Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Publication year range
1.
Cell ; 178(5): 1115-1131.e15, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442404

ABSTRACT

Little is known about how metabolites couple tissue-specific stem cell function with physiology. Here we show that, in the mammalian small intestine, the expression of Hmgcs2 (3-hydroxy-3-methylglutaryl-CoA synthetase 2), the gene encoding the rate-limiting enzyme in the production of ketone bodies, including beta-hydroxybutyrate (ßOHB), distinguishes self-renewing Lgr5+ stem cells (ISCs) from differentiated cell types. Hmgcs2 loss depletes ßOHB levels in Lgr5+ ISCs and skews their differentiation toward secretory cell fates, which can be rescued by exogenous ßOHB and class I histone deacetylase (HDAC) inhibitor treatment. Mechanistically, ßOHB acts by inhibiting HDACs to reinforce Notch signaling, instructing ISC self-renewal and lineage decisions. Notably, although a high-fat ketogenic diet elevates ISC function and post-injury regeneration through ßOHB-mediated Notch signaling, a glucose-supplemented diet has the opposite effects. These findings reveal how control of ßOHB-activated signaling in ISCs by diet helps to fine-tune stem cell adaptation in homeostasis and injury.


Subject(s)
Diet, High-Fat , Ketone Bodies/metabolism , Stem Cells/metabolism , 3-Hydroxybutyric Acid/blood , 3-Hydroxybutyric Acid/pharmacology , Aged, 80 and over , Animals , Cell Differentiation/drug effects , Cell Self Renewal , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxymethylglutaryl-CoA Synthase/deficiency , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Intestines/cytology , Intestines/pathology , Male , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Stem Cells/cytology , Young Adult
2.
Cell ; 175(5): 1307-1320.e22, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30392957

ABSTRACT

In the small intestine, a niche of accessory cell types supports the generation of mature epithelial cell types from intestinal stem cells (ISCs). It is unclear, however, if and how immune cells in the niche affect ISC fate or the balance between self-renewal and differentiation. Here, we use single-cell RNA sequencing (scRNA-seq) to identify MHC class II (MHCII) machinery enrichment in two subsets of Lgr5+ ISCs. We show that MHCII+ Lgr5+ ISCs are non-conventional antigen-presenting cells in co-cultures with CD4+ T helper (Th) cells. Stimulation of intestinal organoids with key Th cytokines affects Lgr5+ ISC renewal and differentiation in opposing ways: pro-inflammatory signals promote differentiation, while regulatory cells and cytokines reduce it. In vivo genetic perturbation of Th cells or MHCII expression on Lgr5+ ISCs impacts epithelial cell differentiation and IEC fate during infection. These interactions between Th cells and Lgr5+ ISCs, thus, orchestrate tissue-wide responses to external signals.


Subject(s)
Cell Differentiation , Cell Self Renewal , Interleukin-10/metabolism , Stem Cells/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Cytokines/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Histocompatibility Antigens Class II/metabolism , Immune System/metabolism , Intestines/cytology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Receptors, G-Protein-Coupled/metabolism , Salmonella enterica/pathogenicity , Stem Cells/metabolism , T-Lymphocytes, Helper-Inducer/cytology
3.
Cell ; 168(5): 775-788.e12, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235195

ABSTRACT

Stem-cell-based therapies can potentially reverse organ dysfunction and diseases, but the removal of impaired tissue and activation of a program leading to organ regeneration pose major challenges. In mice, a 4-day fasting mimicking diet (FMD) induces a stepwise expression of Sox17 and Pdx-1, followed by Ngn3-driven generation of insulin-producing ß cells, resembling that observed during pancreatic development. FMD cycles restore insulin secretion and glucose homeostasis in both type 2 and type 1 diabetes mouse models. In human type 1 diabetes pancreatic islets, fasting conditions reduce PKA and mTOR activity and induce Sox2 and Ngn3 expression and insulin production. The effects of the FMD are reversed by IGF-1 treatment and recapitulated by PKA and mTOR inhibition. These results indicate that a FMD promotes the reprogramming of pancreatic cells to restore insulin generation in islets from T1D patients and reverse both T1D and T2D phenotypes in mouse models. PAPERCLIP.


