Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
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.
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
3.
Eur J Cancer Prev ; 28(5): 383-389, 2019 09.
Article in English | MEDLINE | ID: mdl-30234553

ABSTRACT

Multicomponent therapy has gained interest for its potential to synergize and subsequently lower the effective dose of each constituent required to reduce colon cancer risk. We have previously showed that rapidly cycling Lgr5 stem cells are exquisitely sensitive to extrinsic dietary factors that modulate colon cancer risk. In the present study, we quantified the dose-dependent synergistic properties of dietary n-3 polyunsaturated fatty acids (PUFA) and curcumin (Cur) to promote targeted apoptotic deletion of damaged colonic Lgr5 stem cells. For this purpose, both heterogeneous bulk colonocytes and Lgr5 stem cells were isolated from Lgr5-EGFP-IRES-CreER knock-in mice injected with azoxymethane (AOM). Isolated cells were analyzed for DNA damage (γH2AX), apoptosis (cleaved caspase-3), and targeted apoptosis (both γH2AX and cleaved caspase-3) at 12 h post-AOM injection. Comparison of the percentage of targeted apoptosis in Lgr5 stem cells (GFP) across a broad bioactive dose-range revealed an ED50 of 16.0 mg/day n-3 PUFA + 15.9 mg/day Cur. This corresponded to a human equivalent dose of 3.0 g n-3 PUFA + 3.0 g Cur. In summary, our results provide evidence that a low dose (n-3 PUFA + Cur) combination diet reduces AOM-induced DNA damage in Lgr5 stem cells and enhances targeted apoptosis of DNA-damaged cells, implying that a lower human equivalent dose can be utilized in future human clinical trials.


Subject(s)
Colonic Neoplasms/prevention & control , Curcumin/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Neoplastic Stem Cells/drug effects , Receptors, G-Protein-Coupled/metabolism , Animals , Apoptosis/drug effects , Azoxymethane/toxicity , Carcinogens/toxicity , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Colon/cytology , Colon/drug effects , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dietary Supplements , Dose-Response Relationship, Drug , Female , Gene Knock-In Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/genetics
4.
Sci Rep ; 7(1): 10163, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28860561

ABSTRACT

Aryl hydrocarbon receptor (AhR) ligands are important for gastrointestinal health and play a role in gut inflammation and the induction of T regulatory cells, and the short chain fatty acids (SCFAs) butyrate, propionate and acetate also induce similar protective responses. Initial studies with butyrate demonstrated that this compound significantly increased expression of Ah-responsive genes such as Cyp1a1/CYP1A1 in YAMC mouse colonocytes and Caco-2 human colon cancer cell lines. Butyrate synergistically enhanced AhR ligand-induced Cyp1a1/CYP1A1 in these cells with comparable enhancement being observed for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and also microbiota-derived AhR ligands tryptamine, indole and 1,4-dihydroxy-2-naphthoic acid (DHNA). The effects of butyrate on enhancing induction of Cyp1b1/CYP1B1, AhR repressor (Ahrr/AhRR) and TCDD-inducible poly(ADP-ribose)polymerase (Tiparp/TiPARP) by AhR ligands were gene- and cell context-dependent with the Caco-2 cells being the most responsive cell line. Like butyrate and propionate, the prototypical hydroxyamic acid-derived histone deacetylase (HDAC) inhibitors Panobinostat and Vorinostat also enhanced AhR ligand-mediated induction and this was accompanied by enhanced histone acetylation. Acetate also enhanced basal and ligand-inducible Ah responsiveness and histone acetylation, demonstrating that acetate was an HDAC inhibitor. These results demonstrate SCFA-AhR ligand interactions in YAMC and Caco-2 cells where SCFAs synergistically enhance basal and ligand-induced expression of AhR-responsive genes.


