Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38563893

ABSTRACT

After birth, the development of secondary lymphoid tissues (SLTs) in the colon is dependent on the expression of the Aryl Hydrocarbon Receptor (AhR) in immune cells as a response to the availability of AhR ligands. However, little is known about how AhR activity from intestinal epithelial cells (IECs) may influence the development of tertiary lymphoid tissues (TLTs). As organized structures that develop at sites of inflammation or infection during adulthood, TLTs serve as localized centers of adaptive immune responses, and their presence has been associated with the resolution of inflammation and tumorigenesis in the colon. Here, we investigated the effect of the conditional loss of AhR activity in IECs in the formation and immune cell composition of TLTs in a model of acute inflammation. In females, loss of AhR activity in IECs reduced the formation of TLTs without significantly changing disease outcomes nor immune cell composition within TLTs. In males lacking AhR expression in IECs, increased disease activity index, lower expression of functional-IEC genes, increased number of TLTs, increased T-cell density, and lower B- to T-cell ratio was observed. These findings may represent an unfavorable prognosis when exposed to DSS-induced epithelial damage compared to females. Sex and loss of IEC AhR also resulted in changes in microbial populations in the gut. Collectively, these data suggest that the formation of TLTs in the colon is influenced by sex and AhR expression in IECs.

2.
Receptors (Basel) ; 2(1): 93-99, 2023 Mar.
Article in English | MEDLINE | ID: mdl-38651159

ABSTRACT

The aryl hydrocarbon receptor (AhR) is overexpressed in many tumor types and exhibits tumor-specific tumor promoter and tumor suppressor-like activity. In colon cancer, most but not all studies suggest that the AhR exhibits tumor suppressor activity which is enhanced by AhR ligands acting as agonists. Our studies investigated the role of the AhR in colon tumorigenesis using wild-type and AhR-knockout mice, the inflammation model of colon tumorigenesis using mice treated with azoxymethane (AOM)/dextran sodium sulfate (DSS) and APCS580/+; KrasG12D/+ mice all of which form intestinal tumors. The effects of tissue-specific AhR loss in the intestine of the tumor-forming mice on colonic stem cells, organoid-initiating capacity, colon tumor formation and mechanisms of AhR-mediated effects were investigated. Loss of AhR enhanced stem cell and tumor growth and in the AOM/DSS model AhR-dependent suppression of FOXM1 and downstream genes was important for AhR-dependent anticancer activity. Furthermore, the effectiveness of interleukin-22 (IL22) in colonic epithelial cells was also dependent on AhR expression. IL22 induced phosphorylation of STAT3, inhibited colonic organoid growth, promoted colonic cell proliferation in vivo and enhanced DNA repair in AOM/DSS-induced tumors. In this mouse model, the AhR suppressed SOCS3 expression and enhanced IL22-mediated activation of STAT3, whereas the loss of the AhR increased levels of SOCS3 which in turn inhibited IL22-induced STAT3 activation. In the APCS580/+; KrasG12D/+ mouse model, the loss of the AhR enhanced Wnt signaling and colon carcinogenesis. Results in both mouse models of colon carcinogenesis were complemented by single cell transcriptomics on colonic intestinal crypts which also showed that AhR deletion promoted expression of FOXM1-regulated genes in multiple colonic cell subtypes. These results support the role of the AhR as a tumor suppressor-like gene in the colon.

3.
Int J Mol Sci ; 23(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35216335

ABSTRACT

Chronic low-grade inflammation is a hallmark of aging, which is now coined as inflamm-aging. Inflamm-aging contributes to many age-associated diseases such as obesity, type 2 diabetes, cardiovascular disease, and inflammatory bowel disease (IBD). We have shown that gut hormone ghrelin, via its receptor growth hormone secretagogue receptor (GHS-R), regulates energy metabolism and inflammation in aging. Emerging evidence suggests that gut microbiome has a critical role in intestinal immunity of the host. To determine whether microbiome is an integral driving force of GHS-R mediated immune-metabolic homeostasis in aging, we assessed the gut microbiome profiles of young and old GHS-R global knockout (KO) mice. While young GHS-R KO mice showed marginal changes in Bacteroidetes and Firmicutes, aged GHS-R KO mice exhibited reduced Bacteroidetes and increased Firmicutes, featuring a disease-susceptible microbiome profile. To further study the role of GHS-R in intestinal inflammation in aging, we induced acute colitis in young and aged GHS-R KO mice using dextran sulfate sodium (DSS). The GHS-R KO mice showed more severe disease activity scores, higher proinflammatory cytokine expression, and decreased expression of tight junction markers. These results suggest that GHS-R plays an important role in microbiome homeostasis and gut inflammation during aging; GHS-R suppression exacerbates intestinal inflammation in aging and increases vulnerability to colitis. Collectively, our finding reveals for the first time that GHS-R is an important regulator of intestinal health in aging; targeting GHS-R may present a novel therapeutic strategy for prevention/treatment of aging leaky gut and inflammatory bowel disease.