Subject(s)
Diabetes Mellitus, Type 1/diet therapy , Diabetes Mellitus, Type 2/diet therapy , Fasting , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diet , Glucose Tolerance Test , Humans , In Vitro Techniques , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans , Mice , Nerve Tissue Proteins/genetics , Pancreas/cytology , Pancreas/metabolism , Signal Transduction , Transcriptome
4.
Nature ; 627(8004): 636-645, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418875

ABSTRACT

A hallmark of cancer is the avoidance of immune destruction. This process has been primarily investigated in locally advanced or metastatic cancer1-3; however, much less is known about how pre-malignant or early invasive tumours evade immune detection. Here, to understand this process in early colorectal cancers (CRCs), we investigated how naive colon cancer organoids that were engineered in vitro to harbour Apc-null, KrasG12D and Trp53-null (AKP) mutations adapted to the in vivo native colonic environment. Comprehensive transcriptomic and chromatin analyses revealed that the endoderm-specifying transcription factor SOX17 became strongly upregulated in vivo. Notably, whereas SOX17 loss did not affect AKP organoid propagation in vitro, its loss markedly reduced the ability of AKP tumours to persist in vivo. The small fraction of SOX17-null tumours that grew displayed notable interferon-γ (IFNγ)-producing effector-like CD8+ T cell infiltrates in contrast to the immune-suppressive microenvironment in wild-type counterparts. Mechanistically, in both endogenous Apc-null pre-malignant adenomas and transplanted organoid-derived AKP CRCs, SOX17 suppresses the ability of tumour cells to sense and respond to IFNγ, preventing anti-tumour T cell responses. Finally, SOX17 engages a fetal intestinal programme that drives differentiation away from LGR5+ tumour cells to produce immune-evasive LGR5- tumour cells with lower expression of major histocompatibility complex class I (MHC-I). We propose that SOX17 is a transcription factor that is engaged during the early steps of colon cancer to orchestrate an immune-evasive programme that permits CRC initiation and progression.


Subject(s)
Adenoma , Colorectal Neoplasms , Immune Evasion , SOXF Transcription Factors , Animals , Humans , Mice , Adenoma/immunology , Adenoma/pathology , CD8-Positive T-Lymphocytes/immunology , Chromatin/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Gene Expression Profiling , Interferon-gamma/immunology , Organoids/immunology , Organoids/pathology , SOXF Transcription Factors/metabolism , Tumor Microenvironment/immunology , Mutation , Endoderm/metabolism , Disease Progression
5.
Nat Immunol ; 18(2): 184-195, 2017 02.
Article in English | MEDLINE | ID: mdl-27992400

ABSTRACT

Invariant natural killer T cells (iNKT cells) are innate-like lymphocytes that protect against infection, autoimmune disease and cancer. However, little is known about the epigenetic regulation of iNKT cell development. Here we found that the H3K27me3 histone demethylase UTX was an essential cell-intrinsic factor that controlled an iNKT-cell lineage-specific gene-expression program and epigenetic landscape in a demethylase-activity-dependent manner. UTX-deficient iNKT cells exhibited impaired expression of iNKT cell signature genes due to a decrease in activation-associated H3K4me3 marks and an increase in repressive H3K27me3 marks within the promoters occupied by UTX. We found that JunB regulated iNKT cell development and that the expression of genes that were targets of both JunB and the iNKT cell master transcription factor PLZF was UTX dependent. We identified iNKT cell super-enhancers and demonstrated that UTX-mediated regulation of super-enhancer accessibility was a key mechanism for commitment to the iNKT cell lineage. Our findings reveal how UTX regulates the development of iNKT cells through multiple epigenetic mechanisms.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation , Histone Demethylases/metabolism , Natural Killer T-Cells/physiology , Animals , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Histone Demethylases/genetics , Immunity, Innate/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Genes Dev ; 33(17-18): 1236-1251, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31416966

