TLR4-SNP mice show reduced antibody responses and a deficiency in class switching (also Th2-dependent) in response to immunization to T-dependent antigen when adjuvanted with a lipid A-based adjuvant (161).
Although DSS is widely used to induce colitic disease in mice, we recognize that this model has limitations (162). DSS is toxic to epithelial cells resulting in loss of barrier integrity by redistribution of the tight junction components, increased permeability, increased epithelial apoptosis and proinflammatory cytokines, and entry of luminal antigens and microorganisms that induce inflammation (162). The pathogenesis of IBD is complex, and many mechanisms have been implicated (51 – 53). In healthy individuals, multiple cellular programs coordinate to effectively promote healing of damaged tissues. One cell type that is particularly important for healing is the alternatively activated or repair “M2” Mφ.
Mφ mediate innate immune responses to PAMPs and DAMPs and play a key role in tissue repair and regeneration (163 – 165). The presence of cytokines and other environmental signals promotes Mφ polarization to a variety of phenotypes. Mφ are often described in terms of a “differentiation spectrum,” with “classically activated” “M1” Mφ, induced by potent inflammatory stimuli such as LPS and IFN-γ, at one end of the differentiation spectrum, and “alternatively activated” or “tissue repair” (166) “M2” Mφ at the other end of the spectrum (113). While M2 Mφ differentiation was originally attributed to signaling by IL-4 and IL-13 through a shared IL-4Rα chain to activate M2-associated genes (116, 166), the definition of “M2” Mφ is now far more nuanced and subsets have been defined (M2a, M2b, M2c, M2d, regulatory) based on distinct inducing agents and transcriptomic patterns (112, 114).
By using IL-4Rα−/− mice, our study has provided the first direct experimental evidence for the role of IL-4/13 signaling in the repair of DSS-induced colitis in WT mice (Fig. 3). IL-4 and IL-13 signaling through a shared receptor, IL-4Rα, leads to activation of specific transcription factors, e.g., STAT-6, EGR-2 (112, 167), that, in turn, recruit the transcription factor PPARγ to activate promoters of specific “M2a” genes (112, 130, 167, 168). We extended our findings in the IL-4Rα−/− mice by showing that mice in which PPARγ had been deleted in myeloid cells (133, 147 – 149) were also extremely sensitive to DSS-induced colitis (Fig. 5). We recognize that LysMCre deletion is not absolute and that PPARγ on other cell types (e.g., epithelial cells) remains functional and may even compensate to some degree for the lack of PPARγ on myeloid cells. Nonetheless, like DSS-treated IL-4Rα−/− and TLR4-SNP mice, the PPARγcKO mice phenocopy the impaired repair responses to DSS. When induction of M2a-related mRNA and protein species were measured at Days 9 and 11, colons of DSS-treated TLR4-SNP mice exhibited reduced levels of M2a Mφ RNA and proteins, including PPARγ protein (Fig. 4A through C). These findings are consistent with previous studies showing that in contrast to WT Mφ TLR4−/− Mφ failed to induce Pparg mRNA in response to either IL-4 stimulation or RSV infection (135). Treatment of male and female, WT and TLR4-SNP thioglycollate-induced, peritoneal Mφ with exogenous IL-4 resulted in no significant difference in the levels of Arg1 and Chil3 mRNA, indicating that TLR4-SNP Mφ do not have an intrinsic defect in the ability to respond through IL-4Rα (data not shown).
PPARγ heterodimerizes with the NHR, RXR, to activate transcription of many M2a genes (e.g., Arg1, Mrc1, Chil3) (112, 130, 167), and transcriptional activity can be further modulated by endogenous and exogenous agonist ligands that bind to each component of the heterodimer (169). Many studies have investigated the therapeutic effect of ligand activation of the PPARs in IBD (141, 150, 170 – 173), including human clinical trials (174 – 176) and studies (177). TZDs are a class of structurally related PPARγ ligands originally designed as antidiabetic drugs. In particular, rosiglitazone acts as a highly specific ligand agonist of PPARγ (178, 179) that enhances M2a Mφ polarization (130, 180, 181), reduces DSS-induced inflammation in vivo (150), and leads to overall anti-inflammatory effects (182). Rosiglitazone showed promise in a clinical trial for treatment of active UC (175) but is no longer prescribed due to cardiotoxicity (183). Rosiglitazone therapy resolved lung histology and enhanced the expression of M2a Mφ in rodent models of RSV-induced lung injury (135). Another structurally related TZD, pioglitazone, showed similar outcomes in influenza infection (184). We hypothesized that therapeutic administration of rosiglitazone would also contribute to the resolution of DSS-induced colitis in the TLR4-SNP mice. Indeed, rosiglitazone effectively reversed DSS-induced colonic damage in the TLR4-SNP mice (Fig. 6), further supporting the conclusion that PPARγ may be an effective therapeutic target in patients that express the D299G and T399I TLR4 SNPs. Importantly, recent studies suggest that it is possible to develop PPARγ ligands that dissociate one cell-specific PPARγ function from another (185), offering the possibility of being able to identify a new PPARγ agonist ligand that will ameliorate IBD but not exert cardiotoxicity.
Since the microbiome has been strongly implicated in IBD pathogenesis, future studies will be required to determine whether the microbiomes of WT and TLR4-SNP mice differ and whether potential microbial differences affect the observed phenotypic responses to DSS in the two strains. Since β-HB is a metabolite associated with Gram-negative bacteria, it is possible that bacterial subsets may be reduced or missing in the DSS-treated TLR4-SNP mice such that β-HB is produced to a lesser extent, thereby reducing M2a Mφ differentiation.
In conclusion, TLR4-SNP mice develop more severe DSS-colitis symptoms compared to WT mice and exhibit a reduced capacity to repair colonic damage. IL-4Rα and myeloid PPARγ are necessary for induction of M2a Mφ and we show herein that IL-4Rα and PPARγ (on myeloid cells) are necessary to repair DSS-induced colonic damage. DSS-treated TLR4-SNP mice exhibited reduced M2a Mφ markers including PPARγ but responded to rosiglitazone, providing a potential avenue for future therapeutic development. In the context of personalized medicine, we anticipate that genotyping of IBD patients for the inheritance of TLR4 SNPs may ultimately lead to the identification of a patient subset for which M2a-inducing therapies are likely to be most effective.
MATERIALS AND METHODS
Mice
Six-week-old WT C57BL/6J and BALB/cByJ mice were purchased from the Jackson Laboratory (Bar Harbor, ME) and acclimated in our vivarium until use at 8–10 weeks of age. TLR4-SNP mice were engineered on a C57BL/6J background to express Tlr4 SNPs encoding D298G and N397I in murine TLR4 that are homologous to human TLR4 SNPs that encode D299G and T399I (14). IL-4Rα−/− mice (BALB/c background) were originally provided by Nancy Noben-Trauth (Rockville, MD) and William Paul (NIH, Bethesda, MD). PPARγcKO mice (C57BL/6 background) were provided by Dr. Mary Jane Thomassen (East Carolina University). Originally, homozygous floxed PPARγ mice were crossed into a transgenic mouse strain on a C57BL/6 background containing the Cre gene under control of the Lysozyme M (Lyz2) promoter (PPARγ flox+/+/Cre+/+) to delete the Pparg gene in lysozyme-producing cells (e.g., Mφ, neutrophils) (133, 147 – 149). All transgenic strains were bred homozygously at the University of Maryland, Baltimore (UMB). Animal experiments were conducted with institutional IACUC approvals from UMB. TLR4-SNP mice are available upon request.
DSS-induced colitis; measurements of colonic and histologic scores
Control mice received standard hyperchlorinated drinking water from Days 0 to 14. Experimental mice received 3% DSS (CAS 9011-18-1, MP Biomedicals 160110) in drinking water on Days 0–7 (changed on alternate days) (75, 77). Each new lot of DSS was titrated to ensure equivalent potency. On Day 8, all mice were provided regular drinking water only to allow a period of recovery through Day 14. A “colonic symptom score” was measured on alternate days by combining the stool consistency score (0 = normal; 1 = soft; 2 = very soft; 3 = diarrhea) and the fecal occult blood score [0 = no blood (−occult); 2 = microscopic blood in stool (+occult); 3 = macroscopic visible blood in stool (+occult); 4 = gross rectal bleeding] (75, 77). Fecal occult blood was measured using the Fisher Healthcare Sure-Vue Fecal Occult Blood Test (Fisher Scientific). At the indicated times, colons were resected, cleared of feces, and measured in centimeters distally from the cecum to just prior to the rectum. For most experiments, the most distal 1 cm colon section from control and experimental mice was opened longitudinally and fixed in 4% paraformaldehyde for 2 h at room temperature and sent to the University of Maryland School of Medicine’s Pathology Histology Core or to the University of Maryland School of Medicine’s and Greenebaum Comprehensive Cancer Center’s Pathology Biorepository Shared Services (housed in the Center for Innovative Biomedical Research (CIBR)) for sectioning and/or H&E-staining. At least two sections per mouse and >3 areas per section were analyzed using the ECHO Revolve microscope under 10× magnification for histology scoring or 100× magnification for high-power cellular counts. The total histology score represents the sum of epithelial damage (0–3), mucosal inflammatory infiltrate (0–3), submucosal inflammatory infiltrate (0–3), and muscularis externa inflammatory infiltrate (0–3), each multiplied by the respective extent of damage (1–3) (77). Under high power, the number of monocytic cells and the number of granulocytic cells were recorded in >6 high-power fields per mouse. In some experiments, the DC was flash frozen and subsequently homogenized in 0.5 or 1-mL PBS for Western analysis. The next most distal 1 cm of the colon (medial colon, MC) was flash frozen and used for RNA extraction for qRT-PCR.