Subject(s)
Colon/chemistry , Colonic Neoplasms/genetics , Fatty Acids, Volatile/pharmacology , Gene Regulatory Networks , Receptors, Aryl Hydrocarbon/metabolism , Animals , Butyrates/pharmacology , Caco-2 Cells , Cells, Cultured , Colon/cytology , Colon/metabolism , Colonic Neoplasms/metabolism , Gene Knockout Techniques , Gene Regulatory Networks/drug effects , Humans , Ligands , Mice , Propionates/pharmacology
5.
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
6.
Data Brief ; 6: 398-404, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26862588

ABSTRACT

With the identification of Lgr5 as a definitive marker for intestinal stem cells, we used the highly novel, recently described, Lgr5-EGFP-IRES-cre ER (T2) knock in mouse model. Mice were injected with azoxymethane (AOM, a colon carcinogen) or saline (control) and fed a chemo-protective diet containing n-3 fatty acids and fermentable fiber (n-3 PUFA+pectin) or a control diet (n-6 PUFA + cellulose). Single cells were isolated from colonic mucosa crypts and three discrete populations of cells were collected via fluorescence activated cell sorting (FACS): Lgr5(high) (stem cells), Lgr5(low) (daughter cells) and Lgr5(negative) (differentiated cells). microRNA profiling and RNA sequencing were performed from the same sample and analyzed. These data refer to 'Comparative effects of diet and carcinogen on microRNA expression in the stem cell niche of the mouse colonic crypt' (Shah et al., 2016) [5].

7.
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
8.
Biochim Biophys Acta ; 1862(1): 121-34, 2016 01.
Article in English | MEDLINE | ID: mdl-26493444

ABSTRACT

There is mounting evidence that noncoding microRNAs (miRNA) are modulated by select chemoprotective dietary agents. For example, recently we demonstrated that the unique combination of dietary fish oil (containing n-3 fatty acids) plus pectin (fermented to butyrate in the colon) (FPA) up-regulates a subset of putative tumor suppressor miRNAs in intestinal mucosa, and down-regulates their predicted target genes following carcinogen exposure as compared to control (corn oil plus cellulose (CCA)) diet. To further elucidate the biological effects of diet and carcinogen modulated miR's in the colon, we verified that miR-26b and miR-203 directly target PDE4B and TCF4, respectively. Since perturbations in adult stem cell dynamics are generally believed to represent an early step in colon tumorigenesis and to better understand how the colonic stem cell population responds to environmental factors such as diet and carcinogen, we additionally determined the effects of the chemoprotective FPA diet on miRNAs and mRNAs in colonic stem cells obtained from Lgr5-EGFP-IRES-creER(T2) knock-in mice. Following global miRNA profiling, 26 miRNAs (P<0.05) were differentially expressed in Lgr5(high) stem cells as compared to Lgr5(negative) differentiated cells. FPA treatment up-regulated miR-19b, miR-26b and miR-203 expression as compared to CCA specifically in Lgr5(high) cells. In contrast, in Lgr5(negative) cells, only miR-19b and its indirect target PTK2B were modulated by the FPA diet. These data indicate for the first time that select dietary cues can impact stem cell regulatory networks, in part, by modulating the steady-state levels of miRNAs. To our knowledge, this is the first study to utilize Lgr5(+) reporter mice to determine the impact of diet and carcinogen on miRNA expression in colonic stem cells and their progeny.


Subject(s)
Carcinogens , Colon/pathology , Colonic Neoplasms/genetics , Diet , Fatty Acids, Omega-3/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Stem Cell Niche , Animals , Carcinogens/metabolism , Carcinogens/toxicity , Colon/metabolism , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Focal Adhesion Kinase 2/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Protective Factors , Stem Cell Niche/drug effects , Transcription Factor 4/genetics
9.
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
10.
Am J Physiol Gastrointest Liver Physiol ; 309(1): G1-9, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25977509