ABSTRACT

Tumors display increased uptake and processing of nutrients to fulfill the demands of rapidly proliferating cancer cells. Seminal studies have shown that the proto-oncogene MYC promotes metabolic reprogramming by altering glutamine uptake and metabolism in cancer cells. How MYC regulates the metabolism of other amino acids in cancer is not fully understood. Using high-performance liquid chromatography (HPLC)-tandem mass spectrometry (LC-MS/MS), we found that MYC increased intracellular levels of tryptophan and tryptophan metabolites in the kynurenine pathway. MYC induced the expression of the tryptophan transporters SLC7A5 and SLC1A5 and the enzyme arylformamidase (AFMID), involved in the conversion of tryptophan into kynurenine. SLC7A5, SLC1A5, and AFMID were elevated in colon cancer cells and tissues, and kynurenine was significantly greater in tumor samples than in the respective adjacent normal tissue from patients with colon cancer. Compared with normal human colonic epithelial cells, colon cancer cells were more sensitive to the depletion of tryptophan. Blocking enzymes in the kynurenine pathway caused preferential death of established colon cancer cells and transformed colonic organoids. We found that only kynurenine and no other tryptophan metabolite promotes the nuclear translocation of the transcription factor aryl hydrocarbon receptor (AHR). Blocking the interaction between AHR and kynurenine with CH223191 reduced the proliferation of colon cancer cells. Therefore, we propose that limiting cellular kynurenine or its downstream targets could present a new strategy to reduce the proliferation of MYC-dependent cancer cells.


Subject(s)
Colonic Neoplasms/physiopathology , Kynurenine/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tryptophan/metabolism , Amino Acid Transport System ASC/genetics , Antineoplastic Agents/pharmacology , Arylformamidase/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Indoles/pharmacology , Kynurenine/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Minor Histocompatibility Antigens/genetics , Oximes/pharmacology , Proto-Oncogene Mas , Sulfonamides/pharmacology
7.
Nature ; 571(7765): 398-402, 2019 07.
Article in English | MEDLINE | ID: mdl-31292548

ABSTRACT

A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche1,2. Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor α (PPAR-α)3, and lowered PPAR-α activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.


Subject(s)
Aging , Cellular Senescence , Esterases/metabolism , Intestinal Mucosa/pathology , Paneth Cells/metabolism , Regeneration , Aging/physiology , Animals , Cellular Senescence/physiology , Esterases/antagonists & inhibitors , Esterases/biosynthesis , Female , Humans , Intestinal Mucosa/physiology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , PPAR alpha/metabolism , Paneth Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Stem Cell Niche , Stem Cells/pathology , Wnt Proteins/antagonists & inhibitors , Wnt Signaling Pathway
8.
Gastroenterology ; 164(7): 1137-1151.e15, 2023 06.
Article in English | MEDLINE | ID: mdl-36871599

ABSTRACT

BACKGROUND & AIMS: Fibrosis and tissue stiffening are hallmarks of inflammatory bowel disease (IBD). We have hypothesized that the increased stiffness directly contributes to the dysregulation of the epithelial cell homeostasis in IBD. Here, we aim to determine the impact of tissue stiffening on the fate and function of the intestinal stem cells (ISCs). METHODS: We developed a long-term culture system consisting of 2.5-dimensional intestinal organoids grown on a hydrogel matrix with tunable stiffness. Single-cell RNA sequencing provided stiffness-regulated transcriptional signatures of the ISCs and their differentiated progeny. YAP-knockout and YAP-overexpression mice were used to manipulate YAP expression. In addition, we analyzed colon samples from murine colitis models and human IBD samples to assess the impact of stiffness on ISCs in vivo. RESULTS: We demonstrated that increasing the stiffness potently reduced the population of LGR5+ ISCs and KI-67+-proliferating cells. Conversely, cells expressing the stem cell marker, olfactomedin-4, became dominant in the crypt-like compartments and pervaded the villus-like regions. Concomitantly, stiffening prompted the ISCs to preferentially differentiate toward goblet cells. Mechanistically, stiffening increased the expression of cytosolic YAP, driving the extension of olfactomedin-4+ cells into the villus-like regions, while it induced the nuclear translocation of YAP, leading to preferential differentiation of ISCs toward goblet cells. Furthermore, analysis of colon samples from murine colitis models and patients with IBD demonstrated cellular and molecular remodeling reminiscent of those observed in vitro. CONCLUSIONS: Collectively, our findings highlight that matrix stiffness potently regulates the stemness of ISCs and their differentiation trajectory, supporting the hypothesis that fibrosis-induced gut stiffening plays a direct role in epithelial remodeling in IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Mice , Animals , Goblet Cells , Stem Cells/physiology , Intestinal Mucosa/metabolism , Cell Differentiation/genetics , Inflammatory Bowel Diseases/metabolism , Colitis/metabolism
9.
Nature ; 560(7717): E26, 2018 08.
Article in English | MEDLINE | ID: mdl-29849139

ABSTRACT

In Fig. 4e of this Article, the labels for 'Control' and 'HFD' were reversed ('Control' should have been labelled blue rather than purple, and 'HFD' should have been labelled purple rather than blue). Similarly, in Fig. 4f of this Article, the labels for 'V' and 'GW' were reversed ('V' should have been labelled blue rather than purple, and 'GW' should have been labelled purple instead of blue). The original figure has been corrected online.