Measurement of colonic permeability
On Days 6 or 13, control or DSS-treated mice were fasted overnight before administration of 0.6 mg/g FITC-dextran in PBS (CAS 60842-46-8, Sigma-Aldrich) by oral gavage on Days 7 and 14. Mice were bled 4 h after FITC-dextran administration. Fluorescence was measured in the serum of each mouse (diluted 1:2 in 1× PBS) and compared to a standard curve (0–8,000 ng/mL FITC-dextran) in a black well plate using a fluorescent spectrophotometer (Thermo Electron Corporation Fluoroskan Ascent FL) with 485 nm excitation and 527 nm emission (186).
Gene expression analysis
Flash-frozen colonic tissue taken from the MC was homogenized in TriPure (Roche) and RNA extracted using the manufacturer’s protocol. Since DSS has been reported to interfere with the reverse transcriptase reaction (187), RNA was purified using the LiCl purification method (188). cDNA was synthesized by reverse transcription reaction from 1 µg RNA per sample using the qScript cDNA synthesis kit (Quantabio). The 7500 Fast Real-Time PCR System and software (Applied Biosystems) was used to perform qRT-PCR (189). Forward and reverse primers included those corresponding to the housekeeping gene, Hprt, and M2a markers, Chil3, Arg1, Mrc1, and Pparg (Sigma) (Table S1). Primers were designed to cross intron-exon boundaries, and sequences were confirmed to target the gene of interest by NCBI BLAST. Sample amplification was verified using a negative control containing no cDNA. mRNA levels were normalized to relative Hprt mRNA levels and were reported using the −ΔCt method (14, 190).
Measurement of β-HB
Serum from each mouse was collected at the indicated times and filtered using 10 kDa centrifugal filter units (Millipore). β-HB was measured using the β-HB (ketone body) colorimetric assay kit (Cayman Chemical; 700190). β-HB standards were prepared according to the manufacturer’s protocol. Standards and samples (50 µL) were added to the plate in duplicate. Developer solution (50 µL) was added to each well, and the plate was incubated in the dark for 30 min at 25°C. Absorbance was measured at 450 nm using a ELx808 (BioTek) plate reader.
Western analysis
The DC was homogenized in 0.5 or 1 mL 1× PBS and centrifuged at 1,000 × g for 10 min at 4°C to remove cellular debris. The supernatants were aliquoted and frozen at −20°C for Western analysis (191). Primary anti-PPARγ rabbit polyclonal antibody (Abclonal A0270; 1:2,000 dilution), primary mouse monoclonal anti-Arginase 1 (BD Biosciences 610709; 1:1,000 dilution), primary rabbit polyclonal anti-Ym1 (StemCell 60130; 1:1,000 dilution), primary goat polyclonal anti-Mrc1 (R&D Systems AF2535-SP; 1:1,000 dilution), primary rabbit anti-GAPDH (14C10) monoclonal antibody (Cell Signaling 2118; 1:1,000 dilution), secondary peroxidase-conjugated goat anti-rabbit IgG antibody (Jackson ImmunoResearch 111-035-003; 1:5,000 dilution), secondary peroxidase-conjugated goat anti-mouse IgG light chain-specific antibody (Jackson ImmunoResearch 115-035-174; 1:5,000 dilution), and secondary peroxidase-conjugated rabbit anti-goat IgG antibody (Jackson ImmunoResearch 305-035-003; 1:5,000 dilution) were used to detect species-specific primary antibodies. Proteins were detected using Pierce ECL (or ECLPlus) Western Blotting Substrate (Thermo Scientific). Densitometry was performed on scanned images of the developed film using the open-source ImageJ software (NIH). Pixel intensity across the length of each lane was calculated to create a graph of intensity vs position. The software was used to calculate the area under these curves to provide a measure of total protein present in an individual band. These calculated protein measurements were normalized to the areas under the curves for the corresponding housekeeping protein GAPDH.
Isolation and cultivation of peritoneal Mφ
Thioglycollate-elicited Mφ (192) were treated for 24 h with medium or IL-4 (5 ng/mL) (184). Supernatants from lysates of the cells were analyzed by Western blot analysis.
Statistical analysis
Mean values ± standard error of the mean are shown. Comparisons among more than two groups were analyzed using a two-way ANOVA with Sidak’s multiple comparisons test (GraphPad Prism 9) unless otherwise noted. Comparisons among exactly two groups were analyzed using an unpaired two-tailed Student’s t-test (GraphPad Prism 9). *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. The number and sex of mice per treatment are indicated in the figure legends, and individual data points are presented from the indicated number of experiments. Biostatistical services were provided by CIBR (UMB).
ACKNOWLEDGMENTS
We thank Drs. David Rasko, Jacques Ravel, and Terez Shea-Donohue for their insightful discussions and advice during this study. We thank Dr. Soren Bentzen for guidance regarding statistical analyses of our data. The authors thank the University of Maryland Baltimore’s Pathology Histology Core and the Center for Innovative Biomedical Resources (CIBR) Pathology Biorepository Shared Service and the Biostatistics and Bioinformatics Shared Service of the University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center for their histological services and biostatistics guidance.
Author A.M.V. was supported through The American Association of Immunologists Careers in Immunology Fellowship Program (A.M.V./S.N.V). This work was also supported by NIH T32 AI095190 (S.N.V.), NIH R21 AI163543 (S.N.V.), NIH R01 AI143845 (A.D.K./S.N.V.), NIH R21 AI174374 (D.P./S.N.V.), UMGCCC American Cancer Society Institutional Research Grant (IRG-18-160-16) (M.S.B.), NIH R56DK131277, and UMB SOD INSPIRE Grant (V.T.M.). CIBR was supported by funds through the National Cancer Institute Cancer Center Support Grant (CCSG) P30CA134274.
All authors contributed to the experimental design of this study and the writing of this manuscript. Specifically, A.M.V. performed all experiments and analysis as part of her PhD requirements. D.P., K.A.S., M.S.B., and D.J.P. performed or provided guidance for all experiments and analysis. V.T.M. provided advice and support for microbiome-related studies. A.D.K. provided the IL-4Rα−/− mice and guidance on experiments focused on M2a Mφ differentiation and analysis. S.N.V. provided the TLR4-SNP and PPARγcKO mice, oversaw and directed all experiments, and provided guidance on Mφ and TLR4.
Contributor Information
Stefanie N. Vogel, Email: svogel@som.umaryland.edu.
Alejandro Aballay, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA .
DATA AVAILABILITY
All primary data are available from the corresponding author on reasonable request.
SUPPLEMENTAL MATERIAL
The following material is available online at https://doi.org/10.1128/mbio.01208-23.
Forward and reverse qRT-PCR primer sequences.
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.
REFERENCES
- 1. Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, Birdwell D, Alejos E, Silva M, Galanos C, Freudenberg M, Ricciardi-Castagnoli P, Layton B, Beutler B. 1998. Defective LPS signaling in C3H/Hej and C57Bl/10Sccr mice: mutations in Tlr4 gene. Science 282:2085–2088. doi: 10.1126/science.282.5396.2085 [DOI] [PubMed] [Google Scholar]
- 2. Qureshi ST, Larivière L, Leveque G, Clermont S, Moore KJ, Gros P, Malo D. 1999. Endotoxin-tolerant mice have mutations in toll-like receptor 4 (Tlr4). J Exp Med 189:615–625. doi: 10.1084/jem.189.4.615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Medvedev AE. 2013. Toll-like receptor polymorphisms, inflammatory and infectious diseases, allergies, and cancer. J Interferon Cytokine Res 33:467–484. doi: 10.1089/jir.2012.0140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Akira S, Takeda K. 2004. Toll-like receptor signalling. Nat Rev Immunol 4:499–511. doi: 10.1038/nri1391 [DOI] [PubMed] [Google Scholar]
- 5. Brubaker SW, Bonham KS, Zanoni I, Kagan JC. 2015. Innate immune pattern recognition: a cell biological perspective. Annu Rev Immunol 33:257–290. doi: 10.1146/annurev-immunol-032414-112240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Barton GM, Kagan JC. 2009. A cell biological view of toll-like receptor function: regulation through compartmentalization. Nat Rev Immunol 9:535–542. doi: 10.1038/nri2587 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Fitzgerald KA, Kagan JC. 2020. Toll-like receptors and the control of immunity. Cell 180:1044–1066. doi: 10.1016/j.cell.2020.02.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Arbour NC, Lorenz E, Schutte BC, Zabner J, Kline JN, Jones M, Frees K, Watt JL, Schwartz DA. 2000. TLR4 mutations are associated with endotoxin hyporesponsiveness in humans. Nat Genet 25:187–191. doi: 10.1038/76048 [DOI] [PubMed] [Google Scholar]