ABSTRACT

Evidence suggests that targeting cancer cell energy metabolism might be an effective therapeutic approach for selective ablation of malignancies. Using a Seahorse Extracellular Flux Analyzer, we have demonstrated that select environmental agents can alter colonic mitochondrial function by increasing respiration-induced proton leak, thereby inducing apoptosis, a marker of colon cancer risk. To further probe bioenergetics in primary intestinal cells, we developed methodology that can be modified and adapted to measure the bioenergetic profiles of colonic crypts, the basic functional unit of the colon, and colonic organoids, an ex vivo 3D culture of colonic crypts. Furthermore, in combination with the MoFlo Astrios High-Speed Cell Sorter, we were able to measure the bioenergetic profiles of colonic adult stem and daughter cells from Lgr5-EGFP-IRES-creER(T2) transgenic mice. We examined the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a full arylhydrocarbon receptor agonist, known to affect gastrointestinal function and cancer risk, on the bioenergetic profiles of intestinal epithelial cells. Mouse colonic crypts, organoids, or sorted single cells were seeded onto Matrigel-precoated Seahorse XF24 microplates for extracellular flux analysis. Temporal analyses revealed distinct energy metabolic profiles in crypts and organoids challenged with TCDD. Furthermore, sorted Lgr5(+) stem cells exhibited a Warburg-like metabolic profile. This is noteworthy because perturbations in stem cell dynamics are generally believed to represent the earliest step toward colon tumorigenesis. We propose that our innovative methodology may facilitate future in vivo/ex vivo metabolic studies using environmental agents affecting colonocyte energy metabolism.


Subject(s)
Biological Assay/methods , Cell Separation/methods , Colon/metabolism , Energy Metabolism , Flow Cytometry , Organoids/metabolism , Stem Cells/metabolism , Animals , Cells, Cultured , Colon/cytology , Colon/drug effects , Energy Metabolism/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Organoids/cytology , Organoids/drug effects , Phenotype , Polychlorinated Dibenzodioxins/pharmacology , Stem Cells/cytology , Stem Cells/drug effects , Time Factors , Tissue Culture Techniques
11.
Carcinogenesis ; 35(3): 606-12, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24336194

ABSTRACT

Arachidonic acid (20:4(Δ5,8,11,14), AA)-derived prostaglandin E2 (PGE2) promotes colon cancer development. In contrast, chemoprotective n-3 polyunsaturated fatty acids supplant AA, thereby decreasing PGE2 biosynthesis in colonocytes, with eicosapentaenoic acid (20:5(Δ5,8,11,14,17), EPA) in particular being metabolized to a novel 3-series E-prostaglandin (PGE3), a putative anti-tumorigenic-cyclooxygenase metabolite. Because transformation of adult stem cells is an extremely important route toward initiating intestinal cancer, we utilized the leucine-rich-repeat-containing G-protein-coupled receptor 5 (Lgr5)-enhanced green fluorescent protein-internal ribosome entry site (IRES)-creER(T2) knock-in mouse model to isolate and culture colonic organoids, in order to document ex vivo responses to exogenous PGE2 and PGE3. Colonic crypts were isolated from transgenic mice and cultured in a Matrigel-based three-dimensional platform. Organoids were treated with exogenous PGE2, PGE3 or dimethyl sulfoxide (vehicle control) for 5 days and the number of viable organoids was recorded daily. Subsequently, samples were processed for immunohistochemistry, flow cytometry and real-time PCR analyses. PGE2 promoted optimal organoid growth and induced significantly higher levels of cell proliferation (P < 0.05) compared with PGE3 and control. In contrast, the Lgr5-green fluorescent protein-positive stem cell number was uniquely elevated by >2-fold in PGE2-treated cultures compared with PGE3 and control. This coincided with the upregulation of stem-cell-related Sox9, Axin2 and Cd44 messenger RNAs. Our results demonstrate that relative to AA-derived PGE2, a known promoter of colon tumorigenesis, EPA-derived PGE3 has diminished ability to support colonic stem cell expansion in mouse colonic organoids.