10.
Nature ; 545(7654): 355-359, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28489818

ABSTRACT

The heterogeneity of cellular states in cancer has been linked to drug resistance, cancer progression and the presence of cancer cells with properties of normal tissue stem cells. Secreted Wnt signals maintain stem cells in various epithelial tissues, including in lung development and regeneration. Here we show that mouse and human lung adenocarcinomas display hierarchical features with two distinct subpopulations, one with high Wnt signalling activity and another forming a niche that provides the Wnt ligand. The Wnt responder cells showed increased tumour propagation ability, suggesting that these cells have features of normal tissue stem cells. Genetic perturbation of Wnt production or signalling suppressed tumour progression. Small-molecule inhibitors targeting essential posttranslational modification of Wnt reduced tumour growth and markedly decreased the proliferative potential of lung cancer cells, leading to improved survival of tumour-bearing mice. These results indicate that strategies for disrupting pathways that maintain stem-like and niche cell phenotypes can translate into effective anti-cancer therapies.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Disease Progression , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Stem Cell Niche , Wnt Proteins/biosynthesis , Wnt Signaling Pathway , Adenocarcinoma of Lung , Animals , Cell Proliferation/drug effects , Female , Humans , Male , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/pharmacology , Survival Rate , Wnt Proteins/chemistry , Wnt Proteins/metabolism
11.
Clin Infect Dis ; 74(6): 1081-1084, 2022 03 23.
Article in English | MEDLINE | ID: mdl-34245255

ABSTRACT

The clinical significance of severe acute respiratory syndrome coronavirus 2 (SARS CoV-2) RNA in stool remains uncertain. We found that extrapulmonary dissemination of infection to the gastrointestinal tract, assessed by the presence of SARS-CoV-2 RNA in stool, is associated with decreased coronavirus disease 2019 (COVID-19) survival. Measurement of SARS-CoV-2 RNA in stool may have utility for clinical risk assessment.


Subject(s)
COVID-19 , SARS-CoV-2 , Feces , Gastrointestinal Tract , Humans , RNA, Viral , SARS-CoV-2/genetics
12.
Mod Pathol ; 35(11): 1723-1731, 2022 11.
Article in English | MEDLINE | ID: mdl-35590108

ABSTRACT

Mucinous adenocarcinoma (MAD), the most common subtype of colonic adenocarcinoma (CA), requires >50% intratumoral mucin. There is limited data regarding the impact of MAD on key lymphocyte subsets and therapeutically critical immune elements. In this study we address: (1) the definition of MAD, (2) grading of MAD, and (3) the impact of MAD and extracellular mucin on intratumoral immune milieu. Estimation of the percentage of intratumoral mucin was performed by two pathologists. Tissue microarrays were stained for immune markers including CD8, CD163, PD-L1, FoxP3, ß2 microglobulin, HLA class I, and HLA class II. Immunohistochemistry for BRAF V600E was performed. MMR status was determined on immunohistochemistry for MSH2, MSH6, MLH1, PMS2. Manual and automated HALO platforms were used for quantification. The 903 CAs included 62 (6.9%) MAD and 841 CA with ≤ 50% mucin. We identified 225 CAs with mucinous differentiation, defined by ≥10% mucin. On univariate analysis neither cut point, 50% (p = 0.08) and 10% (p = 0.08) mucin, correlated with disease-specific survival (DSS). There were no differences in key clinical, histological and molecular features between MAD and CA with mucinous differentiation. On univariate analysis of patients with MAD, tumor grade correlated with DSS (p = 0.0001) while MMR status did not (p = 0.86). There was no statistically significant difference in CD8 (P = 0.17) and CD163 (P = 0.05) positive immune cells between MAD and conventional CA. However, deficient (d) MMR MADs showed fewer CD8 (P = 0.0001), CD163 (P = 0.0001) and PD-L1 (P = 0.003) positive immune cells compared to proficient (p)MMR MADs, a finding also seen with at 10% mucin cut point. Although MAD does not impact DSS, this study raises the possibility that the immune milieu of dMMR MADs and tumors with > =10% mucin may differ from pMMR MADs and tumors with <10% mucin, a finding that may impact immune-oncology based therapeutics.