- 9. Lorenz E, Mira JP, Frees KL, Schwartz DA. 2002. Relevance of mutations in the TLR4 receptor in patients with gram-negative septic shock. Arch Intern Med 162:1028–1032. doi: 10.1001/archinte.162.9.1028 [DOI] [PubMed] [Google Scholar]
- 10. Wang H, Zhou S, Zhang J, Lei S, Zhou J. 2019. Correlations between TLR polymorphisms and inflammatory bowel disease: a meta-analysis of 49 case-control studies. Immunol Res 67:142–150. doi: 10.1007/s12026-018-9061-0 [DOI] [PubMed] [Google Scholar]
- 11. Tal G, Mandelberg A, Dalal I, Cesar K, Somekh E, Tal A, Oron A, Itskovich S, Ballin A, Houri S, Beigelman A, Lider O, Rechavi G, Amariglio N. 2004. Association between common toll-like receptor 4 mutations and severe respiratory syncytial virus disease. J Infect Dis 189:2057–2063. doi: 10.1086/420830 [DOI] [PubMed] [Google Scholar]
- 12. Puthothu B, Forster J, Heinzmann A, Krueger M. 2006. TLR-4 and CD14 polymorphisms in respiratory syncytial virus associated disease. Dis Markers 22:303–308. doi: 10.1155/2006/865890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Awomoyi AA, Rallabhandi P, Pollin TI, Lorenz E, Sztein MB, Boukhvalova MS, Hemming VG, Blanco JCG, Vogel SN. 2007. Association of TLR4 polymorphisms with symptomatic respiratory syncytial virus infection in high-risk infants and young children. J Immunol 179:3171–3177. doi: 10.4049/jimmunol.179.5.3171 [DOI] [PubMed] [Google Scholar]
- 14. Richard K, Piepenbrink KH, Shirey KA, Gopalakrishnan A, Nallar S, Prantner DJ, Perkins DJ, Lai W, Vlk A, Toshchakov VY, Feng C, Fanaroff R, Medvedev AE, Blanco JCG, Vogel SN. 2021. A mouse model of human TLR4 D299G/T399I SNPs reveals mechanisms of altered LPS and pathogen responses. J Exp Med 218:e20200675. doi: 10.1084/jem.20200675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Lorenz E, Schwartz DA, Martin PJ, Gooley T, Lin MT, Chien JW, Hansen JA, Clark JG. 2001. Association of TLR4 mutations and the risk for acute GVHD after HLA-matched-sibling hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 7:384–387. doi: 10.1053/bbmt.2001.v7.pm11529488 [DOI] [PubMed] [Google Scholar]
- 16. Lorenz E, Hallman M, Marttila R, Haataja R, Schwartz DA. 2002. Association between the Asp299Gly polymorphisms in the toll-like receptor 4 and premature births in the Finnish population. Pediatr Res 52:373–376. doi: 10.1203/00006450-200209000-00011 [DOI] [PubMed] [Google Scholar]
- 17. Carvalho A, Pasqualotto AC, Pitzurra L, Romani L, Denning DW, Rodrigues F. 2008. Polymorphisms in toll-like receptor genes and susceptibility to pulmonary aspergillosis. J Infect Dis 197:618–621. doi: 10.1086/526500 [DOI] [PubMed] [Google Scholar]
- 18. Chaiwiang N, Poyomtip T. 2019. The associations between toll-like receptor 4 gene polymorphisms and hepatitis C virus infection: a systematic review and meta-analysis. Biosci Rep 39:BSR20182470. doi: 10.1042/BSR20182470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Vidyant S, Chatterjee A, Dhole TN. 2019. A single-nucleotide polymorphism in TLR4 is linked with the risk of HIV-1 infection. Br J Biomed Sci 76:59–63. doi: 10.1080/09674845.2018.1559486 [DOI] [PubMed] [Google Scholar]
- 20. Ferwerda B, Kibiki GS, Netea MG, Dolmans WM, van der Ven AJ. 2007. The toll-like receptor 4 Asp299Gly variant and tuberculosis susceptibility in HIV-infected patients in Tanzania. AIDS 21:1375–1377. doi: 10.1097/QAD.0b013e32814e6b2d [DOI] [PubMed] [Google Scholar]
- 21. Pulido I, Leal M, Genebat M, Pacheco YM, Sáez ME, Soriano-Sarabia N. 2010. The TLR4 ASP299GLY polymorphism is a risk factor for active tuberculosis in Caucasian HIV-infected patients. Curr HIV Res 8:253–258. doi: 10.2174/157016210791111052 [DOI] [PubMed] [Google Scholar]
- 22. Lorenz E, Schwartz DA, Martin PJ, Gooley T, Lin MT, Chien JW, Hansen JA, Clark JG. 2001. Association of TLR4 mutations and the risk for acute GVHD after HLA-matched-sibling hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 7:384–387. doi: 10.1053/bbmt.2001.v7.pm11529488 [DOI] [PubMed] [Google Scholar]
- 23. Kiechl S, Lorenz E, Reindl M, Wiedermann CJ, Oberhollenzer F, Bonora E, Willeit J, Schwartz DA. 2002. Toll-like receptor 4 polymorphisms and atherogenesis. N Engl J Med 347:185–192. doi: 10.1056/NEJMoa012673 [DOI] [PubMed] [Google Scholar]
- 24. Ameziane N, Beillat T, Verpillat P, Chollet-Martin S, Aumont M-C, Seknadji P, Lamotte M, Lebret D, Ollivier V, de Prost D. 2003. Association of the toll-like receptor 4 gene Asp299Gly polymorphism with acute coronary events. ATVB 23. doi: 10.1161/01.ATV.0000101191.92392.1D [DOI] [PubMed] [Google Scholar]
- 25. Radstake TRDJ, Franke B, Hanssen S, Netea MG, Welsing P, Barrera P, Joosten LAB, van Riel PLCM, van den Berg WB. 2004. The toll-like receptor 4 Asp299Gly functional variant is associated with decreased rheumatoid arthritis disease susceptibility but does not influence disease severity and/or outcome. Arthritis Rheum 50:999–1001. doi: 10.1002/art.20114 [DOI] [PubMed] [Google Scholar]
- 26. Bochud PY, Sinsimer D, Aderem A, Siddiqui MR, Saunderson P, Britton S, Abraham I, Tadesse Argaw A, Janer M, Hawn TR, Kaplan G. 2009. Polymorphisms in toll-like receptor 4 (TLR4) are associated with protection against leprosy. Eur J Clin Microbiol Infect Dis 28:1055–1065. doi: 10.1007/s10096-009-0746-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Rallabhandi P, Bell J, Boukhvalova MS, Medvedev A, Lorenz E, Arditi M, Hemming VG, Blanco JCG, Segal DM, Vogel SN. 2006. Analysis of TLR4 polymorphic variants: new insights into TLR4/MD-2/CD14 stoichiometry, structure, and signaling. J Immunol 177:322–332. doi: 10.4049/jimmunol.177.1.322 [DOI] [PubMed] [Google Scholar]
- 28. Prohinar P, Rallabhandi P, Weiss JP, Gioannini TL. 2010. Expression of functional D299G.T399I polymorphic variant of TLR4 depends more on coexpression of MD-2 than does wild-type TLR4. J Immunol 184:4362–4367. doi: 10.4049/jimmunol.0903142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Figueroa L, Xiong Y, Song C, Piao W, Vogel SN, Medvedev AE. 2012. The Asp299Gly polymorphism alters TLR4 signaling by interfering with recruitment of MyD88 and TRIF. J Immunol 188:4506–4515. doi: 10.4049/jimmunol.1200202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Ohto U, Yamakawa N, Akashi-Takamura S, Miyake K, Shimizu T. 2012. Structural analyses of human toll-like receptor 4 polymorphisms D299G and T399I. J Biol Chem 287:40611–40617. doi: 10.1074/jbc.M112.404608 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Yamakawa N, Ohto U, Akashi-Takamura S, Takahashi K, Saitoh SI, Tanimura N, Suganami T, Ogawa Y, Shibata T, Shimizu T, Miyake K. 2013. Human TLR4 polymorphism D299G/T399I alters TLR4/MD-2 conformation and response to a weak ligand monophosphoryl lipid A. Int Immunol 25:45–52. doi: 10.1093/intimm/dxs084 [DOI] [PubMed] [Google Scholar]
- 32. Hajjar AM, Ernst RK, Yi J, Yam CS, Miller SI. 2017. Expression level of human TLR4 rather than sequence is the key determinant of LPS responsiveness. PLoS One 12:e0186308. doi: 10.1371/journal.pone.0186308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Ferwerda B, McCall MBB, Alonso S, Giamarellos-Bourboulis EJ, Mouktaroudi M, Izagirre N, Syafruddin D, Kibiki G, Cristea T, Hijmans A, Hamann L, Israel S, ElGhazali G, Troye-Blomberg M, Kumpf O, Maiga B, Dolo A, Doumbo O, Hermsen CC, Stalenhoef AFH, van Crevel R, Brunner HG, Oh D-Y, Schumann RR, de la Rúa C, Sauerwein R, Kullberg B-J, van der Ven AJAM, van der Meer JWM, Netea MG. 2007. TLR4 polymorphisms, infectious diseases, and evolutionary pressure during migration of modern humans. Proc Natl Acad Sci U S A 104:16645–16650. doi: 10.1073/pnas.0704828104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Loftus EV Jr. 2004. Clinical epidemiology of inflammatory bowel disease: incidence, prevalence, and environmental influences. Gastroenterology 126:1504–1517. doi: 10.1053/j.gastro.2004.01.063 [DOI] [PubMed] [Google Scholar]
- 35. Crohn BB, Ginzburg L, Oppenheimer GD. 1952. Regional ileitis; a pathologic and clinical entity. Am J Med 13:583–590. doi: 10.1016/0002-9343(52)90025-9 [DOI] [PubMed] [Google Scholar]
- 36. Flynn S, Eisenstein S. 2019. Inflammatory bowel disease presentation and diagnosis. Surg Clin North Am 99:1051–1062. doi: 10.1016/j.suc.2019.08.001 [DOI] [PubMed] [Google Scholar]
- 37. Greuter T, Manser C, Pittet V, Vavricka SR, Biedermann L, on behalf of Swiss IBDnet, an official working group of the swiss Society of Gastroenterology . 2020. Gender differences in inflammatory bowel disease. Digestion 101 Suppl 1:98–104. doi: 10.1159/000504701 [DOI] [PubMed] [Google Scholar]
- 38. Basso PJ, Fonseca MTC, Bonfá G, Alves VBF, Sales-Campos H, Nardini V, Cardoso CRB. 2014. Association among genetic predisposition, gut Microbiota, and host immune response in the etiopathogenesis of inflammatory bowel disease. Braz J Med Biol Res 47:727–737. doi: 10.1590/1414-431X20143932 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. 2014. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev 13:3–10. doi: 10.1016/j.autrev.2013.06.004 [DOI] [PubMed] [Google Scholar]