Subject(s)
Cell Division/drug effects , Colon/drug effects , Prostaglandins/pharmacology , Stem Cells/drug effects , Colon/cytology , Green Fluorescent Proteins/genetics , Humans , Real-Time Polymerase Chain Reaction , Stem Cells/cytology
12.
Am J Physiol Cell Physiol ; 304(9): C905-17, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23426968

ABSTRACT

Epidermal growth factor receptor (EGFR)-mediated signaling is required for optimal intestinal wound healing. Since n-3 polyunsaturated fatty acids (PUFA), specifically docosahexaenoic acid (DHA), alter EGFR signaling and suppress downstream activation of key signaling pathways, we hypothesized that DHA would be detrimental to the process of intestinal wound healing. Using a mouse immortalized colonocyte model, DHA uniquely reduced EGFR ligand-induced receptor activation, whereas DHA and its metabolic precursor eicosapentaenoic acid (EPA) reduced wound-induced EGFR transactivation compared with control (no fatty acid or linoleic acid). Under wounding conditions, the suppression of EGFR activation was associated with a reduction in downstream activation of cytoskeletal remodeling proteins (PLCγ1, Rac1, and Cdc42). Subsequently, DHA and EPA reduced cell migration in response to wounding. Mice were fed a corn oil-, DHA-, or EPA-enriched diet prior to intestinal wounding (2.5% dextran sodium sulfate for 5 days followed by termination after 0, 3, or 6 days of recovery). Mortality was increased in EPA-fed mice and colonic histological injury scores were increased in EPA- and DHA-fed mice compared with corn oil-fed (control) mice. Although kinetics of colonic EGFR activation and downstream signaling (PLCγ1, Rac1, and Cdc42) were delayed by both n-3 PUFA, colonic repair was increased in EPA- relative to DHA-fed mice. These results indicate that, during the early response to intestinal wounding, DHA and EPA uniquely delay the activation of key wound-healing processes in the colon. This effect is mediated, at least in part, via suppression of EGFR-mediated signaling and downstream cytoskeletal remodeling.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , ErbB Receptors/metabolism , Protein Processing, Post-Translational , Wound Healing , Animals , Arachidonic Acid/metabolism , Cell Movement , Cells, Cultured , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Colon/drug effects , Colon/pathology , Corn Oil/administration & dosage , Dextran Sulfate , Dietary Supplements , Docosahexaenoic Acids/physiology , Eicosapentaenoic Acid/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neuropeptides/metabolism , Oxygen Consumption , Phosphorylation , Signal Transduction , alpha-Linolenic Acid/pharmacology , alpha-Linolenic Acid/physiology , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
13.
Biochim Biophys Acta ; 1822(10): 1600-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22750333

ABSTRACT

Since aberrant wound healing and chronic inflammation can promote malignant transformation, we determined whether dietary bioactive fish oil (FO)-derived n-3 polyunsaturated fatty acids (n-3 PUFA) modulate stem cell kinetics in a colitis-wounding model. Lgr5-LacZ and Lgr5-EGFP-IRES-creER(T2) mice were fed diets enriched with n-3 PUFA vs n-6 PUFA (control) and exposed to dextran sodium sulfate (DSS) for 5days in order to induce crypt damage and colitis throughout the colon. Stem cell number, cell proliferation, apoptosis, expression of stem cell (Lgr5, Sox9, Bmi1, Hopx, mTert, Ascl2, and DCAMKL-1) and inflammation (STAT3) markers were quantified. DSS treatment resulted in the ablation of Lgr5(+) stem cells in the distal colon, concurrent with the loss of distal crypt structure and proliferating cells. Lgr5, Ascl2 and Hopx mRNA expression levels were decreased in damaged colonic mucosa. Lgr5(+) stem cells reappeared at day 5 of DSS recovery, with normal levels attained by day 6 of recovery. There was no effect of diet on the recovery of stem cells. FO fed animals exhibited higher levels of phospho-STAT3 at all time points, consistent with a higher wounding by DSS in FO feeding. n-3 PUFA-fed mice exhibited a reduction in stem cell associated factors, Ascl2, Axin2 and EphB3. These results indicate that rapidly cycling Lgr5(+) stem cells residing at position 1 in the colon epithelium are highly susceptible to DSS-induced damage and that dietary cues can impact stem cell regulatory networks.


Subject(s)
Colon/physiology , Regeneration/physiology , Stem Cells/physiology , Wound Healing/physiology , Animals , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Dextran Sulfate , Diet , Fatty Acids, Omega-3/metabolism , Fish Oils/metabolism , Gene Expression/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Regeneration/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Wound Healing/genetics
14.
J Biomed Opt ; 17(1): 016006, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22352656

ABSTRACT

Large area confocal microscopy may provide fast, high-resolution image acquisition for evaluation of tissue in pre-clinical studies with reduced tissue processing in comparison to histology. We present a rapid beam and stage-scanning confocal fluorescence microscope to image cellular and tissue features along the length of the entire excised mouse colon. The beam is scanned at 8,333 lines/sec by a polygon scanning mirror while the specimen is scanned in the orthogonal axis by a motorized translation stage with a maximum speed of 7 mm/sec. A single 1 × 60 mm(2) field of view image spanning the length of the mouse colon is acquired in 10 s. Z-projection images generated from axial image stacks allow high resolution imaging of the surface of non-flat specimens. In contrast to the uniform size, shape, and distribution of colon crypts in confocal images of normal colon, confocal images of chronic bowel inflammation exhibit heterogeneous tissue structure with localized severe crypt distortion.


Subject(s)
Colitis/pathology , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Animals , Colon/pathology , Disease Models, Animal , Equipment Design , Histocytochemistry , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Mice , Mice, Inbred C57BL , Microscopy, Confocal/instrumentation , Microscopy, Fluorescence/instrumentation
15.
Cancer Prev Res (Phila) ; 4(8): 1267-74, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21490130

ABSTRACT

An alteration of mitochondrial function can result in disruption of redox homeostasis and is associated with abnormal cancer cell growth. Manganese superoxide dismutase (SOD2) and glutathione peroxidase 4 (Gpx4) are two of the most important antioxidant defense enzymes that protect cells against oxidative stress. We had previously shown that n-3 polyunsaturated fatty acids (PUFA) promote colonocyte apoptosis, a marker of colon cancer risk, in part by enhancing phospholipid oxidation. To elucidate the mechanisms regulating oxidative stress-induced apoptosis in vivo, we fed heterozygous SOD2(Het), Gpx4(Het), and transgenic Gpx4(Tg) mice diets containing either 15% corn oil by weight (CO, enriched in n-6 PUFA) or 3.5% CO + 11.5% fish oil (FO, enriched in n-3 PUFA) for 4 weeks. Our data showed that (i) genetic predeposition to oxidative stress facilitates apoptosis in the mouse colon (Gpx4(Het) > SOD2(Het) > Wt > Gpx4(Tg)), (ii) dietary n-3 PUFA have an additive effect on the induction of apoptosis in Gpx4(Het) and SOD2(Het) mice; and (iii) dietary n-3 PUFA reverse the phenotype in oxidatively protected Gpx4(Tg) mice by elevating apoptosis to a level observed in wild-type (Wt; control) animals. Complimentary experiments examining colonic mitochondrial bioenergetic profiles indicate that FO-fed mice exhibit a significantly (P < 0.05) increased respiration-induced proton leak relative to control CO treatment. This finding was consistent with a loss of membrane potential in response to chronic oxidative stress and supports the contention that n-3 PUFA alter mitochondrial metabolic activity, thereby enhancing apoptosis and reducing colon cancer risk.