Subject(s)
Aging/metabolism , Colitis/metabolism , Dysbiosis/metabolism , Receptors, Ghrelin/metabolism , Animals , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/physiology , Gastrointestinal Microbiome/physiology , Inflammation/metabolism , Inflammatory Bowel Diseases/metabolism , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/physiology , Obesity/metabolism
4.
Essays Biochem ; 65(6): 1003-1013, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34342357

ABSTRACT

In recent years, researchers have demonstrated that estrogen and its receptors, aside from their role in regulating several biological functions, contribute to the development and progression/severity of inflammatory bowel diseases (IBDs). IBDs include both ulcerative colitis (UC) and Crohn's disease (CD). Epidemiological data indicate a clear difference in the incidence, severity, and complications of IBDs between sexes. Men present a higher risk of developing colitis than women and a higher risk of developing colorectal cancer, a common complication of this condition. However, fluctuations of estrogen levels have yielded inconsistent data, where oral contraceptives and hormone replacement therapy have been associated with an increased risk of IBDs in premenopausal women but significantly reduce disease activity after menopause. Likewise, improvement of symptoms related to CD has been reported during pregnancy, but not in UC, who often experience worsening symptoms. In the colonic epithelium, estrogen receptor ß (ERß) is the predominant form of the protein expressed, and it helps maintain normal epithelial function and organization. Preclinical data suggest that ER expression and activation via estrogen confers different responses on disease severity depending on the model used to induce colitis, which may reflect what is observed in patients with IBDs. Hence, this review aims to provide an overview of estrogen and its receptors, particularly ERß, in the pathophysiology of IBDs.


Subject(s)
Colitis, Ulcerative , Colitis , Crohn Disease , Inflammatory Bowel Diseases , Colitis, Ulcerative/epidemiology , Colitis, Ulcerative/metabolism , Crohn Disease/epidemiology , Crohn Disease/metabolism , Female , Humans , Inflammatory Bowel Diseases/metabolism , Male , Pregnancy , Receptors, Estrogen
5.
Microb Cell Fact ; 19(1): 219, 2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33256731

ABSTRACT

BACKGROUND: Diet, loss of aryl hydrocarbon receptor (AhR) expression and their modification of the gut microbiota community composition and its metabolites affect the development of colorectal cancer (CRC). However, the concordance between fecal microbiota composition and the fecal metabolome is poorly understood. Mice with specific AhR deletion (AhRKO) in intestinal epithelial cell and their wild-type littermates were fed a low-fat diet or a high-fat diet. Shifts in the fecal microbiome and metabolome associated with diet and loss of AhR expression were assessed. Microbiome and metabolome data were integrated to identify specific microbial taxa that contributed to the observed metabolite shifts. RESULTS: Our analysis shows that diet has a more pronounced effect on mouse fecal microbiota composition than the impact of the loss of AhR. In contrast, metabolomic analysis showed that the loss of AhR in intestinal epithelial cells had a more pronounced effect on metabolite profile compared to diet. Integration analysis of microbiome and metabolome identified unclassified Clostridiales, unclassified Desulfovibrionaceae, and Akkermansia as key contributors to the synthesis and/or utilization of tryptophan metabolites. CONCLUSIONS: Akkermansia are likely to contribute to the synthesis and/or degradation of tryptophan metabolites. Our study highlights the use of multi-omic analysis to investigate the relationship between the microbiome and metabolome and identifies possible taxa that can be targeted to manipulate the microbiome for CRC treatment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Diet , Feces/microbiology , Metabolome , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Akkermansia/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Colonic Neoplasms/microbiology , DNA, Bacterial , Female , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , RNA, Ribosomal, 16S , Receptors, Aryl Hydrocarbon/genetics
6.
EMBO J ; 39(19): e104319, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32915464

ABSTRACT

The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that senses xenobiotics, diet, and gut microbial-derived metabolites, is increasingly recognized as a key regulator of intestinal biology. However, its effects on the function of colonic stem and progenitor cells remain largely unexplored. Here, we observed that inducible deletion of AhR in Lgr5+ stem cells increases the percentage of colonic stem cells and enhances organoid initiating capacity and growth of sorted stem and progenitor cells, while AhR activation has the opposite effect. Moreover, intestinal-specific AhR knockout increases basal stem cell and crypt injury-induced cell proliferation and promotes colon tumorigenesis in a preclinical colitis-associated tumor model by upregulating FoxM1 signaling. Mechanistically, AhR transcriptionally suppresses FoxM1 expression. Activation of AhR in human organoids recapitulates phenotypes observed in mice, such as reduction in the percentage of colonic stem cells, promotion of stem cell differentiation, and attenuation of FoxM1 signaling. These findings indicate that the AhR-FoxM1 axis, at least in part, mediates colonic stem/progenitor cell behavior.