- 40. Na YR, Stakenborg M, Seok SH, Matteoli G. 2019. Macrophages in intestinal inflammation and resolution: a potential therapeutic target in IBD. Nat Rev Gastroenterol Hepatol 16:531–543. doi: 10.1038/s41575-019-0172-4 [DOI] [PubMed] [Google Scholar]
- 41. Sultzer BM. 1968. Genetic control of leucocyte responses to endotoxin. Nature 219:1253–1254. doi: 10.1038/2191253a0 [DOI] [PubMed] [Google Scholar]
- 42. Vogel SN, Hansen CT, Rosenstreich DL. 1979. Characterization of a congenitally LPS-resistant, athymic mouse strain. J Immunol 122:619–622. [PubMed] [Google Scholar]
- 43. Mähler M, Bristol IJ, Leiter EH, Workman AE, Birkenmeier EH, Elson CO, Sundberg JP. 1998. Differential susceptibility of inbred Mouse strains to dextran sulfate sodium-induced colitis. Am J Physiol 274:G544–51. doi: 10.1152/ajpgi.1998.274.3.G544 [DOI] [PubMed] [Google Scholar]
- 44. Lange S, Delbro DS, Jennische E, Mattsby-Baltzer I. 1996. The role of the Lps gene in experimental ulcerative colitis in mice. APMIS 104:823–833. doi: 10.1111/j.1699-0463.1996.tb04948.x [DOI] [PubMed] [Google Scholar]
- 45. Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, Takeda K, Akira S. 1999. Cutting edge: toll-like receptor 4 (TLR4)-Deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J Immunol 162:3749–3752. [PubMed] [Google Scholar]
- 46. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. 2004. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118:229–241. doi: 10.1016/j.cell.2004.07.002 [DOI] [PubMed] [Google Scholar]
- 47. Fukata M, Michelsen KS, Eri R, Thomas LS, Hu B, Lukasek K, Nast CC, Lechago J, Xu R, Naiki Y, Soliman A, Arditi M, Abreu MT. 2005. Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis. Am J Physiol Gastrointest Liver Physiol 288:G1055–65. doi: 10.1152/ajpgi.00328.2004 [DOI] [PubMed] [Google Scholar]
- 48. Abreu MT, Arnold ET, Thomas LS, Gonsky R, Zhou Y, Hu B, Arditi M. 2002. TLR4 and MD-2 expression is regulated by immune-mediated signals in human intestinal epithelial cells. J Biol Chem 277:20431–20437. doi: 10.1074/jbc.M110333200 [DOI] [PubMed] [Google Scholar]
- 49. Hausmann M, Kiessling S, Mestermann S, Webb G, Spöttl T, Andus T, Schölmerich J, Herfarth H, Ray K, Falk W, Rogler G. 2002. Toll-like receptors 2 and 4 are up-regulated during intestinal inflammation. Gastroenterology 122:1987–2000. doi: 10.1053/gast.2002.33662 [DOI] [PubMed] [Google Scholar]
- 50. Frosali S, Pagliari D, Gambassi G, Landolfi R, Pandolfi F, Cianci R. 2015. How the intricate interaction among toll-like receptors, microbiota, and intestinal immunity can influence gastrointestinal pathology. J Immunol Res 2015:489821. doi: 10.1155/2015/489821 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Strober W, Fuss I, Mannon P. 2007. The fundamental basis of inflammatory bowel disease. J Clin Invest 117:514–521. doi: 10.1172/JCI30587 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Lakatos PL, Fischer S, Lakatos L, Gal I, Papp J. 2006. Current concept on the pathogenesis of inflammatory bowel disease-crosstalk between genetic and microbial factors: pathogenic bacteria and altered bacterial sensing or changes in mucosal integrity take “toll World J Gastroenterol 12:1829–1841. doi: 10.3748/wjg.v12.i12.1829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Khor B, Gardet A, Xavier RJ. 2011. Genetics and pathogenesis of inflammatory bowel disease. Nature 474:307–317. doi: 10.1038/nature10209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Berg DJ, Davidson N, Kühn R, Müller W, Menon S, Holland G, Thompson-Snipes L, Leach MW, Rennick D. 1996. Enterocolitis and colon cancer in Interleukin-10-deficient mice are associated with aberrant cytokine production and CD4(+) TH1-like responses. J Clin Invest 98:1010–1020. doi: 10.1172/JCI118861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Arseneau KO, Tamagawa H, Pizarro TT, Cominelli F. 2007. Innate and adaptive immune responses related to IBD pathogenesis. Curr Gastroenterol Rep 9:508–512. doi: 10.1007/s11894-007-0067-3 [DOI] [PubMed] [Google Scholar]
- 56. Wang A-J, Smith A, Li Y, Urban JF, Ramalingam TR, Wynn TA, Lu N, Shea-Donohue T, Yang Z, Zhao A. 2014. Genetic deletion of IL-25 (IL-17E) confers resistance to dextran sulfate sodium-induced colitis in mice. Cell Biosci 4:72. doi: 10.1186/2045-3701-4-72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Xavier RJ, Podolsky DK. 2007. Unravelling the pathogenesis of inflammatory bowel disease. Nature 448:427–434. doi: 10.1038/nature06005 [DOI] [PubMed] [Google Scholar]
- 58. Corr SC, O’Neill LAJ. 2009. Genetic variation in toll-like receptor signalling and the risk of inflammatory and immune diseases. J Innate Immun 1:350–357. doi: 10.1159/000200774 [DOI] [PubMed] [Google Scholar]
- 59. Kordjazy N, Haj-Mirzaian A, Haj-Mirzaian A, Rohani MM, Gelfand EW, Rezaei N, Abdolghaffari AH. 2018. Role of toll-like receptors in inflammatory bowel disease. Pharmacol Res 129:204–215. doi: 10.1016/j.phrs.2017.11.017 [DOI] [PubMed] [Google Scholar]
- 60. Franchimont D, Vermeire S, El Housni H, Pierik M, Van Steen K, Gustot T, Quertinmont E, Abramowicz M, Van Gossum A, Devière J, Rutgeerts P. 2004. Deficient host-bacteria interactions in inflammatory bowel disease? the toll-like receptor (TLR)-4 Asp299Gly polymorphism is associated with Crohn’s disease and ulcerative colitis. Gut 53:987–992. doi: 10.1136/gut.2003.030205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Török H-P, Glas J, Tonenchi L, Mussack T, Folwaczny C. 2004. Polymorphisms of the lipopolysaccharide-signaling complex in inflammatory bowel disease: association of a mutation in the toll-like receptor 4 gene with ulcerative colitis. Clin Immunol 112:85–91. doi: 10.1016/j.clim.2004.03.002 [DOI] [PubMed] [Google Scholar]
- 62. Braat H, Stokkers P, Hommes T, Cohn D, Vogels E, Pronk I, Spek A, van Kampen A, van Deventer S, Peppelenbosch M, Hommes D. 2005. Consequence of functional Nod2 and Tlr4 mutations on gene transcription in Crohn's disease patients. J Mol Med (Berl) 83:601–609. doi: 10.1007/s00109-005-0685-x [DOI] [PubMed] [Google Scholar]
- 63. Brand S, Staudinger T, Schnitzler F, Pfennig S, Hofbauer K, Dambacher J, Seiderer J, Tillack C, Konrad A, Crispin A, Göke B, Lohse P, Ochsenkühn T. 2005. The role of Toll-like receptor 4 Asp299Gly and Thr399Ile polymorphisms and CARD15/NOD2 mutations in the susceptibility and phenotype of Crohn’s disease. Inflamm Bowel Dis 11:645–652. doi: 10.1097/01.mib.0000168372.94907.d2 [DOI] [PubMed] [Google Scholar]
- 64. Gazouli M, Mantzaris G, Kotsinas A, Zacharatos P, Papalambros E, Archimandritis A, Ikonomopoulos J, Gorgoulis V-G. 2005. Association between polymorphisms in the toll-like receptor 4, CD14, and CARD15/NOD2 and inflammatory bowel disease in the Greek population. World J Gastroenterol 11:681–685. doi: 10.3748/wjg.v11.i5.681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Browning BL, Huebner C, Petermann I, Gearry RB, Barclay ML, Shelling AN, Ferguson LR. 2007. Has toll-like receptor 4 been prematurely dismissed as an inflammatory bowel disease gene? association study combined with meta-analysis shows strong evidence for association. Am J Gastroenterol 102:2504–2512. doi: 10.1111/j.1572-0241.2007.01463.x [DOI] [PubMed] [Google Scholar]
- 66. De Jager PL, Franchimont D, Waliszewska A, Bitton A, Cohen A, Langelier D, Belaiche J, Vermeire S, Farwell L, Goris A, Libioulle C, Jani N, Dassopoulos T, Bromfield GP, Dubois B, Cho JH, Brant SR, Duerr RH, Yang H, Rotter JI, Silverberg MS, Steinhart AH, Daly MJ, Podolsky DK, Louis E, Hafler DA, Rioux JD, Quebec IBD Genetics Consortium and NIDDK IBD Genetics Consortium . 2007. The role of the toll receptor pathway in susceptibility to inflammatory bowel diseases. Genes Immun 8:387–397. doi: 10.1038/sj.gene.6364398 [DOI] [PubMed] [Google Scholar]
- 67. Hong J, Leung E, Fraser AG, Merriman TR, Vishnu P, Krissansen GW. 2007. TLR2, TLR4 and TLR9 polymorphisms and Crohn’s disease in a New Zealand caucasian cohort. J Gastroenterol Hepatol 22:1760–1766. doi: 10.1111/j.1440-1746.2006.04727.x [DOI] [PubMed] [Google Scholar]
- 68. Hume GE, Fowler EV, Doecke J, Simms LA, Huang N, Palmieri O, Griffiths LR, Florin THJ, Annese V, Radford-Smith GL. 2008. Novel NOD2 haplotype strengthens the association between TLR4 Asp299Gly and Crohn’s disease in an Australian population. Inflamm Bowel Dis 14:585–590. doi: 10.1002/ibd.20362 [DOI] [PubMed] [Google Scholar]