Subject(s)
Apoptosis , Colon/pathology , Dietary Fats, Unsaturated/metabolism , Fish Oils/metabolism , Mitochondria/metabolism , Animals , Cardiolipins/metabolism , Colon/metabolism , Crosses, Genetic , Genotype , Glutathione Peroxidase/metabolism , Mice , Mice, Inbred C57BL , Oxidative Stress , Oxygen Consumption , Phospholipid Hydroperoxide Glutathione Peroxidase , Protons
16.
Br J Nutr ; 106(4): 519-29, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21401974

ABSTRACT

Both fish oil (FO) and curcumin have potential as anti-tumour and anti-inflammatory agents. To further explore their combined effects on dextran sodium sulphate (DSS)-induced colitis, C57BL/6 mice were randomised to four diets (2 × 2 design) differing in fatty acid content with or without curcumin supplementation (FO, FO+2 % curcumin, maize oil (control, MO) or MO+2 % curcumin). Mice were exposed to one or two cycles of DSS in the drinking-water to induce either acute or chronic intestinal inflammation, respectively. FO-fed mice exposed to the single-cycle DSS treatment exhibited the highest mortality (40 %, seventeen of forty-three) compared with MO with the lowest mortality (3 %, one of twenty-nine) (P = 0·0008). Addition of curcumin to MO increased (P = 0·003) mortality to 37 % compared with the control. Consistent with animal survival data, following the one- or two-cycle DSS treatment, both dietary FO and curcumin promoted mucosal injury/ulceration compared with MO. In contrast, compared with other diets, combined FO and curcumin feeding enhanced the resolution of chronic inflammation and suppressed (P < 0·05) a key inflammatory mediator, NF-κB, in the colon mucosa. Mucosal microarray analysis revealed that dietary FO, curcumin and FO plus curcumin combination differentially modulated the expression of genes induced by DSS treatment. These results suggest that dietary lipids and curcumin interact to regulate mucosal homeostasis and the resolution of chronic inflammation in the colon.


Subject(s)
Colitis/diet therapy , Colon/metabolism , Curcumin/therapeutic use , Cytokines/metabolism , Dietary Supplements , Fish Oils/therapeutic use , Gene Expression Regulation , Acute Disease , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chronic Disease , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Curcumin/adverse effects , Cytokines/genetics , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Fish Oils/adverse effects , Gene Expression Profiling , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Irritants/administration & dosage , Irritants/toxicity , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Random Allocation , Survival Analysis
17.
J Nutr Biochem ; 20(8): 649-55, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18926688

ABSTRACT

Inflammatory bowel disease (IBD) patients are at high risk for developing folate deficiency and colon cancer. Since it is difficult to study the subtle global and gene-specific epigenetic mechanisms involved in folate-mediated tumor initiation and promotion, we have generated genetically modified mouse models by targeting the reduced folate carrier (RFC1) and folate-binding protein (Folbp1) genes. The transgenic mice were fed semi-purified diets for 8 weeks containing either normal (2 mg) or deficient (0.1 mg folate/kg diet) levels of folate. Compound heterozygous mice (Folbp1(+/-); RFC1(+/-)) fed an adequate folate diet exhibited a reduction in plasma folate concentrations compared to heterozygous (Folbp1(+/-)) and littermate wild-type mice (P<.05). In contrast, no differences were observed in colonic mucosa. Consumption of a low folate diet significantly reduced (three- to fourfold) plasma and tissue folate levels in all animal models, although plasma homocysteine levels were not altered. In order to elucidate the relationship between folate status and inflammation-associated colon cancer, animals were injected with azoxymethane followed by dextran sodium sulphate treatment in the drinking water. Mice were fed a normal folate diet and were terminated 5 weeks after carcinogen injection. The number of high multiplicity aberrant crypt foci per centimeter of colon was significantly elevated (P<.05) in compound Folbp1(+/-); RFC1(+/-) (3.5+/-0.4) mice as compared to Folbp1(+/-) (1.9+/-0.3) and wild-type control mice (1.1+/-0.1). These data demonstrate that the ablation of two receptor/carrier-mediated pathways for folate transport increases the risk for developing inflammation-associated colon cancer.


Subject(s)
Carrier Proteins/genetics , Colitis/complications , Colonic Neoplasms/etiology , Folic Acid Deficiency/complications , Folic Acid/metabolism , Membrane Transport Proteins/genetics , Precancerous Conditions/etiology , Receptors, Cell Surface/genetics , Animals , Azoxymethane/toxicity , Biomarkers , Brain Chemistry , Carcinogens/toxicity , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Folate Receptors, GPI-Anchored , Folic Acid/analysis , Folic Acid/blood , Folic Acid Deficiency/metabolism , Folic Acid Deficiency/pathology , Food, Formulated , Heterozygote , Homocysteine/blood , Intestinal Mucosa/chemistry , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Models, Animal , Nutritional Status , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Reduced Folate Carrier Protein
18.
Am J Physiol Gastrointest Liver Physiol ; 293(5): G935-43, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17717041

ABSTRACT

Butyrate, a short-chain fatty acid fiber fermentation product, induces colonocyte apoptosis in part via a Fas-mediated (extrinsic) pathway. In previous studies, we demonstrated that docosahexaenoic acid (DHA, 22:6(Delta4,7,10,13,16,19)) enhances the effect of butyrate by increasing mitochondrial lipid oxidation and mitochondrial Ca(2+)-dependent apoptosis in the colon. In this study, we further examined the mechanism of DHA-butyrate synergism in 1) human colon tumor (HCT-116 isogenic p53+/+ vs. p53-/-) cells and 2) primary cultures of rat colonic crypts. Herein, we show that DHA and butyrate promote apoptosis by enhancing mitochondrial Ca(2+) accumulation in both isogenic cell lines. Ca(2+) accumulation and apoptosis were inhibited by blockade of mitochondrial uniporter-mediated Ca(2+) uptake. In addition, Mito-Q, a mitochondria-targeted antioxidant, also blocked apoptosis induced by DHA and butyrate. In complementary experiments, rats were fed diets supplemented with either corn oil (control, contains no DHA) or fish oil (contains DHA). Colonic crypts were isolated and incubated with or without butyrate, after which the mitochondria-to-cytosol Ca(2+) ratio and crypt viability were measured. No significant difference (P > 0.05) in basal mitochondrial Ca(2+) levels was observed between fish oil- or corn oil-fed animals. In contrast, when fish oil was the dietary lipid source, crypts incubated with butyrate exhibited a significant increase (3.6-fold, P < 0.001) in mitochondrial Ca(2+) compared with corn oil plus butyrate treatment. On the basis of these data, we propose that the combination of DHA and butyrate compared with butyrate alone further enhances colonocyte apoptosis by inducing a p53-independent, oxidation-sensitive, mitochondrial Ca(2+) -dependent (intrinsic) pathway.


Subject(s)
Apoptosis/physiology , Butyrates/pharmacology , Colon/physiology , Docosahexaenoic Acids/pharmacology , Intestinal Mucosa/physiology , Mitochondria/physiology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/drug effects , Calcium/physiology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Colon/cytology , Colon/drug effects , Colonic Neoplasms , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Lipid Peroxidation/drug effects , Male , Mitochondria/drug effects , Rats , Rats, Sprague-Dawley , Ruthenium Compounds/pharmacology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
19.
Cancer Res ; 65(3): 887-97, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15705887