Subject(s)
Colon/metabolism , Forkhead Box Protein M1/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Signal Transduction , Stem Cells/metabolism , Animals , Female , Forkhead Box Protein M1/genetics , Gene Knockout Techniques , Humans , Male , Mice , Mice, Transgenic , Receptors, Aryl Hydrocarbon/metabolism
7.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G451-G463, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31905023

ABSTRACT

Consumption of a high-fat diet has been associated with an increased risk of developing colorectal cancer (CRC). However, the effects of the interaction between dietary fat content and the aryl hydrocarbon receptor (AhR) on colorectal carcinogenesis remain unclear. Mainly known for its role in xenobiotic metabolism, AhR has been identified as an important regulator for maintaining intestinal epithelial homeostasis. Although previous research using whole body AhR knockout mice has revealed an increased incidence of colon and cecal tumors, the unique role of AhR activity in intestinal epithelial cells (IECs) and modifying effects of fat content in the diet at different stages of sporadic CRC development are yet to be elucidated. In the present study, we have examined the effects of a high-fat diet on IEC-specific AhR knockout mice in a model of sporadic CRC. Although loss of AhR activity in IECs significantly induced the development of premalignant lesions, in a separate experiment, no significant changes in colon mass incidence were observed. Moreover, consumption of a high-fat diet promoted cell proliferation in crypts at the premalignant colon cancer lesion stage and colon mass multiplicity as well as ß-catenin expression and nuclear localization in actively proliferating cells in colon masses. Our data demonstrate the modifying effects of high-fat diet and AhR deletion in IECs on tumor initiation and progression.NEW & NOTEWORTHY Through the use of an intestinal-specific aryl hydrocarbon receptor (AhR) knockout mouse model, this study demonstrates that the expression of AhR in intestinal epithelial cells is required to reduce the formation of premalignant colon cancer lesions. Furthermore, consumption of a high-fat diet and the loss of AhR in intestinal epithelial cells influences the development of colorectal cancer at various stages.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/deficiency , Cell Transformation, Neoplastic/metabolism , Colon/metabolism , Colonic Neoplasms/metabolism , Diet, High-Fat , Epithelial Cells/metabolism , Gene Deletion , Intestinal Mucosa/metabolism , Precancerous Conditions/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Animals , Azoxymethane , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Damage , Disease Models, Animal , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Intestinal Mucosa/pathology , Mice, Inbred C57BL , Mice, Knockout , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
8.
Chem Res Toxicol ; 32(11): 2353-2364, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31621310

ABSTRACT

Many of the protective responses observed for flavonoids in the gastrointestinal track resemble aryl hydrocarbon receptor (AhR)-mediated effects. Therefore, we examined the structure-activity relationships of isoflavones and isomeric flavone and flavanones as AhR ligands on the basis of their induction of CYP1A1, CYP1B1, and UGT1A1 gene expression in colon cancer Caco2 cells and young adult mouse colonocyte (YAMC) cells. Caco2 cells were significantly more Ah-responsive than YAMC cells, and this was due, in part, to flavonoid-induced cytotoxicity in the latter cell lines. The structure-activity relationships for the flavonoids were complex and both response and cell context specific; however, there was significant variability in the AhR activities of the isomeric substituted isoflavones and flavones. For example, 4',5,7-trihydroxyisoflavone (genistein) was AhR-inactive whereas 4',5,7-trihydroxyflavone (apigenin) induced CYP1A1, CYP1B1, and UGT1A1 in Caco2 cells. In contrast, both 5,7-dihydroxy-4-methoxy substituted isoflavone (biochanin A) and flavone (acacetin) induced all three AhR-responsive genes; 4',5,7-trimethoxyisoflavone was a potent AhR agonist, and the isomeric flavone was AhR-inactive. These results coupled with simulation studies modeling flavonoid interaction within the AhR binding pocket demonstrate that the orientation of the substituted phenyl ring at C-2 (flavones) or C-3 (isoflavones) on the common 4-H-chromen-4-one ring strongly influences the activities of isoflavones and flavones as AhR agonists.