Subject(s)
Adenocarcinoma, Mucinous , Colonic Neoplasms , Colorectal Neoplasms , Humans , DNA Mismatch Repair , B7-H1 Antigen/genetics , MutS Homolog 2 Protein/genetics , Mismatch Repair Endonuclease PMS2/genetics , Proto-Oncogene Proteins B-raf/genetics , Adenocarcinoma, Mucinous/genetics , Colonic Neoplasms/pathology , Biomarkers , Forkhead Transcription Factors , Mucins , Colorectal Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis
13.
Histopathology ; 81(3): 380-388, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35789111

ABSTRACT

BACKGROUND: Serrated adenocarcinoma (SAC), a recognised WHO variant of colonic adenocarcinoma, is the purported end-product of serrated neoplasia. However, the diagnosis of SAC is infrequently rendered, and little is known about its prognosis, immune microenvironment and molecular alterations. MATERIALS AND METHODS: We assessed 903 consecutive colon carcinomas and recognised tumours with ≥ 5% (n = 77) serrated and ≥ 50% serrated patterns (n = 13). We assessed precursor polyps and synchronous polyps. We recorded demographic/clinical parameters, histological features and mismatch repair (MMR) status. We performed immunohistochemistry and quantification on tissue microarray for HLA class I/II proteins, B2MG, CD8, CD163, LAG3, FoxP3, PD-L1 and BRAF V600E. RESULTS: We identified ≥ 5% epithelial serration prevalence in 8.5% of cases and ≥ 50% epithelial serration prevalence in 1.4% of cases. Precursor lesions were present in 21.4% of cases; these were mostly tubular adenomas with two traditional serrated adenomas identified. SAC with ≥ 5% serrations exhibited lower numbers of CD8-positive lymphocytes (P = 0.002) and lower B2MG expression (P = 0.048), although neither value was significant at ≥ 50% serration threshold. There was no difference in HLA class I/II, or PD-L1 expression on tumour cells and no difference in PD-L1, LAG3, FoxP3 and CD163 expression on immune cells. There was no association with MMR status, or BRAFV600E relative to conventional adenocarcinoma. There was improved disease-specific survival on univariate (but not multivariate) analysis between carcinomas with serrated pattern and non-mucinous conventional colonic carcinomas at ≥ 5% epithelial serrations (P = 0.04). CONCLUSION: SAC category shows a limited impact on survival, and this phenotype may harbour a unique immunological milieu.


Subject(s)
Adenocarcinoma , Adenoma , Carcinoma , Colonic Neoplasms , Colonic Polyps , Colorectal Neoplasms , Adenocarcinoma/pathology , Adenoma/pathology , B7-H1 Antigen/genetics , Biomarkers, Tumor/analysis , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Forkhead Transcription Factors , Humans , Proto-Oncogene Proteins B-raf/genetics , Tumor Microenvironment
14.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26935695

ABSTRACT

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/pathology , Diet, High-Fat/adverse effects , Intestines/pathology , Stem Cells/drug effects , Stem Cells/pathology , Animals , Cell Count , Cell Self Renewal/drug effects , Female , Genes, APC , Humans , Male , Mice , Obesity/chemically induced , Obesity/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , PPAR delta/metabolism , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Stem Cells/metabolism , beta Catenin/metabolism
15.
J Nutr ; 151(12): 3678-3688, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34590119