- 69. Sivaram G, Tiwari SK, Bardia A, Anjum F, Vishnupriya S, Habeeb A, Khan AA. 2012. Macrophage migration inhibitory factor, toll-like receptor 4, and CD14 polymorphisms with altered expression levels in patients with ulcerative colitis. Hum Immunol 73:201–205. doi: 10.1016/j.humimm.2011.12.006 [DOI] [PubMed] [Google Scholar]
- 70. Feki S, Bouzid D, Abida O, Chtourou L, Elloumi N, Toumi A, Hachicha H, Amouri A, Tahri N, Masmoudi H. 2017. Genetic Association and Phenotypic correlation of TLR4 but not NOD2 variants with Tunisian inflammatory bowel disease. J Dig Dis 18:625–633. doi: 10.1111/1751-2980.12552 [DOI] [PubMed] [Google Scholar]
- 71. Arnott IDR, Landers CJ, Nimmo EJ, Drummond HE, Smith BKR, Targan SR, Satsangi J. 2004. Sero-reactivity to microbial components in Crohn's disease is associated with disease severity and progression, but not Nod2/Card15 genotype. Am J Gastroenterol 99:2376–2384. doi: 10.1111/j.1572-0241.2004.40417.x [DOI] [PubMed] [Google Scholar]
- 72. Oostenbrug LE, Drenth JPH, de Jong DJ, Nolte IM, Oosterom E, van Dullemen HM, van der Linde K, te Meerman GJ, van der Steege G, Kleibeuker JH, Jansen PLM. 2005. Association between toll-like receptor 4 and inflammatory bowel disease. Inflamm Bowel Dis 11:567–575. doi: 10.1097/01.mib.0000161305.81198.0f [DOI] [PubMed] [Google Scholar]
- 73. Figueroa C, Peralta A, Herrera L, Castro P, Gutiérrez A, Valenzuela J, Aguillón JC, Quera R, Hermoso MA. 2006. NOD2/CARD15 and toll-like 4 receptor gene polymorphism in Chilean patients with inflammatory bowel disease. Eur Cytokine Netw 17:125–130. [PubMed] [Google Scholar]
- 74. Shea-Donohue T, Thomas K, Cody MJ, Detolla LJ, Kopydlowski KM, Fukata M, Lira SA, Vogel SN. 2008. Mice deficient in the CXCR2 ligand, CXCL1 (KC/GRO-α), exhibit increased susceptibility to dextran sodium sulfate (DSS)-Induced colitis. Innate Immun 14:117–124. doi: 10.1177/1753425908088724 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Kim JJ, Shajib MS, Manocha MM, Khan WI. 2012. Investigating intestinal inflammation in DSS-induced model of IBD. J Vis Exp 1–6:3678. doi: 10.3791/3678 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Netzel-Arnett S, Buzza MS, Shea-Donohue T, Désilets A, Leduc R, Fasano A, Bugge TH, Antalis TM. 2012. Matriptase protects against experimental colitis and promotes intestinal barrier recovery. Inflamm Bowel Dis 18:1303–1314. doi: 10.1002/ibd.21930 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Chassaing B, Aitken JD, Malleshappa M, Vijay-Kumar M. 2014. Dextran sulfate sodium (DSS)-Induced colitis in mice. Curr Protoc Immunol 104:15. doi: 10.1002/0471142735.im1525s104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Buzza MS, Johnson TA, Conway GD, Martin EW, Mukhopadhyay S, Shea-Donohue T, Antalis TM. 2017. Inflammatory cytokines down-regulate the barrier-protective prostasin-matriptase proteolytic cascade early in experimental colitis. J Biol Chem 292:10801–10812. doi: 10.1074/jbc.M116.771469 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Ukabam SO, Clamp JR, Cooper BT. 1983. Abnormal small intestinal permeability to sugars in patients with Crohn’s disease of the terminal ileum and colon. Digestion 27:70–74. doi: 10.1159/000198932 [DOI] [PubMed] [Google Scholar]
- 80. Hollander D, Vadheim CM, Brettholz E, Petersen GM, Delahunty T, Rotter JI. 1986. Increased intestinal permeability in patients with Crohn’s disease and their relatives. a possible etiologic factor. Ann Intern Med 105:883. doi: 10.7326/0003-4819-105-6-883 [DOI] [PubMed] [Google Scholar]
- 81. Andre F, Andre C, Emery Y, Forichon J, Descos L, Minaire Y. 1988. Assessment of the lactulose-mannitol test in Crohn’s disease. Gut 29:511–515. doi: 10.1136/gut.29.4.511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Poritz LS, Garver KI, Green C, Fitzpatrick L, Ruggiero F, Koltun WA. 2007. Loss of the tight junction protein ZO-1 in dextran sulfate sodium induced colitis. J Surg Res 140:12–19. doi: 10.1016/j.jss.2006.07.050 [DOI] [PubMed] [Google Scholar]
- 83. Arrieta MC, Madsen K, Doyle J, Meddings J. 2009. Reducing small intestinal permeability attenuates colitis in the Il10 gene-deficient Mouse. Gut 58:41–48. doi: 10.1136/gut.2008.150888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Mennigen R, Nolte K, Rijcken E, Utech M, Loeffler B, Senninger N, Bruewer M. 2009. Probiotic mixture VSL#3 protects the epithelial barrier by maintaining tight junction protein expression and preventing apoptosis in a murine model of colitis. Am J Physiol Gastrointest Liver Physiol 296:G1140–9. doi: 10.1152/ajpgi.90534.2008 [DOI] [PubMed] [Google Scholar]
- 85. Tripathi A, Lammers KM, Goldblum S, Shea-Donohue T, Netzel-Arnett S, Buzza MS, Antalis TM, Vogel SN, Zhao A, Yang S, Arrietta M-C, Meddings JB, Fasano A. 2009. Identification of human zonulin, a physiological modulator of tight junctions, as prehaptoglobin-2. Proc Natl Acad Sci U S A 106:16799–16804. doi: 10.1073/pnas.0906773106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Guo S, Nighot M, Al-Sadi R, Alhmoud T, Nighot P, Ma TY. 2015. Lipopolysaccharide regulation of intestinal tight junction permeability is mediated by TLR4 signal transduction pathway activation of FAK and Myd88. J Immunol 195:4999–5010. doi: 10.4049/jimmunol.1402598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Hans W, Schölmerich J, Gross V, Falk W. 2000. The role of the resident intestinal flora in acute and chronic dextran sulfate sodium-induced colitis in mice. Eur J Gastroenterol Hepatol 12:267–273. doi: 10.1097/00042737-200012030-00002 [DOI] [PubMed] [Google Scholar]
- 88. Glassner KL, Abraham BP, Quigley EMM. 2020. The microbiome and inflammatory bowel disease. J Allergy Clin Immunol 145:16–27. doi: 10.1016/j.jaci.2019.11.003 [DOI] [PubMed] [Google Scholar]
- 89. Lee Y, Sugihara K, Gillilland MG, Jon S, Kamada N, Moon JJ. 2020. Hyaluronic acid–bilirubin nanomedicine for targeted modulation of dysregulated intestinal barrier, microbiome and immune responses in colitis. Nat Mater 19:118–126. doi: 10.1038/s41563-019-0462-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Yang Y, Gharaibeh RZ, Newsome RC, Jobin C. 2020. Amending microbiota by targeting intestinal inflammation with TNF blockade attenuates development of colorectal cancer. Nat Cancer 1:723–734. doi: 10.1038/s43018-020-0078-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Zou J, Liu C, Jiang S, Qian D, Duan J, Richardson AR. 2021. Cross talk between gut microbiota and intestinal mucosal immunity in the development of ulcerative colitis. Infect Immun 89. doi: 10.1128/IAI.00014-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Hu J, Cheng S, Yao J, Lin X, Li Y, Wang W, Weng J, Zou Y, Zhu L, Zhi M. 2022. Correlation between altered gut microbiota and elevated inflammation markers in patients with Crohn’s disease. Front Immunol 13:947313. doi: 10.3389/fimmu.2022.947313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. McCoy KD, Geuking MB, Ronchi F. 2017. Gut microbiome standardization in control and experimental mice. Curr Protoc Immunol 117:23. doi: 10.1002/cpim.25 [DOI] [PubMed] [Google Scholar]
- 94. Lipinski JH, Zhou X, Gurczynski SJ, Erb-Downward JR, Dickson RP, Huffnagle GB, Moore BB, O’Dwyer DN, Raffatellu M. 2021. Cage environment regulates gut microbiota independent of toll-like receptors. Infect Immun 89. doi: 10.1128/IAI.00187-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Lin Y, Yang X, Yue W, Xu X, Li B, Zou L, He R. 2014. Chemerin aggravates DSS-induced colitis by suppressing M2 macrophage polarization. Cell Mol Immunol 11:355–366. doi: 10.1038/cmi.2014.15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. You Y, Zhou C, Li D, Cao Z-L, Shen W, Li W-Z, Zhang S, Hu B, Shen X. 2016. Sorting nexin 10 acting as a novel regulator of macrophage polarization mediates inflammatory response in experimental mouse colitis. Sci Rep 6:20630. doi: 10.1038/srep20630 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Yang R, Liao Y, Wang L, He P, Hu Y, Yuan D, Wu Z, Sun X. 2019. Exosomes derived from M2B macrophages attenuate DSS-induced colitis. Front Immunol 10:2346. doi: 10.3389/fimmu.2019.02346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Jang SE, Min SW. 2020. Amelioration of colitis in mice by Leuconostoc lactis EJ-1 by M1 to M2 macrophage polarization. Microbiol Immunol 64:133–142. doi: 10.1111/1348-0421.12752 [DOI] [PubMed] [Google Scholar]
- 99. Seyedizade SS, Afshari K, Bayat S, Rahmani F, Momtaz S, Rezaei N, Abdolghaffari AH. 2020. Current status of M1 and M2 macrophages pathway as drug targets for inflammatory bowel disease. Arch Immunol Ther Exp (Warsz) 68:10. doi: 10.1007/s00005-020-00576-4 [DOI] [PubMed] [Google Scholar]
- 100. Wang Z, Hao C, Zhuang Q, Zhan B, Sun X, Huang J, Cheng Y, Zhu X. 2020. Excretory/secretory products from trichinella spiralis adult Worms attenuated DSS-induced colitis in mice by driving PD-1-mediated M2 macrophage polarization. Front Immunol 11:563784. doi: 10.3389/fimmu.2020.563784 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Deng F, Yan J, Lu J, Luo M, Xia P, Liu S, Wang X, Zhi F, Liu D. 2021. M2 macrophage-derived exosomal miR-590-3p attenuates DSS-induced mucosal damage and promotes epithelial repair via the LATS1/YAP/β-Catenin signalling axis. J Crohns Colitis 15:665–677. doi: 10.1093/ecco-jcc/jjaa214 [DOI] [PubMed] [Google Scholar]
- 102. Kang Z-P, Wang M-X, Wu T-T, Liu D-Y, Wang H-Y, Long J, Zhao H-M, Zhong Y-B. 2021. Curcumin alleviated dextran sulfate sodium-induced colitis by regulating M1/M2 macrophage polarization and TLRs signaling pathway. Evid Based Complement Alternat Med 2021:3334994. doi: 10.1155/2021/3334994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Huang C, Wang J, Liu H, Huang R, Yan X, Song M, Tan G, Zhi F. 2022. Ketone body β-hydroxybutyrate ameliorates colitis by promoting M2 macrophage polarization through the STAT6-dependent signaling pathway. BMC Med 20:148. doi: 10.1186/s12916-022-02352-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Long J, Liu X-K, Kang Z-P, Wang M-X, Zhao H-M, Huang J-Q, Xiao Q-P, Liu D-Y, Zhong Y-B. 2022. Ginsenoside Rg1 ameliorated experimental colitis by regulating the balance of M1/M2 macrophage polarization and the homeostasis of intestinal flora. Eur J Pharmacol 917:174742. doi: 10.1016/j.ejphar.2022.174742 [DOI] [PubMed] [Google Scholar]
- 105. Meng Z, Fu B, Yang Z, Xu Y, Huang H, Bai Y, Fang X, Shen S, Yang J, Yong J, Tao X, Ma C, Miao W, Ren H, Wang Y, Xu H, Li X. 2023. Polydopamine-coated thalidomide nanocrystals promote DSS-induced murine colitis recovery through macrophage M2 polarization together with the synergistic anti-inflammatory and anti-angiogenic effects. Int J Pharm 630:122376. doi: 10.1016/j.ijpharm.2022.122376 [DOI] [PubMed] [Google Scholar]
- 106. Ma S, Zhang J, Liu H, Li S, Wang Q. 2022. The role of tissue-resident macrophages in the development and treatment of inflammatory bowel disease. Front Cell Dev Biol 10:896591. doi: 10.3389/fcell.2022.896591 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Wang J, Chen W-D, Wang Y-D. 2020. The relationship between gut microbiota and inflammatory diseases: the role of macrophages. Front Microbiol 11:1065. doi: 10.3389/fmicb.2020.01065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Cao L, Xu H, Wang G, Liu M, Tian D, Yuan Z. 2019. Extracellular vesicles derived from bone marrow mesenchymal stem cells attenuate dextran sodium sulfate-induced ulcerative colitis by promoting M2 macrophage polarization. Int Immunopharmacol 72:264–274. doi: 10.1016/j.intimp.2019.04.020 [DOI] [PubMed] [Google Scholar]
- 109. Cosín-Roger J, Ortiz-Masiá D, Calatayud S, Hernández C, Esplugues JV, Barrachina MD. 2016. The activation of WNT signaling by a STAT6-dependent macrophage phenotype promotes mucosal repair in murine IBD. Mucosal Immunol 9:986–998. doi: 10.1038/mi.2015.123 [DOI] [PubMed] [Google Scholar]
- 110. Hunter MM, Wang A, Parhar KS, Johnston MJG, Van Rooijen N, Beck PL, McKay DM. 2010. In vitro-derived alternatively activated macrophages reduce colonic inflammation in mice. Gastroenterology 138:1395–1405. doi: 10.1053/j.gastro.2009.12.041 [DOI] [PubMed] [Google Scholar]
- 111. Martinez FO, Gordon S. 2014. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 6:13. doi: 10.12703/P6-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Muraille E, Leo O, Moser M. 2014. TH1/TH2 paradigm extended: macrophage polarization as an unappreciated pathogen-driven escape mechanism. Front Immunol 5:603. doi: 10.3389/fimmu.2014.00603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, Locati M, Mantovani A, Martinez FO, Mege J-L, Mosser DM, Natoli G, Saeij JP, Schultze JL, Shirey KA, Sica A, Suttles J, Udalova I, van Ginderachter JA, Vogel SN, Wynn TA. 2014. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41:14–20. doi: 10.1016/j.immuni.2014.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Fleming BD, Chandrasekaran P, Dillon LAL, Dalby E, Suresh R, Sarkar A, El-Sayed NM, Mosser DM. 2015. The generation of macrophages with anti-inflammatory activity in the absence of STAT6 signaling. J Leukoc Biol 98:395–407. doi: 10.1189/jlb.2A1114-560R [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Heller NM, Qi X, Gesbert F, Keegan AD. 2012. The extracellular and transmembrane domains of the γC and interleukin (IL)-13 receptor Α1 chains, not their cytoplasmic domains, dictate the nature of signaling responses to IL-4 and IL-13. J Biol Chem 287:31948–31961. doi: 10.1074/jbc.M112.348896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Keegan AD, Leonard WJ, Zhu J. 2021. Recent advances in understanding the role of IL-4 signaling. Fac Rev 10:71. doi: 10.12703/r/10-71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Noben-Trauth N, Shultz LD, Brombacher F, Urban JF, Gu H, Paul WE. 1997. An interleukin 4 (IL-4)-independent pathway for CD4+ T cell IL-4 production is revealed in IL-4 receptor-deficient mice. Proc Natl Acad Sci U S A 94:10838–10843. doi: 10.1073/pnas.94.20.10838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Anderson AJ, Dawes EA. 1990. Occurrence, metabolism, metabolic role, and industrial uses of bacterial polyhydroxyalkanoates. Microbiol Rev 54:450–472. doi: 10.1128/mr.54.4.450-472.1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Sasaki K, Sasaki D, Hannya A, Tsubota J, Kondo A. 2020. In vitro human colonic microbiota Utilises D-β-hydroxybutyrate to increase butyrogenesis. Sci Rep 10:8516. doi: 10.1038/s41598-020-65561-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Mierziak J, Burgberger M, Wojtasik W. 2021. 3-Hydroxybutyrate as a metabolite and a signal molecule regulating processes of living organisms. Biomolecules 11:1–21. doi: 10.3390/biom11030402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Forsyth WGC, Hayward AC, Roberts JB. 1958. Occurrence of poly-Β-hydroxybutyric acid in aerobic gram-negative bacteria. Nature 182:800–801. doi: 10.1038/182800a0 [DOI] [PubMed] [Google Scholar]
- 122. Griebel R, Smith Z, Merrick JM. 1968. Metabolism of poly-beta-hydroxybutyrate. I. purification, composition, and properties of native poly-beta-hydroxybutyrate granules from Bacillus Megaterium. Biochemistry 7:3676–3681. doi: 10.1021/bi00850a047 [DOI] [PubMed] [Google Scholar]
- 123. Peoples OP, Sinskey AJ. 1989. Poly-beta-hydroxybutyrate (PHB) biosynthesis in Alcaligenes eutrophus H16. identification and characterization of the PHB polymerase gene (phbC). J Biol Chem 264:15298–15303. [PubMed] [Google Scholar]
- 124. Sudesh K, Abe H, Doi Y. 2000. Synthesis, structure and properties of polyhydroxyalkanoates: biological polyesters. Prog Polym Sci 25:1503–1555. doi: 10.1016/S0079-6700(00)00035-6 [DOI] [Google Scholar]
- 125. Dawiskiba T, Deja S, Mulak A, Ząbek A, Jawień E, Pawełka D, Banasik M, Mastalerz-Migas A, Balcerzak W, Kaliszewski K, Skóra J, Barć P, Korta K, Pormańczuk K, Szyber P, Litarski A, Młynarz P. 2014. Serum and urine metabolomic fingerprinting in diagnostics of inflammatory bowel diseases. World J Gastroenterol 20:163–174. doi: 10.3748/wjg.v20.i1.163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Zhang Y, Lin L, Xu Y, Lin Y, Jin Y, Zheng C. 2013. 1H NMR-based spectroscopy detects metabolic alterations in serum of patients with early-stage ulcerative colitis. Biochem Biophys Res Commun 433:547–551. doi: 10.1016/j.bbrc.2013.03.012 [DOI] [PubMed] [Google Scholar]
- 127. Yan X, Liu XY, Zhang D, Zhang YD, Li ZH, Liu X, Wu F, Chen GQ. 2021. Construction of a sustainable 3-hydroxybutyrate-producing probiotic Escherichia coli for treatment of colitis. Cell Mol Immunol 18:2344–2357. doi: 10.1038/s41423-021-00760-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Bouhlel MA, Derudas B, Rigamonti E, Dièvart R, Brozek J, Haulon S, Zawadzki C, Jude B, Torpier G, Marx N, Staels B, Chinetti-Gbaguidi G. 2007. PPARγ activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab 6:137–143. doi: 10.1016/j.cmet.2007.06.010 [DOI] [PubMed] [Google Scholar]
- 129. Coste A, Dubourdeau M, Linas MD, Cassaing S, Lepert J-C, Balard P, Chalmeton S, Bernad J, Orfila C, Séguéla J-P, Pipy B. 2003. PPARgamma promotes mannose receptor gene expression in murine macrophages and contributes to the induction of this receptor by IL-13. Immunity 19:329–339. doi: 10.1016/s1074-7613(03)00229-2 [DOI] [PubMed] [Google Scholar]
- 130. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F, Ferrante AW, Chawla A. 2007. Macrophage-specific PPARγ controls alternative activation and improves insulin resistance. Nature 447:1116–1120. doi: 10.1038/nature05894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Bonfield TL, Thomassen MJ, Farver CF, Abraham S, Koloze MT, Zhang X, Mosser DM, Culver DA. 2008. Peroxisome proliferator-activated receptor-gamma regulates the expression of alveolar macrophage macrophage colony-stimulating factor. J Immunol 181:235–242. doi: 10.4049/jimmunol.181.1.235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Stienstra R, Duval C, Keshtkar S, van der Laak J, Kersten S, Müller M. 2008. Peroxisome proliferator-activated receptor gamma activation promotes infiltration of alternatively activated macrophages into adipose tissue. J Biol Chem 283:22620–22627. doi: 10.1074/jbc.M710314200 [DOI] [PubMed] [Google Scholar]
- 133. Malur A, Mccoy AJ, Arce S, Barna BP, Kavuru MS, Malur AG, Thomassen MJ. 2009. Deletion of PPAR gamma in alveolar macrophages is associated with a Th-1 pulmonary inflammatory response. J Immunol 182:5816–5822. doi: 10.4049/jimmunol.0803504 [DOI] [PubMed] [Google Scholar]
- 134. Chawla A. 2010. Control of macrophage activation and function by PPARs. Circ Res 106:1559–1569. doi: 10.1161/CIRCRESAHA.110.216523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Shirey KA, Pletneva LM, Puche AC, Keegan AD, Prince GA, Blanco JCG, Vogel SN. 2010. Control of RSV-induced lung injury by alternatively activated macrophages is IL-4Rα-, TLR4-, and IFN-Β-dependent. Mucosal Immunol 3:291–300. doi: 10.1038/mi.2010.6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Date D, Das R, Narla G, Simon DI, Jain MK, Mahabeleshwar GH. 2014. Kruppel-like transcription factor 6 regulates inflammatory macrophage polarization. J Biol Chem 289:10318–10329. doi: 10.1074/jbc.M113.526749 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Abdalla HB, Napimoga MH, Lopes AH, de Macedo Maganin AG, Cunha TM, Van Dyke TE, Clemente Napimoga JT. 2020. Activation of PPAR-Γ induces macrophage polarization and reduces neutrophil migration mediated by heme oxygenase 1. Int Immunopharmacol 84:106565. doi: 10.1016/j.intimp.2020.106565 [DOI] [PubMed] [Google Scholar]
- 138. Kim CE, Park HY, Won HJ, Kim M, Kwon B, Lee S-J, Kim DH, Shin J-G, Seo S-K. 2021. Repression of PPARγ reduces the ABCG2-mediated efflux activity of M2 macrophages. Int J Biochem Cell Biol 130:105895. doi: 10.1016/j.biocel.2020.105895 [DOI] [PubMed] [Google Scholar]
- 139. Desreumaux P, Dubuquoy L, Nutten S, Peuchmaur M, Englaro W, Schoonjans K, Derijard B, Desvergne B, Wahli W, Chambon P, Leibowitz MD, Colombel JF, Auwerx J. 2001. Attenuation of colon inflammation through Activators of the Retinoid X receptor (RXR)/Peroxisome Proliferator-activated receptor gamma (PPARgamma) heterodimer. a basis for new therapeutic strategies. J Exp Med 193:827–838. doi: 10.1084/jem.193.7.827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Glass CK, Ogawa S. 2006. Combinatorial roles of nuclear receptors in inflammation and immunity. Nat Rev Immunol 6:44–55. doi: 10.1038/nri1748 [DOI] [PubMed] [Google Scholar]
- 141. Dubuquoy L, Rousseaux C, Thuru X, Peyrin-Biroulet L, Romano O, Chavatte P, Chamaillard M, Desreumaux P. 2006. PPARgamma as a new therapeutic target in inflammatory bowel diseases. Gut 55:1341–1349. doi: 10.1136/gut.2006.093484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Adachi M, Kurotani R, Morimura K, Shah Y, Sanford M, Madison BB, Gumucio DL, Marin HE, Peters JM, Young HA, Gonzalez FJ. 2006. Peroxisome proliferator activated receptor γ in colonic epithelial cells protects against experimental inflammatory bowel disease. Gut 55:1104–1113. doi: 10.1136/gut.2005.081745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Shah YM, Morimura K, Gonzalez FJ. 2007. Expression of peroxisome proliferator-activated receptor-in macrophage suppresses experimentally induced colitis. Am J Physiol Gastrointest Liver Physiol 292:G657–66. doi: 10.1152/ajpgi.00381.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Dubuquoy Laurent, Jansson EA, Deeb S, Rakotobe S, Karoui M, Colombel J-F, Auwerx J, Pettersson S, Desreumaux P. 2003. Impaired expression of peroxisome proliferator-activated receptor γin ulcerative colitis. Gastroenterology 124:1265–1276. doi: 10.1016/s0016-5085(03)00271-3 [DOI] [PubMed] [Google Scholar]
- 145. Yamamoto-Furusho JK, Peñaloza-Coronel A, Sánchez-Muñoz F, Barreto-Zuñiga R, Dominguez-Lopez A. 2011. Peroxisome Proliferator-activated receptor-gamma (PPAR-γ) expression is downregulated in patients with active ulcerative colitis. Inflamm Bowel Dis 17:680–681. doi: 10.1002/ibd.21322 [DOI] [PubMed] [Google Scholar]
- 146. Lytle C, Tod TJ, Vo KT, Lee JW, Atkinson RD, Straus DS. 2005. The peroxisome proliferator-activated receptor gamma ligand rosiglitazone delays the onset of inflammatory bowel disease in mice with interleukin 10 deficiency. Inflamm Bowel Dis 11:231–243. doi: 10.1097/01.mib.0000160805.46235.eb [DOI] [PubMed] [Google Scholar]
- 147. Clausen BE, Burkhardt C, Reith W, Renkawitz R, Förster I. 1999. Conditional gene targeting in macrophages and granulocytes using Lysmcre mice. Transgenic Res 8:265–277. doi: 10.1023/a:1008942828960 [DOI] [PubMed] [Google Scholar]
- 148. Akiyama TE, Sakai S, Lambert G, Nicol CJ, Matsusue K, Pimprale S, Lee Y-H, Ricote M, Glass CK, Brewer HB, Gonzalez FJ. 2002. Conditional disruption of the peroxisome proliferator-activated receptor γ gene in mice results in lowered expression of ABCA1, ABCG1, and apoE in macrophages and reduced cholesterol efflux. Mol Cell Biol 22:2607–2619. doi: 10.1128/MCB.22.8.2607-2619.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Jones JR, Shelton KD, Guan Y, Breyer MD, Magnuson MA. 2002. Generation and functional confirmation of a conditional null PPARgamma allele in mice. genesis 32:134–137. doi: 10.1002/gene.10042 [DOI] [PubMed] [Google Scholar]
- 150. Su CG, Wen X, Bailey ST, Jiang W, Rangwala SM, Keilbaugh SA, Flanigan A, Murthy S, Lazar MA, Wu GD. 1999. A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response. J Clin Invest 104:383–389. doi: 10.1172/JCI7145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Saubermann LJ, Nakajima A, Wada K, Zhao S, Terauchi Y, Kadowaki T, Aburatani H, Matsuhashi N, Nagai R, Blumberg RS. 2002. Peroxisome proliferator-activated receptor gamma agonist ligands stimulate a Th2 cytokine response and prevent acute colitis. Inflamm Bowel Dis 8:330–339. doi: 10.1097/00054725-200209000-00004 [DOI] [PubMed] [Google Scholar]
- 152. Celinski K, Dworzanski T, Fornal R, Korolczuk A, Madro A, Slomka M. 2012. Comparison of the anti-inflammatory and therapeutic actions of PPAR-gamma agonists rosiglitazone and troglitazone in experimental colitis. J Physiol Pharmacol 63:631–640. [PubMed] [Google Scholar]
- 153. Celinski K, Dworzanski T, Fornal R, Korolczuk A, Madro A, Brzozowski T, Slomka M. 2013. Comparison of anti-inflammatory properties of peroxisome proliferator-activated receptor gamma agonists rosiglitazone and troglitazone in prophylactic treatment of experimental colitis. J Physiol Pharmacol 64:587–595. [PubMed] [Google Scholar]
- 154. Sun T, Kwong CHT, Gao C, Wei J, Yue L, Zhang J, Ye RD, Wang R. 2020. Amelioration of ulcerative colitis via inflammatory regulation by macrophage-biomimetic nanomedicine. Theranostics 10:10106–10119. doi: 10.7150/thno.48448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Sánchez-Hidalgo M, Martín AR, Villegas I, de la Lastra CA. 2007. Rosiglitazone, a PPARγ ligand, modulates signal transduction pathways during the development of acute TNBS-induced colitis in rats. Eur J Pharmacol 562:247–258. doi: 10.1016/j.ejphar.2007.01.047 [DOI] [PubMed] [Google Scholar]
- 156. Schröder NWJ, Schumann RR. 2005. Single nucleotide polymorphisms of Toll-like receptors and susceptibility to infectious disease. Lancet Infect Dis 5:156–164. doi: 10.1016/S1473-3099(05)01308-3 [DOI] [PubMed] [Google Scholar]
- 157. Hennessy EJ, Parker AE, O’Neill LAJ. 2010. Targeting toll-like receptors: emerging therapeutics Nat Rev Drug Discov 9:293–307. doi: 10.1038/nrd3203 [DOI] [PubMed] [Google Scholar]
- 158. Ungaro R, Fukata M, Hsu D, Hernandez Y, Breglio K, Chen A, Xu R, Sotolongo J, Espana C, Zaias J, Elson G, Mayer L, Kosco-Vilbois M, Abreu MT. 2009. A novel toll-like receptor 4 antagonist antibody ameliorates inflammation but impairs mucosal healing in murine colitis. Am J Physiol Gastrointest Liver Physiol 296:G1167–79. doi: 10.1152/ajpgi.90496.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Fort MM, Mozaffarian A, Stöver AG, Correia J da S, Johnson DA, Crane RT, Ulevitch RJ, Persing DH, Bielefeldt-Ohmann H, Probst P, Jeffery E, Fling SP, Hershberg RM. 2005. A synthetic TLR4 antagonist has anti-inflammatory effects in two murine models of inflammatory bowel disease. J Immunol 174:6416–6423. doi: 10.4049/jimmunol.174.10.6416 [DOI] [PubMed] [Google Scholar]
- 160. Fink MY, Qi X, Shirey KA, Fanaroff R, Chapoval S, Viscardi RM, Vogel SN, Keegan AD. 2022. Mice expressing cosegregating single nucleotide polymorphisms (D298G and N397I). J Immunol 208:2085–2097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Gopalakrishnan A, Richard K, Wahid R, Harley R, Sztein MB, Hawkins LD, Vogel SN. 2022. E6020, a TLR4 agonist adjuvant, enhances both antibody titers and Isotype switching in response to immunization with hapten-protein antigens and is diminished in mice with TLR4 signaling insufficiency. J Immunol 209:1950–1959. doi: 10.4049/jimmunol.2200495 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Perše M, Cerar A. 2012. Dextran sodium sulphate colitis mouse model: traps and tricks. J Biomed Biotechnol 2012:718617. doi: 10.1155/2012/718617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Novak ML, Koh TJ. 2013. Macrophage phenotypes during tissue repair. J Leukoc Biol 93:875–881. doi: 10.1189/jlb.1012512 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Oishi Y, Manabe I. 2018. Macrophages in inflammation, repair and regeneration. Int Immunol 30:511–528. doi: 10.1093/intimm/dxy054 [DOI] [PubMed] [Google Scholar]
- 165. Kubelkova K, Macela A. 2019. Innate immune recognition: an issue more complex than expected. Front Cell Infect Microbiol 9:241. doi: 10.3389/fcimb.2019.00241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Gordon S. 2003. Alternative activation of macrophages. Nat Rev Immunol 3:23–35. doi: 10.1038/nri978 [DOI] [PubMed] [Google Scholar]
- 167. Széles L, Töröcsik D, Nagy L. 2007. PPARgamma in immunity and inflammation: cell types and diseases. Biochim Biophys Acta 1771:1014–1030. doi: 10.1016/j.bbalip.2007.02.005 [DOI] [PubMed] [Google Scholar]
- 168. Orecchioni M, Ghosheh Y, Pramod AB, Ley K. 2019. Macrophage polarization: different gene signatures in M1(LPS+) vs. classically and M2(LPS-) vs. Front Immunol 10:1084. doi: 10.3389/fimmu.2019.01084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Wang L, Waltenberger B, Pferschy-Wenzig E-M, Blunder M, Liu X, Malainer C, Blazevic T, Schwaiger S, Rollinger JM, Heiss EH, Schuster D, Kopp B, Bauer R, Stuppner H, Dirsch VM, Atanasov AG. 2014. Natural product agonists of peroxisome proliferator-activated receptor gamma (PPARγ): a review. Biochem Pharmacol 92:73–89. doi: 10.1016/j.bcp.2014.07.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Milestone AN, Iacucci M, Ghosh S. 2008. Is rosiglitazone a promising treatment for ulcerative colitis Nat Rev Gastroenterol Hepatol 5:486–487. doi: 10.1038/ncpgasthep1226 [DOI] [PubMed] [Google Scholar]
- 171. Liu Y, Li Z, Xu Y, Liu Y, Xue C. 2022. Enzymatic modification of d-mannose alleviates DSS-induced colonic inflammation in mice through macrophage polarization mediated by PPARγ . Food Funct 13:11467–11475. doi: 10.1039/D1FO03897D [DOI] [PubMed] [Google Scholar]
- 172. Decara J, Rivera P, López-Gambero AJ, Serrano A, Pavón FJ, Baixeras E, Rodríguez de Fonseca F, Suárez J. 2020. Peroxisome proliferator-activated receptors: experimental targeting for the treatment of inflammatory bowel diseases. Front Pharmacol 11:730. doi: 10.3389/fphar.2020.00730 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, Evans RM. 2013. Pparγ signaling and metabolism: the good, the bad and the future. Nat Med 19:557–566. doi: 10.1038/nm.3159 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Lewis JD, Lichtenstein GR, Deren JJ, Sands BE, Hanauer SB, Katz JA, Lashner B, Present DH, Chuai S, Ellenberg JH, Nessel L, Wu GD, Rosiglitazone for Ulcerative Colitis Study Group . 2008. Rosiglitazone for active ulcerative colitis: a randomized placebo-controlled trial. Gastroenterology 134:688–695. doi: 10.1053/j.gastro.2007.12.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Lewis JD, Lichtenstein GR, Stein RB, Deren JJ, Judge TA, Fogt F, Furth EE, Demissie EJ, Hurd LB, Su CG, Keilbaugh SA, Lazar MA, Wu GD. 2001. An open-label trial of the PPAR-gamma ligand rosiglitazone for active ulcerative colitis. Am J Gastroenterology 96:3323–3328. doi: 10.1111/j.1572-0241.2001.05333.x [DOI] [PubMed] [Google Scholar]
- 176. Liang HL, Ouyang Q. 2008. A clinical trial of combined use of rosiglitazone and 5-aminosalicylate for ulcerative colitis. World J Gastroenterol 14:114–119. doi: 10.3748/wjg.14.114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Pedersen G, Brynskov J. 2010. Topical rosiglitazone treatment improves ulcerative colitis by restoring peroxisome proliferator-activated receptor-gamma activity. Am J Gastroenterol 105:1595–1603. doi: 10.1038/ajg.2009.749 [DOI] [PubMed] [Google Scholar]
- 178. Powers AC, D’Alessio D.. 2011. Hormones and hormone antagonists , p. 1260. In Brunton, LL (ed.), Goodman & Gilman’s The Pharmacological Basis of Therapeutics 12th edition, 12th ed. McGraw Hill. [Google Scholar]
- 179. Cuzzocrea S, Pisano B, Dugo L, Ianaro A, Maffia P, Patel NSA, Di Paola R, Ialenti A, Genovese T, Chatterjee PK, Di Rosa M, Caputi AP, Thiemermann C. 2004. Rosiglitazone, a ligand of the peroxisome proliferator-activated receptor-γ, reduces acute inflammation. Eur J Pharmacol 483:79–93. doi: 10.1016/j.ejphar.2003.10.056 [DOI] [PubMed] [Google Scholar]
- 180. Hasegawa H, Takano H, Zou Y, Qin Y, Hizukuri K, Odaka K, Toyozaki T, Komuro I. 2005. Pioglitazone, a peroxisome proliferator-activated receptor γ activator, ameliorates experimental autoimmune myocarditis by modulating Th1/Th2 balance. J Mol Cell Cardiol 38:257–265. doi: 10.1016/j.yjmcc.2004.11.010 [DOI] [PubMed] [Google Scholar]
- 181. Yao Q, Liu J, Zhang Z, Li F, Zhang C, Lai B, Xiao L, Wang N. 2018. Peroxisome proliferator-activated receptor γ (PPARγ) induces the gene expression of integrin ΑVβ5 to promote macrophage M2 polarization. J Biol Chem 293:16572–16582. doi: 10.1074/jbc.RA118.003161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Zhou J-P, Yang X-N, Song Y, Zhou F, Liu J-J, Hu Y-Q, Chen L-G. 2021. Rosiglitazone alleviates lipopolysaccharide-induced inflammation in RAW264.7 cells via inhibition of NF-ΚB and in a PPARγ-dependent manner. Exp Ther Med 22:743. doi: 10.3892/etm.2021.10175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Wallach JD, Wang K, Zhang AD, Cheng D, Grossetta Nardini HK, Lin H, Bracken MB, Desai M, Krumholz HM, Ross JS. 2020. Updating insights into rosiglitazone and cardiovascular risk through shared data: individual patient and summary level meta-analyses. BMJ 368:l7078. doi: 10.1136/bmj.l7078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Gopalakrishnan A, Joseph J, Shirey KA, Keegan AD, Boukhvalova MS, Vogel SN, Blanco JCG. 2022. Protection against influenza-induced acute lung injury (ALI) by enhanced induction of M2A macrophages: possible role of PPARγ/RXR ligands in IL-4-induced M2A macrophage differentiation. Front Immunol 13:968336. doi: 10.3389/fimmu.2022.968336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Liu C, Feng T, Zhu N, Liu P, Han X, Chen M, Wang X, Li N, Li Y, Xu Y, Si S. 2015. Identification of a novel selective agonist of PPARγ with no promotion of adipogenesis and less inhibition of osteoblastogenesis. Sci Rep 5:9530. doi: 10.1038/srep09530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Gupta J, Nebreda A. 2014. Analysis of intestinal permeability in mice. Bio-ProtocoL 4. doi: 10.21769/BioProtoc.1289 [DOI] [Google Scholar]
- 187. Viennois E, Chen F, Laroui H, Baker MT, Merlin D. 2013. Dextran sodium sulfate inhibits the activities of both polymerase and reverse transcriptase: lithium chloride purification, a rapid and efficient technique to purify RNA. BMC Res Notes 6:360. doi: 10.1186/1756-0500-6-360 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Viennois E, Tahsin A, Merlin D. 2018. Purification of total RNA from DSS-treated murine tissue via lithium chloride precipitation. Bio-Protocol 8. doi: 10.21769/BioProtoc.2829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Cuesta N, Salkowski CA, Thomas KE, Vogel SN. 2003. Regulation of lipopolysaccharide sensitivity by IFN regulatory factor-2. J Immunol 170:5739–5747. doi: 10.4049/jimmunol.170.11.5739 [DOI] [PubMed] [Google Scholar]
- 190. Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25:402–408. doi: 10.1006/meth.2001.1262 [DOI] [PubMed] [Google Scholar]
- 191. Prantner D, Nallar S, Richard K, Spiegel D, Collins KD, Vogel SN. 2021. Classically activated mouse macrophages produce methylglyoxal that induces a TLR4- and RAGE-independent proinflammatory response. J Leukoc Biol 109:605–619. doi: 10.1002/JLB.3A0520-745RR [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Salkowski CA, Kopydlowski K, Blanco J, Cody MJ, McNally R, Vogel SN. 1999. IL-12 is dysregulated in macrophages from IRF-1 and IRF-2 knockout mice. J Immunol 163:1529–1536. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Forward and reverse qRT-PCR primer sequences.
Data Availability Statement
All primary data are available from the corresponding author on reasonable request.