Subject(s)
Flavonoids/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Animals , Cell Line , Colon/cytology , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/metabolism , Flavonoids/chemistry , Glucuronosyltransferase/metabolism , Humans , Mice , Models, Molecular , Structure-Activity Relationship
9.
J Vis Exp ; (150)2019 08 30.
Article in English | MEDLINE | ID: mdl-31524856

ABSTRACT

Pancreatic islets, also called the Islets of Langerhans, are a cluster of endocrine cells which produces hormones for glucose regulation and other important biological functions. The islets primarily consist of five types of hormone-secreting cells: α cells secrete glucagon, ß cells secrete insulin, δ cells secrete somatostatin, ε cells secrete ghrelin, and PP cells secrete pancreatic polypeptide. Sixty to 80% of the cells in the islets are ß cells, which are the most important cell population to study insulin secretion. Pancreatic islets are a crucial model system to study ex vivo insulin secretion. Acquiring high quality islets is of great importance for diabetes research. Most islet isolation procedures require technically difficult to access site of collagenase injection, harsh and complex digestion procedures, and multiple density gradient purification steps. This paper features a simple high yield mouse islet isolation method with detailed descriptions and realistic demonstrations, showing the following specific steps: 1) injection of collagenase P at the ampulla of Vater, a small area joining the pancreatic duct and the common bile duct, 2) enzymatic digestion and mechanical separation of the exocrine pancreas, and 3) a single gradient purification step. The advantages of this method are the injection of digestive enzyme using the more accessible ampulla of Vater, more complete digestion using combination of enzymatic and mechanical approaches, and a simpler single gradient purification step. This protocol produces approximately 250-350 islets per mouse; and islets are suitable for various ex vivo studies. Possible caveats of this procedure are potentially damaged islets due to enzymatic digestion and/or prolonged gradient incubation, all of which can be largely avoided by careful ad justification of incubation time.


Subject(s)
Cell Separation/methods , Islets of Langerhans/cytology , Animals , Collagenases , Mice
10.
Exp Biol Med (Maywood) ; 243(10): 864-875, 2018 06.
Article in English | MEDLINE | ID: mdl-29874946

ABSTRACT

Inflammatory bowel disease is a complex collection of disorders. Microbial dysbiosis as well as exposure to toxins including xenoestrogens are thought to be risk factors for inflammatory bowel disease development and relapse. Bisphenol-A has been shown to exert estrogenic activity in the colon and alter intestinal function, but the role that xenoestrogens, such as bisphenol-A , play in colonic inflammation has been previously described but with conflicting results. We investigated the ability of bisphenol-A to exacerbate colonic inflammation and alter microbiota metabolites derived from aromatic amino acids in an acute dextran sulfate sodium-induced colitis model. Female C57BL/6 mice were ovariectomized and exposed to bisphenol-A daily for 15 days. Disease activity measures include body weight, fecal consistency, and rectal bleeding. Colons were scored for inflammation, injury, and nodularity. Alterations in the levels of microbiota metabolites derived from aromatic amino acids known to reflect phenotypic changes in the gut microbiome were analyzed. Bisphenol-A exposure increased mortality and worsened disease activity as well as inflammation and nodularity scores in the middle colon region following dextran sulfate sodium exposure. Unique patterns of metabolites were associated with bisphenol-A consumption. Regardless of dextran sulfate sodium treatment, bisphenol-A reduced levels of tryptophan and several metabolites associated with decreased inflammation in the colon. This is the first study to show that bisphenol-A treatment alone can reduce microbiota metabolites derived from aromatic amino acids in the colon which may be associated with increased colonic inflammation and inflammatory bowel disease. Impact statement As rates of inflammatory bowel disease rise, discovery of the mechanisms related to the development of these conditions is important. Environmental exposure is hypothesized to play a role in etiology of the disease, as are alterations in the gut microbiome and the metabolites they produce. This study is the first to show that bisphenol-A alone alters tryptophan and microbiota metabolites derived from aromatic amino acids in a manner consistent with autoimmune diseases, specifically inflammatory bowel diseases, regardless of dextran sulfate sodium treatment. These findings indicate a potential mechanism by which bisphenol-A negatively affects gut physiology to exacerbate inflammation.


Subject(s)
Amino Acids, Aromatic/metabolism , Benzhydryl Compounds/metabolism , Colitis/pathology , Estrogens, Non-Steroidal/metabolism , Gastrointestinal Microbiome/drug effects , Phenols/metabolism , Animals , Benzhydryl Compounds/administration & dosage , Colitis/chemically induced , Colon/pathology , Disease Models, Animal , Estrogens, Non-Steroidal/administration & dosage , Female , Mice, Inbred C57BL , Phenols/administration & dosage , Survival Analysis
11.
Curr Oncol Rep ; 20(8): 59, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29869224

ABSTRACT

PURPOSE OF REVIEW: Flaxseed and its bioactive components have been associated with a decreased risk of colorectal cancer incidence and progression. This review aims to summarize recent research regarding the role of flaxseed and each of its major dietary bioactive components in reducing colorectal cancer. RECENT FINDINGS: In both human and animal model experiments, flaxseed consumption had beneficial effects on colon physiology associated with reduction in colorectal cancer risk or occurrence. Considered separately, each of flaxseed's major bioactive components, including fiber, alpha-linolenic acid, lignans, and other phytochemicals, is also associated with decreased risk of colonic neoplasms and regulation of cell growth through several potential mechanisms. Collectively, experimental data suggests that consumption of flaxseed and/or its bioactive components may reduce colorectal cancer risk by a variety of mechanisms. Future studies should focus on the mechanisms by which whole flaxseed can prevent colorectal cancer.


Subject(s)
Colorectal Neoplasms/prevention & control , Flax , Phytochemicals/chemistry , Seeds/chemistry , Animals , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Dietary Fiber/pharmacology , Humans , Lignans/pharmacology , Phytochemicals/pharmacology , alpha-Linolenic Acid/pharmacology
12.
Dig Dis Sci ; 62(8): 1977-1984, 2017 08.
Article in English | MEDLINE | ID: mdl-28573506

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) increases the risk of developing colon cancer. This risk is higher in men compared to women, implicating a role for female hormones in the protection against this disease. Studies from our laboratory demonstrated that estradiol (E2) protects against inflammation-associated colon tumor formation when administered following chemical carcinogen and induction of chronic colitis. AIM: This study seeks to better understand the effect of E2 on acute colitis in the presence and absence of estrogen receptor ß (ERß). METHODS: Inflammation was induced by 2,4,6-trinitrobenzenesulfonic acid in wild-type (WT) and ERß knockout (ERßKO) mice implanted with a control or E2-containing pellet and killed 5 days later. Inflammation and injury were scored by a pathologist. Apoptosis and proliferation were assessed by immunohistochemistry. Cytokines were measured by multiplex analysis. RESULTS: E2 treatment reduced inflammation in the middle colon in WT mice and the distal colon in ERßKO mice compared to control mice. WT mice had reduced IL-6, IL-12, IL-17, GM-CSF, IFN-γ, MCP-1, MIP-1α, and TNF-α, and ERßKO had reduced IL-6 and IFN-γ expression in response to E2. Injury scores were lower in E2-treated ERßKO mice compared to control ERßKO mice. ERßKO mice had increased proliferation in the basal third of crypts in the distal colon and decreased apoptosis in the proximal colon. CONCLUSIONS: These data suggest that E2 has differential protective effects against acute colitis in the presence or absence of ERß and provide insight into how E2 may protect against IBD.


Subject(s)
Colitis/drug therapy , Colitis/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/prevention & control , Cytokines/analysis , Cytokines/drug effects , Estrogen Receptor beta/analysis , Estrogen Receptor beta/genetics , Female , Immunohistochemistry , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Trinitrobenzenesulfonic Acid
13.
Cell Death Dis ; 7(11): e2460, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27831561

ABSTRACT

The majority of colon tumors are driven by aberrant Wnt signaling in intestinal stem cells, which mediates an efficient route toward initiating intestinal cancer. Natural lipophilic polyphenols and long-chain polyunsaturated fatty acids (PUFAs) generally suppress Wnt- and NF-κB- (nuclear factor-κ light-chain enhancer of activated B-cell) related pathways. However, the effects of these extrinsic agents on colonic leucine-rich repeat-containing G-protein-coupled receptor 5-positive (Lgr5+) stem cells, the cells of origin of colon cancer, have not been documented to date. Therefore, we examined the effect of n-3 PUFA and polyphenol (curcumin) combination on Lgr5+ stem cells during tumor initiation and progression in the colon compared with an n-6 PUFA-enriched control diet. Lgr5-EGFP-IRES-creERT2 knock-in mice were fed diets containing n-6 PUFA (control), n-3 PUFA, n-6 PUFA+curcumin or n-3 PUFA+curcumin for 3 weeks, followed by 6 azoxymethane (AOM) injections, and terminated 17 weeks after the last injection. To further elucidate the effects of the dietary bioactives at the tumor initiation stage, Lgr5+ stem cells were also assessed at 12 and 24 h post AOM injection. Only n-3 PUFA+curcumin feeding reduced nuclear ß-catenin in aberrant crypt foci (by threefold) compared with control at the progression time point. n-3 PUFA+curcumin synergistically increased targeted apoptosis in DNA-damaged Lgr5+ stem cells by 4.5-fold compared with control at 12 h and maximally reduced damaged Lgr5+ stem cells at 24 h, down to the level observed in saline-treated mice. Finally, RNAseq analysis indicated that p53 signaling in Lgr5+ stem cells from mice exposed to AOM was uniquely upregulated only following n-3 PUFA+curcumin cotreatment. These novel findings demonstrate that Lgr5+ stem cells are uniquely responsive to external dietary cues following the induction of DNA damage, providing a therapeutic strategy for eliminating damaged Lgr5+ stem cells to reduce colon cancer initiation.