ABSTRACT

BACKGROUND: Obesity increases the colorectal cancer risk, in part by elevating colonic proinflammatory cytokines. Curcumin (CUR) and supplemental vitamin B-6 each suppress colonic inflammation. OBJECTIVES: We examined whether the combination of CUR and vitamin B-6 amplifies each supplement's effects and thereby suppress obesity-promoted tumorigenesis. METHODS: Male Friend Virus B (FVB) mice (4-week-old; n = 110) received 6 weekly injections of azoxymethane beginning 1 week after arrival. Thereafter, they were randomized to receive a low-fat diet (10% energy from fat), a high-fat diet (HFD; 60% energy from fat), a HFD containing 0.2% CUR, a HFD containing supplemental vitamin B-6 (24 mg pyridoxine HCl/kg), or a HFD containing both CUR and supplemental vitamin B-6 (C + B) for 15 weeks. Colonic inflammation, assessed by fecal calprotectin, and tumor metrics were the primary endpoints. The anti-inflammatory efficacy of the combination was also determined in human colonic organoids. RESULTS: HFD-induced obesity produced a 2.6-fold increase in plasma IL-6 (P < 0.02), a 1.9-fold increase in fecal calprotectin (P < 0.05), and a 2.2-fold increase in tumor multiplicity (P < 0.05). Compared to the HFD group, the C + B combination, but not the individual agents, decreased fecal calprotectin (66%; P < 0.01) and reduced tumor multiplicity and the total tumor burden by 60%-80% (P < 0.03) in an additive fashion. The combination of C + B also significantly downregulated colonic phosphatidylinositol-4,5-bisphosphate 3-kinase, Wnt, and NF-κB signaling by 31%-47% (P < 0.05), effects largely absent with the single agents. Observations that may explain how the 2 agents work additively include a 2.8-fold increased colonic concentration of 3-hydroxyanthranillic acid (P < 0.05) and a 1.3-fold higher colonic concentration of the active coenzymatic form of vitamin B-6 (P < 0.05). In human colonic organoids, micromolar concentrations of CUR, vitamin B-6, and their combination suppressed secreted proinflammatory cytokines by 41%-93% (P < 0.03), demonstrating relevance to humans. CONCLUSIONS: In this mouse model, C + B is superior to either agent alone in preventing obesity-promoted colorectal carcinogenesis. Augmented suppression of procancerous signaling pathways may be the means by which this augmentation occurs.


Subject(s)
Colorectal Neoplasms , Curcumin , Animals , Male , Mice , Carcinogenesis , Colorectal Neoplasms/etiology , Colorectal Neoplasms/prevention & control , Curcumin/pharmacology , Diet, High-Fat , Dietary Supplements , Mice, Inbred C57BL , Obesity/drug therapy , Pyridoxine , Vitamin B 6/pharmacology , Vitamins
16.
Mol Syst Biol ; 15(8): e8849, 2019 08.
Article in English | MEDLINE | ID: mdl-31464373

ABSTRACT

Obesity-associated type 2 diabetes and accompanying diseases have developed into a leading human health risk across industrialized and developing countries. The complex molecular underpinnings of how lipid overload and lipid metabolites lead to the deregulation of metabolic processes are incompletely understood. We assessed hepatic post-translational alterations in response to treatment of cells with saturated and unsaturated free fatty acids and the consumption of a high-fat diet by mice. These data revealed widespread tyrosine phosphorylation changes affecting a large number of enzymes involved in metabolic processes as well as canonical receptor-mediated signal transduction networks. Targeting two of the most prominently affected molecular features in our data, SRC-family kinase activity and elevated reactive oxygen species, significantly abrogated the effects of saturated fat exposure in vitro and high-fat diet in vivo. In summary, we present a comprehensive view of diet-induced alterations of tyrosine signaling networks, including proteins involved in fundamental metabolic pathways.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Liver/drug effects , Obesity/metabolism , Phosphotyrosine/metabolism , Protein Processing, Post-Translational , Animals , Cell Line, Tumor , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Fatty Acids/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/etiology , Obesity/genetics , Obesity/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Rats , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction , src-Family Kinases/genetics , src-Family Kinases/metabolism
17.
Gut ; 68(4): 684-692, 2019 04.
Article in English | MEDLINE | ID: mdl-29666172

ABSTRACT

OBJECTIVE: Serrated colorectal cancer (CRC) accounts for approximately 25% of cases and includes tumours that are among the most treatment resistant and with worst outcomes. This CRC subtype is associated with activating mutations in the mitogen-activated kinase pathway gene, BRAF, and epigenetic modifications termed the CpG Island Methylator Phenotype, leading to epigenetic silencing of key tumour suppressor genes. It is still not clear which (epi-)genetic changes are most important in neoplastic progression and we begin to address this knowledge gap herein. DESIGN: We use organoid culture combined with CRISPR/Cas9 genome engineering to sequentially introduce genetic alterations associated with serrated CRC and which regulate the stem cell niche, senescence and DNA mismatch repair. RESULTS: Targeted biallelic gene alterations were verified by DNA sequencing. Organoid growth in the absence of niche factors was assessed, as well as analysis of downstream molecular pathway activity. Orthotopic engraftment of complex organoid lines, but not BrafV600E alone, quickly generated adenocarcinoma in vivo with serrated features consistent with human disease. Loss of the essential DNA mismatch repair enzyme, Mlh1, led to microsatellite instability. Sphingolipid metabolism genes are differentially regulated in both our mouse models of serrated CRC and human CRC, with key members of this pathway having prognostic significance in the human setting. CONCLUSION: We generate rapid, complex models of serrated CRC to determine the contribution of specific genetic alterations to carcinogenesis. Analysis of our models alongside patient data has led to the identification of a potential susceptibility for this tumour type.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Organoids/pathology , Proto-Oncogene Proteins B-raf/genetics , Adenocarcinoma/metabolism , Alleles , Colon/metabolism , Colorectal Neoplasms/metabolism , CpG Islands/genetics , DNA Mismatch Repair , DNA Mutational Analysis , Disease Progression , Epigenomics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Models, Genetic , Mutation , Organoids/metabolism , Phenotype , Proto-Oncogene Proteins B-raf/metabolism
18.
Am J Physiol Cell Physiol ; 317(3): C405-C419, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31216420