ABSTRACT

Low dietary folate intake is associated with an increased risk for colon cancer; however, relevant genetic animal models are lacking. We therefore investigated the effect of targeted ablation of two folate transport genes, folate binding protein 1 (Folbp1) and reduced folate carrier 1 (RFC1), on folate homeostasis to elucidate the molecular mechanisms of folate action on colonocyte cell proliferation, gene expression, and colon carcinogenesis. Targeted deletion of Folbp1 (Folbp1(+/-) and Folbp1(-/-)) significantly reduced (P < 0.05) colonic Folbp1 mRNA, colonic mucosa, and plasma folate concentration. In contrast, subtle changes in folate homeostasis resulted from targeted deletion of RFC1 (RFC1(+/-)). These animals had reduced (P < 0.05) colonic RFC1 mRNA and exhibited a 2-fold reduction in the plasma S-adenosylmethionine/S-adenosylhomocysteine. Folbp1(+/-) and Folbp1(-/-) mice had larger crypts expressed as greater (P < 0.05) numbers of cells per crypt column relative to Folbp1(+/+) mice. Colonic cell proliferation was increased in RFC1(+/-) mice relative to RFC1(+/+) mice. Microarray analysis of colonic mucosa showed distinct changes in gene expression specific to Folbp1 or RFC1 ablation. The effect of folate transporter gene ablation on colon carcinogenesis was evaluated 8 and 38 weeks post-azoxymethane injection in wild-type and heterozygous mice. Relative to RFC1(+/+) mice, RFC1(+/-) mice developed increased (P < 0.05) numbers of aberrant crypt foci at 8 weeks. At 38 weeks, RFC1(+/-) mice developed local inflammatory lesions with or without epithelial dysplasia as well as adenocarcinomas, which were larger relative to RFC1(+/+) mice. In contrast, Folbp1(+/-) mice developed 4-fold (P < 0.05) more lesions relative to Folbp1(+/+) mice. In conclusion, Folbp1 and RFC1 genetically modified mice exhibit distinct changes in colonocyte phenotype and therefore have utility as models to examine the role of folate homeostasis in colon cancer development.


Subject(s)
Carrier Proteins/genetics , Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Membrane Transport Proteins/genetics , Receptors, Cell Surface/genetics , Animals , Azoxymethane , Carcinogens , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Cell Cycle/genetics , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Colon/metabolism , Colon/physiology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Folate Receptors, GPI-Anchored , Gene Expression Profiling , Gene Silencing , Genetic Predisposition to Disease , Kidney/metabolism , Kidney/physiology , Male , Membrane Transport Modulators , Membrane Transport Proteins/antagonists & inhibitors , Membrane Transport Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , Reduced Folate Carrier Protein , Reverse Transcriptase Polymerase Chain Reaction , S-Adenosylhomocysteine/blood , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/blood , S-Adenosylmethionine/metabolism
20.
Cancer Res ; 64(18): 6797-804, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374999

ABSTRACT

The mechanisms by which n-3 polyunsaturated fatty acids (PUFAs) decrease colon tumor formation have not been fully elucidated. Examination of genes up- or down-regulated at various stages of tumor development via the monitoring of gene expression relationships will help to determine the biological processes ultimately responsible for the protective effects of n-3 PUFA. Therefore, using a 3 x 2 x 2 factorial design, we used Codelink DNA microarrays containing approximately 9000 genes to help decipher the global changes in colonocyte gene expression profiles in carcinogen-injected Sprague Dawley rats. Animals were assigned to three dietary treatments differing only in the type of fat (corn oil/n-6 PUFA, fish oil/n-3 PUFA, or olive oil/n-9 monounsaturated fatty acid), two treatments (injection with the carcinogen azoxymethane or with saline), and two time points (12 hours and 10 weeks after first injection). Only the consumption of n-3 PUFA exerted a protective effect at the initiation (DNA adduct formation) and promotional (aberrant crypt foci) stages. Importantly, microarray analysis of colonocyte gene expression profiles discerned fundamental differences among animals treated with n-3 PUFA at both the 12 hours and 10-week time points. Thus, in addition to demonstrating that dietary fat composition alters the molecular portrait of gene expression profiles in the colonic epithelium at both the initiation and promotional stages of tumor development, these findings indicate that the chemopreventive effect of fish oil is due to the direct action of n-3 PUFA and not to a reduction in the content of n-6 PUFA.


Subject(s)
Anticarcinogenic Agents/pharmacology , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Fatty Acids, Unsaturated/pharmacology , Triglycerides/pharmacology , Animals , Apoptosis/drug effects , Azoxymethane , Carcinogens , Cell Differentiation/drug effects , Colonic Neoplasms/metabolism , DNA Adducts/antagonists & inhibitors , DNA Adducts/biosynthesis , Disease Progression , Down-Regulation/drug effects , Eating/drug effects , Fatty Acids, Omega-3 , Fatty Acids, Omega-6/pharmacology , Fish Oils/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Male , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...