Subject(s)
Cell Cycle , Colonic Neoplasms/pathology , Diet , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Aberrant Crypt Foci/metabolism , Animals , Apoptosis/drug effects , Azoxymethane , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogens , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chemoprevention , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Curcumin/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Fatty Acids, Omega-3 , Fish Oils/pharmacology , Green Fluorescent Proteins/metabolism , Mice , Regeneration/drug effects , Risk Factors , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism
14.
Carcinogenesis ; 37(2): 206-14, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26717997

ABSTRACT

Perturbations in DNA damage, DNA repair, apoptosis and cell proliferation in the base of the crypt where stem cells reside are associated with colorectal cancer (CRC) initiation and progression. Although the transformation of leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5)(+) cells is an extremely efficient route towards initiating small intestinal adenomas, the role of Lgr5(+) cells in CRC pathogenesis has not been well investigated. Therefore, we further characterized the properties of colonic Lgr5(+) cells compared to differentiated cells in Lgr5-EGFP-IRES-creER(T2) knock-in mice at the initiation stage of carcinogen azoxymethane (AOM)-induced tumorigenesis using a quantitative immunofluorescence microscopy approach. At 12 and 24h post-AOM treatment, colonic Lgr5(+) stem cells (GFP(high)) were preferentially damaged by carcinogen, exhibiting a 4.7-fold induction of apoptosis compared to differentiated (GFP(neg)) cells. Furthermore, with respect to DNA repair, O(6)-methylguanine DNA methyltransferase (MGMT) expression was preferentially induced (by 18.5-fold) in GFP(high) cells at 24h post-AOM treatment compared to GFP(neg) differentiated cells. This corresponded with a 4.3-fold increase in cell proliferation in GFP(high) cells. These data suggest that Lgr5(+) stem cells uniquely respond to alkylation-induced DNA damage by upregulating DNA damage repair, apoptosis and cell proliferation compared to differentiated cells in order to maintain genomic integrity. These findings highlight the mechanisms by which colonic Lgr5(+) stem cells respond to cancer-causing environmental factors.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Homeostasis/drug effects , Intestinal Mucosa/cytology , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Carcinogens/toxicity , Cell Proliferation/drug effects , Cell Proliferation/physiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , DNA Damage/drug effects , DNA Damage/physiology , DNA Repair/drug effects , DNA Repair/physiology , Disease Models, Animal , Gene Knock-In Techniques , Homeostasis/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice , Mutagens/toxicity , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Stem Cells/pathology
15.
Food Chem Toxicol ; 87: 23-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26593444

ABSTRACT

Epidemiological and animal data have demonstrated the protective effects of estrogen signaling on colon carcinogenesis. Nonetheless, studies have suggested that estrogen replacement therapy is positively correlated to increased risk of breast cancer. Therefore, there is considerable interest in investigating novel phytoestrogens that mimic the protective actions of estrogen in the colon. Trigonelline (Trig) and 3,3-diindolylmethane (DIM) have been reported as phytoestrogens in spite of their distinct chemical structures from other phytoestrogens. Both compounds elicit estrogenic responses without directly interacting with the binding domain of the estrogen receptor (ER). We examined the influence of Trig and DIM on non-malignant colonocytes. Both compounds reduced cell growth of young adult mouse colonocytes (YAMCs). Trig and DIM induced cell cycle arrest in the G0/G1 phase and enhanced apoptosis in YAMCs. The inhibitory effect of Trig on cell growth was disrupted by co-treatment of ICI 182,780, an ER antagonist. DIM elevated ER mediated transcriptional activity. Both compounds changed gene expression related to apoptosis and cell proliferation in unique ways. In conclusion, Trig and DIM impact cell physiology and gene expression in YAMCs via novel estrogenic actions and these data suggest that intake of novel phytoestrogens may activate protective effects of estrogen signaling in the colon.


Subject(s)
Alkaloids/pharmacology , Cell Proliferation/drug effects , Colon/cytology , Estrogens/pharmacology , Indoles/pharmacology , Alkaloids/chemistry , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cells, Cultured , Estrogens/chemistry , Indoles/chemistry , Mice , Molecular Structure , Signal Transduction
16.
Cancer Res ; 75(24): 5392-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26631266

ABSTRACT

p53 has been shown to mediate cancer stem-like cell function by suppressing pluripotency and cellular dedifferentiation. However, there have been no studies to date that have addressed the specific effects of p53 loss in colonic adult stem cells. In this study, we investigated the consequences of conditionally ablating p53 in the highly relevant Lgr5(+) stem cell population on tumor initiation and progression in the colon. In a mouse model of carcinogen (AOM)-induced colon cancer, tamoxifen-inducible Lgr5-driven deletion of p53 reduced apoptosis and increased proliferation of crypt stem cells, but had no effect on tumor incidence or size. Conversely, in a mouse model of colitis-associated cancer, in which mice are exposed to AOM and the potent inflammation inducer DSS, stem cell-specific p53 deletion greatly enhanced tumor size and incidence in the colon. These novel findings suggest that the loss of p53 function in stem cells enables colonic tumor formation only when combined with DNA damage and chronic inflammation. Furthermore, we propose that stem cell targeting approaches are valuable for interrogating prevention and therapeutic strategies that aim to specifically eradicate genetically compromised stem cells.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Neoplastic Stem Cells/pathology , Tumor Suppressor Protein p53/genetics , Animals , Colitis/complications , Colon/pathology , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Mutant Strains , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism
17.
Food Funct ; 6(3): 749-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25553799

ABSTRACT

Activation of estrogen receptor-ß (ERß) is an important mechanism for colon cancer prevention. Specific sorghum varieties that contain flavones were shown to activate ER in non-malignant colonocytes at low concentrations. This study aimed to determine positive interactions among estrogenic flavonoids most relevant in sorghum. Apigenin and naringenin were tested separately and in combination for their ability to influence ER-mediated cell growth in non-malignant young adult mouse colonocytes (YAMC). Sorghum extracts high in specific flavanones and flavones were also tested. Apigenin reduced ER-mediated YAMC cell growth comparable to physiological levels of estradiol (E2, 1 nM) at 1 µM; naringenin had similar effect at 10 µM. However, when combined, 0.1 µM apigenin plus 0.05 µM naringenin produced similar effect as 1 nM E2; these concentrations represented 1/10th and 1/200th, respectively, of the active concentrations of apigenin and naringenin, demonstrating a strong enhanced action. A sorghum extract higher in flavones (apigenin and luteolin) (4.8 mg g(-1)) was more effective (5 µg mL(-1)) at activating ER in YAMC than a higher flavanone (naringenin and eriodictyol) (28.1 mg g(-1)) sorghum extract (10 µg mL(-1)). Enhanced actions observed for apigenin and naringenin were adequate to explain the level of effects produced by the high flavone and flavanone sorghum extracts. Strong positive interactions among sorghum flavonoids may enhance their ability to contribute to colon cancer prevention beyond what can be modeled using target compounds in isolation.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apigenin/pharmacology , Colon/drug effects , Flavanones/pharmacology , Intestinal Mucosa/drug effects , Phytoestrogens/pharmacology , Sorghum/chemistry , Animals , Anticarcinogenic Agents/analysis , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/isolation & purification , Apigenin/agonists , Apigenin/chemistry , Apigenin/isolation & purification , Cell Line , Cell Proliferation/drug effects , Colon/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/prevention & control , Drug Synergism , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Flavanones/agonists , Flavanones/analysis , Flavanones/chemistry , Flavanones/isolation & purification , Functional Food/analysis , Intestinal Mucosa/metabolism , Luteolin/analysis , Luteolin/isolation & purification , Luteolin/pharmacology , Mice , Osmolar Concentration , Phytoestrogens/agonists , Phytoestrogens/chemistry , Phytoestrogens/isolation & purification , Pigments, Biological/biosynthesis , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Seeds/chemistry , Seeds/metabolism , Sorghum/metabolism
18.
Appl Environ Microbiol ; 79(18): 5763-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23872567

ABSTRACT

Intestinal microbial dysbiosis contributes to the dysmetabolism of luminal factors, including steroid hormones (sterones) that affect the development of chronic gastrointestinal inflammation and the incidence of sterone-responsive cancers of the breast, prostate, and colon. Little is known, however, about the role of specific host sterone nucleoreceptors, including estrogen receptor ß (ERß), in microbiota maintenance. Herein, we test the hypothesis that ERß status affects microbiota composition and determine if such compositionally distinct microbiota respond differently to changes in diet complexity that favor Proteobacteria enrichment. To this end, conventionally raised female ERß(+/+) and ERß(-/-) C57BL/6J mice (mean age of 27 weeks) were initially reared on 8604, a complex diet containing estrogenic isoflavones, and then fed AIN-76, an isoflavone-free semisynthetic diet, for 2 weeks. 16S rRNA gene surveys revealed that the fecal microbiota of 8604-fed mice and AIN-76-fed mice differed, as expected. The relative diversity of Proteobacteria, especially the Alphaproteobacteria and Gammaproteobacteria, increased significantly following the transition to AIN-76. Distinct patterns for beneficial Lactobacillales were exclusive to and highly abundant among 8604-fed mice, whereas several Proteobacteria were exclusive to AIN-76-fed mice. Interestingly, representative orders of the phyla Proteobacteria, Bacteroidetes, and Firmicutes, including the Lactobacillales, also differed as a function of murine ERß status. Overall, these interactions suggest that sterone nucleoreceptor status and diet complexity may play important roles in microbiota maintenance. Furthermore, we envision that this model for gastrointestinal dysbiosis may be used to identify novel probiotics, prebiotics, nutritional strategies, and pharmaceuticals for the prevention and resolution of Proteobacteria-rich dysbiosis.


Subject(s)
Bacteria/classification , Biota , Diet/methods , Estrogen Receptor beta/metabolism , Gastrointestinal Tract/microbiology , Animals , Bacteria/genetics , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Estrogen Receptor beta/deficiency , Isoflavones/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
19.
Endocr Relat Cancer ; 20(4): 515-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23702470

ABSTRACT

Postmenopausal women on estrogen replacement therapy (ERT) have a reduced risk of developing colon cancer compared with postmenopausal women not on ERT, suggesting a role for estradiol (E2) in protection against this disease. To determine whether E2 protects against inflammation-associated colon cancer when administered following the initiation of colonic DNA damage, in this study, we implanted E2-containing pellets into mice after co-treatment with azoxymethane and two rounds of dextran sulfate sodium (DSS). Wild-type (WT) E2-treated mice had reduced numbers and average area of adenocarcinomas compared with the control mice. These effects were lost in estrogen receptor-ß (Erß (Esr2)) knockout mice. Surprisingly, apoptosis was reduced and cell proliferation was increased in sections from tumors of the WT E2 mice compared with the WT control mice. These findings are probably due, in part, to a reduction in ERß expression in colonic epithelial cells as the cells progressed from a non-malignant to a cancerous state as enhanced apoptosis was observed in normal colonocytes expressing higher levels of ERß. Furthermore, epithelial cells within the tumors had dramatically increased ERα mRNA and protein expression compared with the non-diseased mice. We conclude that while E2 treatment resulted in an overall suppression of colonic adenocarcinoma formation, reduced ERß expression accompanied by enhanced ERα expression caused an altered colonocyte response to E2 treatment compared with the earlier stages of colon cancer development. These data are the first examples of decreased ERß expression concurrent with increased ERα expression as a disease develops and highlight the importance of understanding the timing of E2 exposure with regard to the prevention of inflammation-associated colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Animals , Azoxymethane , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Dextran Sulfate , Estradiol/blood , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogens/blood , Female , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Nutr Cancer ; 64(3): 419-27, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22369068

ABSTRACT

Evidence indicates sorghum may be protective against colon cancer; however, the mechanisms are unknown. Estrogen is believed to protect against colon cancer development by inducing apoptosis in damaged nonmalignant colonocytes. Three sorghum extracts (white, red, and black) were screened for estrogenic activity using cell models expressing estrogen receptor α (ER-α; MCF-7 breast cancer cells) and ß [ER-ß; nonmalignant young adult mouse colonocytes (YAMC)]. Black and white sorghum extracts had significant estrogenic activity mediated through both estrogen receptors at 1-5 and 5-10 µg/mL, respectively; but red sorghum did not. Activation of ER-ß in YAMC reduced cell growth via induction of apoptosis. Only the black and red sorghums contained 3-deoxyanthocyanins; however, these compounds were non-estrogenic. Flavones with estrogenic properties, luteolin (0.41-2.12 mg/g) and apigenin (1.1-1.4 mg/g), and their O-methyl derivatives (0.70-0.95 mg/g) were detected in white and black sorghums, but not in the red sorghum. On the other hand, naringenin, a flavanone known to interfere with transcriptional activities of estrogen, was only detected in the red sorghum extract (as its 7-O-glycoside) at relatively high concentration (11.8 mg/g). Sorghum flavonoid composition has important implications on possible modes of chemoprotection by sorghum against colon carcinogenesis.


Subject(s)
Apoptosis/drug effects , Colon/cytology , Flavanones/pharmacology , Plant Extracts/pharmacology , Sorghum/chemistry , Animals , Apigenin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/pathology , Epithelial Cells/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Luteolin/pharmacology , Mice , Phytoestrogens/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...