ABSTRACT

In vitro, cell cultures are essential tools in the study of intestinal function and disease. For the past few decades, monolayer cellular cultures, such as cancer cell lines or immortalized cell lines, have been widely applied in gastrointestinal research. Recently, the development of three-dimensional cultures known as organoids has permitted the growth of normal crypt-villus units that recapitulate many aspects of intestinal physiology. Organoid culturing has also been applied to study gastrointestinal diseases, intestinal-microbe interactions, and colorectal cancer. These models are amenable to CRISPR gene editing and drug treatments, including high-throughput small-molecule testing. Three-dimensional intestinal cultures have been transplanted into mice to develop versatile in vivo models of intestinal disease, particularly cancer. Limitations of currently available organoid models include cost and challenges in modeling nonepithelial intestinal cells, such as immune cells and the microbiota. Here, we describe the development of organoid models of intestinal biology and the applications of organoids for study of the pathophysiology of intestinal diseases and cancer.


Subject(s)
Gastrointestinal Diseases/pathology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiology , Organoids/pathology , Organoids/physiology , Animals , Cells, Cultured , Gastrointestinal Diseases/physiopathology , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/physiopathology , Gastrointestinal Tract/physiopathology , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Intestinal Mucosa/physiopathology , Organoids/physiopathology
19.
J Am Chem Soc ; 141(4): 1537-1545, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30599508

ABSTRACT

Phenanthriplatin, a monofunctional anticancer agent derived from cisplatin, shows significantly more rapid DNA covalent-binding activity compared to its parent complex. To understand the underlying molecular mechanism, we used single-molecule studies with optical tweezers to probe the kinetics of DNA-phenanthriplatin binding as well as DNA binding to several control complexes. The time-dependent extensions of single λ-DNA molecules were monitored at constant applied forces and compound concentrations, followed by rinsing with a compound-free solution. DNA-phenanthriplatin association consisted of fast and reversible DNA lengthening with time constant τ ≈ 10 s, followed by slow and irreversible DNA elongation that reached equilibrium in ∼30 min. In contrast, only reversible fast DNA elongation occured for its stereoisomer  trans-phenanthriplatin, suggesting that the distinct two-rate kinetics of phenanthriplatin is sensitive to the geometric conformation of the complex. Furthermore, no DNA unwinding was observed for pyriplatin, in which the phenanthridine ligand of phenanthriplatin is replaced by the smaller pyridine molecule, indicating that the size of the aromatic group is responsible for the rapid DNA elongation. These findings suggest that the mechanism of binding of phenanthriplatin to DNA involves rapid, partial intercalation of the phenanthridine ring followed by slower substitution of the adjacent chloride ligand by, most likely, the N7 atom of a purine base. The cis isomer affords the proper stereochemistry at the metal center to facilitate essentially irreversible DNA covalent binding, a geometric advantage not afforded by trans-phenanthriplatin. This study demonstrates that reversible DNA intercalation provides a robust transition state that is efficiently converted to an irreversible DNA-Pt bound state.


Subject(s)
DNA/chemistry , Intercalating Agents/chemistry , Organoplatinum Compounds/chemistry , Phenanthridines/chemistry , DNA/metabolism , HCT116 Cells , Humans , Intercalating Agents/metabolism , Intercalating Agents/pharmacology , Molecular Docking Simulation , Nucleic Acid Conformation , Organoplatinum Compounds/metabolism , Organoplatinum Compounds/pharmacology , Phenanthridines/metabolism , Phenanthridines/pharmacology , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL