Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 999
Filter
1.
Int Immunopharmacol ; 140: 112828, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39094359

ABSTRACT

Changes in isocitrate dehydrogenases (IDH) lead to the production of the cancer-causing metabolite 2-hydroxyglutarate, making them a cause of cancer. However, the specific role of IDH in the progression of colon cancer is still not well understood. Our current study provides evidence that IDH2 is significantly increased in colorectal cancer (CRC) cells and actively promotes cell growth in vitro and the development of tumors in vivo. Inhibiting the activity of IDH2, either through genetic silencing or pharmacological inhibition, results in a significant increase in α-ketoglutarate (α-KG), indicating a decrease in the reductive citric acid cycle. The excessive accumulation of α-KG caused by the inactivation of IDH2 obstructs the generation of ATP in mitochondria and promotes the downregulation of HIF-1A, eventually inhibiting glycolysis. This dual metabolic impact results in a reduction in ATP levels and the suppression of tumor growth. Our study reveals a metabolic trait of colorectal cancer cells, which involves the active utilization of glutamine through reductive citric acid cycle metabolism. The data suggests that IDH2 plays a crucial role in this metabolic process and has the potential to be a valuable target for the advancement of treatments for colorectal cancer.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Isocitrate Dehydrogenase , Signal Transduction , Isocitrate Dehydrogenase/metabolism , Isocitrate Dehydrogenase/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Animals , Cell Line, Tumor , Mice , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Ketoglutaric Acids/metabolism , Citric Acid Cycle , Glycolysis , Mice, Nude , Disease Progression , Adenosine Triphosphate/metabolism , Cell Proliferation , Cellular Reprogramming , Mitochondria/metabolism , Intestinal Neoplasms/pathology , Intestinal Neoplasms/metabolism , Metabolic Reprogramming
2.
Crit Rev Oncol Hematol ; 203: 104460, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39153703

ABSTRACT

Neuroendocrine neoplasms (NENs) arise from neuroendocrine cells in a wide variety of organs. One of the most affected disease sites is the gastrointestinal system, which originates the gastro-entero-pancreatic NENs (GEP-NENs), a heterogenous group of malignancies that are rapidly increasing in incidence. These tumors can be functioning, with secretory activity leading to identifiable clinical syndromes, or non-functioning, with no secretory activity but with local symptoms of tumor growth and metastasis. A limitation in biomarkers is a crucial unmet need in non-secretory NEN management, as clinical decision-making is made more difficult by obstacles in tumor classification, prognostic evaluation, assessment of treatment response and surveillance. The objective of this review is to present existing and novel biomarkers for NENs that can function as prognostic factors and monitor disease progression or regression longitudinally, with a special emphasis on innovative research into novel multianalyte biomarkers.


Subject(s)
Biomarkers, Tumor , Neuroendocrine Tumors , Pancreatic Neoplasms , Humans , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/pathology , Intestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/metabolism
3.
Nat Immunol ; 25(9): 1637-1649, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39060651

ABSTRACT

Approximately 25% of cancers are preceded by chronic inflammation that occurs at the site of tumor development. However, whether this multifactorial oncogenic process, which commonly occurs in the intestines, can be initiated by a specific immune cell population is unclear. Here, we show that an intestinal T cell subset, derived from interleukin-17 (IL-17)-producing helper T (TH17) cells, induces the spontaneous transformation of the intestinal epithelium. This subset produces inflammatory cytokines, and its tumorigenic potential is not dependent on IL-17 production but on the transcription factors KLF6 and T-BET and interferon-γ. The development of this cell type is inhibited by transforming growth factor-ß1 (TGFß1) produced by intestinal epithelial cells. TGFß signaling acts on the pretumorigenic TH17 cell subset, preventing its progression to the tumorigenic stage by inhibiting KLF6-dependent T-BET expression. This study therefore identifies an intestinal T cell subset initiating cancer.


Subject(s)
Intestinal Mucosa , Kruppel-Like Factor 6 , T-Box Domain Proteins , Th17 Cells , Animals , Th17 Cells/immunology , Mice , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Kruppel-Like Factor 6/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Signal Transduction/immunology , Mice, Inbred C57BL , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Transforming Growth Factor beta1/metabolism , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Mice, Knockout , Interferon-gamma/metabolism , Interferon-gamma/immunology , Interleukin-17/metabolism , Interleukin-17/immunology , Mice, Transgenic , Proto-Oncogene Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Intestinal Neoplasms/immunology , Intestinal Neoplasms/pathology , Intestinal Neoplasms/metabolism , Humans
4.
PLoS One ; 19(7): e0307757, 2024.
Article in English | MEDLINE | ID: mdl-39074116

ABSTRACT

Feline chronic enteropathies (FCE), include food-responsive-enteropathy (FRE), inflammatory bowel disease (IBD), and low-grade intestinal T-cell lymphoma (LGITL), and are common causes of chronic gastrointestinal signs in cats. Distinguishing between different subgroups of FCE can be challenging due to the frequent overlap of anamnestic, clinical, and laboratory data. While dysregulation in lipid metabolism has been reported in humans and dogs with chronic IBD, similar changes in cats are not yet completely understood. Assessing the fatty acid (FA) profile of red blood cell (RBC) membranes offers a valuable method for evaluating the quantity and quality of structural and functional molecular components in the membranes. Therefore, this study aimed to examine the FA composition of RBC membranes in FCE in comparison to healthy cats (HC). Gas-chromatography was used to quantitatively analyze a cluster of 11 FA, and based on these results, parameters of lipid homeostasis and enzyme activity indexes were calculated. A total of 41 FCE cats (17 FRE, 15 IBD, 9 LGITL) and 43 HC were enrolled. In FCE cats, the values of docosapentaenoic acid (p = 0.0002) and docosahexaenoic acid (p = 0.0246), were significantly higher, resulting in an overall increase in ω-3 polyunsaturated fatty acids (PUFA) (p = 0.006), and that of linoleic acid (p = 0.0026) was significantly lower. Additionally, FCE cats exhibited an increased PUFA balance (p = 0.0019) and Δ6-desaturase index (p = 0.0151), along with a decreased ω-6/ω-3 ratio (p = 0.0019). No differences were observed among cats affected by FRE, IBD and LGITL. Like humans and dogs, the results of this study indicate that FCE cats also display changes in their FA lipid profile at the level of the RBC membrane. The non-invasive analysis of RBC membrane shows promise as a potential tool for gaining a better understanding of lipid imbalances in this disease.


Subject(s)
Cat Diseases , Erythrocyte Membrane , Fatty Acids , Inflammatory Bowel Diseases , Animals , Cats , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/veterinary , Inflammatory Bowel Diseases/blood , Fatty Acids/metabolism , Erythrocyte Membrane/metabolism , Cat Diseases/metabolism , Cat Diseases/blood , Male , Female , Lipidomics/methods , Intestinal Diseases/veterinary , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Lymphoma, T-Cell/veterinary , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/veterinary , Intestinal Neoplasms/pathology
5.
Mol Biol Rep ; 51(1): 704, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824233

ABSTRACT

BACKGROUND: Tumor modeling using organoids holds potential in studies of cancer development, enlightening both the intracellular and extracellular molecular mechanisms behind different cancer types, biobanking, and drug screening. Intestinal organoids can be generated in vitro using a unique type of adult stem cells which are found at the base of crypts and are characterized by their high Lgr5 expression levels. METHODS AND RESULTS: In this study, we successfully established intestinal cancer organoid models by using both the BALB/c derived and mouse embryonic stem cells (mESCs)-derived intestinal organoids. In both cases, carcinogenesis-like model was developed by using azoxymethane (AOM) treatment. Carcinogenesis-like model was verified by H&E staining, immunostaining, relative mRNA expression analysis, and LC/MS analysis. The morphologic analysis demonstrated that the number of generated organoids, the number of crypts, and the intensity of the organoids were significantly augmented in AOM-treated intestinal organoids compared to non-AOM-treated ones. Relative mRNA expression data revealed that there was a significant increase in both Wnt signaling pathway-related genes and pluripotency transcription factors in the AOM-induced intestinal organoids. CONCLUSION: We successfully developed simple carcinogenesis-like models using mESC-based and Lgr5 + stem cell-based intestinal organoids. Intestinal organoid based carcinogenesi models might be used for personalized cancer therapy in the future.


Subject(s)
Azoxymethane , Carcinogenesis , Mouse Embryonic Stem Cells , Organoids , Wnt Signaling Pathway , Animals , Organoids/metabolism , Organoids/pathology , Mice , Azoxymethane/toxicity , Carcinogenesis/pathology , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Mouse Embryonic Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Mice, Inbred BALB C , Intestines/pathology , Intestinal Neoplasms/pathology , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Disease Models, Animal , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology
6.
Biochem Pharmacol ; 228: 116259, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38705538

ABSTRACT

Mounting evidence from preclinical and clinical studies suggests that persistent inflammation functions as a driving force in the journey to cancer. Cyclooxygenase-2 (COX-2) is a key enzyme involved in inflammatory signaling. While being transiently upregulated upon inflammatory stimuli, COX-2 has been found to be consistently overexpressed in human colorectal cancer and several other malignancies. The association between chronic inflammation and cancer has been revisited: cancer can arise when inflammation fails to resolve. Besides its proinflammatory functions, COX-2 also catalyzes the production of pro-resolving as well as anti-inflammatory metabolites from polyunsaturated fatty acids. This may account for the side effects caused by long term use of some COX-2 inhibitory drugs during the cancer chemopreventive trials. This review summarizes the latest findings highlighting the dual functions of COX-2 in the context of its implications in the development, maintenance, and progression of cancer.


Subject(s)
Cyclooxygenase 2 Inhibitors , Cyclooxygenase 2 , Inflammation , Humans , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2 Inhibitors/pharmacology , Animals , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/prevention & control , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Intestinal Neoplasms/prevention & control , Intestinal Neoplasms/metabolism , Chemoprevention/methods , Chemoprevention/trends
7.
Nucl Med Commun ; 45(8): 736-744, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38745508

ABSTRACT

OBJECTIVE: We aimed to compare different segmentation methods used to calculate prognostically valuable volumetric parameters, somatostatin receptor expressing tumor volume (SRETV), and total lesion somatostatin receptor expression (TLSRE), measured by 68 Ga-DOTATATE PET/CT and to find the optimal segmentation method to predict prognosis. PATIENTS AND METHODS: Images of 34 patients diagnosed with gastroenteropancreatic neuroendocrine tumor (GEPNET) who underwent 68 Ga-DOTATATE PET/CT imaging were reanalyzed. Four different threshold-based methods (fixed relative threshold method, normal liver background threshold method, fixed absolute standardized uptake value (SUV) threshold method, and adaptive threshold method) were used to calculate SRETV and TLSRE values. SRETV of all lesions of a patient was summarized as whole body SRETV (WB-SRETV) and TLSRE of all lesions of a patient was computed as whole body TLSRE (WB-TLSRE). RESULTS: WB-SRETVs calculated with all segmentation methods were statistically significantly associated with progression-free survival except WB-SRETV at which was calculated using adaptive threshold method. The fixed relative threshold methods calculated by using 45% (WB-SRETV 45% ) and 60% (WB-SRETV 60% ) of the SUV value as threshold respectively, were found to have statistically significant highest prognostic value (C-index = 0.704, CI = 0.622-0.786, P  = 0.007). Among WB-TLSRE parameters, WB-TLSRE 35% , WB-TLSRE 40% , and WB-TLSRE 50% had the highest prognostic value (C-index = 0.689, CI = 0.604-0.774, P  = 0.008). CONCLUSION: The fixed relative threshold method was found to be the most effective and easily applicable method to measure SRETV on pretreatment 68 Ga-DOTATATE PET/CT to predict prognosis in GEPNET patients. WB-SRETV 45% (cutoff value of 11.8 cm 3 ) and WB-SRETV 60% (cutoff value of 6.3 cm 3 ) were found to be the strongest predictors of prognosis in GEPNET patients.


Subject(s)
Intestinal Neoplasms , Neuroendocrine Tumors , Organometallic Compounds , Pancreatic Neoplasms , Positron Emission Tomography Computed Tomography , Receptors, Somatostatin , Stomach Neoplasms , Tumor Burden , Humans , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Receptors, Somatostatin/metabolism , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Male , Intestinal Neoplasms/diagnostic imaging , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Female , Middle Aged , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Prognosis , Adult , Aged , Image Processing, Computer-Assisted/methods , Retrospective Studies , Aged, 80 and over
8.
J Vet Med Sci ; 86(7): 748-755, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38811188

ABSTRACT

Nuclear expression of ß-catenin has been reported in canine intestinal epithelial tumors (IETs) and colorectal inflammatory polyps (CIPs) with dysplastic epithelia. However, the role of the Wnt/ß-catenin signaling pathway in these lesions remains unclear. To investigate the association between the nuclear ß-catenin expression and the activation of the Wnt/ß-catenin signaling pathway, immunohistochemistry and mutation analyses were conducted on 64 IETs and 20 CIPs. IETs and CIPs with ß-catenin nuclear and/or cytoplasm immunolabeling were classified as ß-catenin (+). The immunostaining scores of c-Myc and Cyclin D1 and Ki-67 index were significantly higher in ß-catenin (+) cases than in ß-catenin (-) cases. Identical APC mutations (p.E154D and p.K155X) were detected in 6/41 ß-catenin (+) IETs; all 6 of IETs with APC mutations were Jack Russell Terriers. CTNNB1 mutations were detected in 29/42 ß-catenin (+) IETs, 3/11 ß-catenin (+) CIPs, and 2/22 ß-catenin (-) IETs, most of which were hotspots associated with human colorectal carcinoma. In one Miniature Dachshund diagnosed with a CIP that subsequently developed into an IET, the same CTNNB1 mutation was detected in both lesions. The immunohistochemical results suggest that cell proliferative activity in ß-catenin (+) cases may be associated with activation of the Wnt/ß-catenin signaling pathway. The mutation analysis results suggest that CTNNB1 mutations may be associated with cytoplasmic ß-catenin accumulation in IET and CIP. Furthermore, the dysplastic epithelium in CIP may progress to IET through the activation of the Wnt/ß-catenin signaling pathway by the CTNNB1 mutation.


Subject(s)
Dog Diseases , Mutation , Wnt Signaling Pathway , beta Catenin , Animals , Dogs , beta Catenin/genetics , beta Catenin/metabolism , Dog Diseases/genetics , Dog Diseases/pathology , Dog Diseases/metabolism , Wnt Signaling Pathway/genetics , Female , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Immunohistochemistry/veterinary , Intestinal Neoplasms/veterinary , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestinal Neoplasms/metabolism
9.
PLoS One ; 19(5): e0302813, 2024.
Article in English | MEDLINE | ID: mdl-38709790

ABSTRACT

Toll-like receptors (TLRs) are pattern recognition receptors of the innate immunity. TLRs are known to mediate both antitumor effects and tumorigenesis. TLRs are abundant in many cancers, but their expression in small bowel neuroendocrine tumors (SB-NETs) is unknown. We aimed to characterize the expression of TLRs 1-9 in SB-NETs and lymph node metastases and evaluate their prognostic relevance. The present study included 125 patients with SB-NETs, of whom 95 had lymph node metastases, from two Finnish hospitals. Tissue samples were stained immunohistochemically for TLR expression, assessed based on cytoplasmic and nucleic staining intensity and percentage of positively stained cells. Statistical methods for survival analysis included Kaplan-Meier method and Cox regression adjusted for confounding factors. Disease-specific survival (DSS) was the primary outcome. TLRs 1-2 and 4-9 were expressed in SB-NETs and lymph node metastases. TLR3 showed no positive staining. In primary SB-NETs, TLRs 1-9 were not associated with survival. For lymph node metastases, high cytoplasmic TLR7 intensity associated with worse DSS compared to low cytoplasmic intensity (26.4% vs. 84.9%, p = 0.028). Adjusted mortality hazard (HR) was 3.90 (95% CI 1.07-14.3). The expression of TLRs 1-6 and 8-9 in lymph node metastases were not associated with survival. SB-NETs and their lymph node metastases express cytoplasmic TLR 1-2 and 4-9 and nucleic TLR5. High TLR7 expression in SB-NET lymph node metastases was associated with worse prognosis. The current research has future perspective, as it can help create base for clinical drug trials to target specific TLRs with agonists or antagonists to treat neuroendocrine tumors.


Subject(s)
Intestinal Neoplasms , Intestine, Small , Neuroendocrine Tumors , Toll-Like Receptors , Humans , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/metabolism , Female , Male , Middle Aged , Prognosis , Aged , Toll-Like Receptors/metabolism , Intestinal Neoplasms/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/mortality , Intestine, Small/pathology , Intestine, Small/metabolism , Lymphatic Metastasis , Adult , Aged, 80 and over , Clinical Relevance
10.
Nutrients ; 16(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38732562

ABSTRACT

Iron supplements are widely consumed. However, excess iron may accelerate intestinal tumorigenesis. To determine the effect of excess iron on intestinal tumor burden and protein expression changes between tumor and normal tissues, ApcMin/+ mice were fed control (adequate) and excess iron (45 and 450 mg iron/kg diet, respectively; n = 9/group) for 10 wk. Tumor burden was measured, and two-dimensional fluorescence difference gel electrophoresis was used to identify differentially expressed proteins in tumor and normal intestinal tissues. There was a significant increase (78.3%; p ≤ 0.05) in intestinal tumor burden (mm2/cm) with excess iron at wk 10. Of 980 analyzed protein spots, 69 differentially expressed (p ≤ 0.05) protein isoforms were identified, representing 55 genes. Of the isoforms, 56 differed (p ≤ 0.05) between tumor vs. normal tissues from the adequate iron group and 23 differed (p ≤ 0.05) between tumors from the adequate vs. excess iron. Differentially expressed proteins include those involved in cell integrity and adaptive response to reactive oxygen species (including, by gene ID: ANPEP, DPP7, ITGB1, PSMA1 HSPA5). Biochemical pathway analysis found that iron supplementation modulated four highly significant (p ≤ 0.05) functional networks. These findings enhance our understanding of interplay between dietary iron and intestinal tumorigenesis and may help develop more specific dietary guidelines regarding trace element intake.


Subject(s)
Dietary Supplements , Disease Models, Animal , Intestinal Neoplasms , Tumor Burden , Animals , Mice , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intestinal Neoplasms/genetics , Iron/metabolism , Iron, Dietary/administration & dosage , Mice, Inbred C57BL , Male , Gene Expression Regulation, Neoplastic/drug effects , Proteomics/methods
11.
Chemosphere ; 359: 142332, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754493

ABSTRACT

Perfluorooctanesulfonic acid (PFOS) is a widely recognized environment pollutant known for its high bioaccumulation potential and a long elimination half-life. Several studies have shown that PFOS can alter multiple biological pathways and negatively affect human health. Considering the direct exposure to the gastrointestinal (GI) tract to environmental pollutants, PFOS can potentially disrupt intestinal homeostasis. However, there is limited knowledge about the effect of PFOS exposure on normal intestinal tissues, and its contribution to GI-associated diseases remains to be determined. In this study, we examined the effect of PFOS exposure on the gene expression profile of intestinal tissues of C57BL/6 mice using RNAseq analysis. We found that PFOS exposure in drinking water significantly downregulates mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), a rate-limiting ketogenic enzyme, in intestinal tissues of mice. We found that diets containing the soluble fibers inulin and pectin, which are known to be protective against PFOS exposure, were ineffective in reversing the downregulation of HMGCS2 expression in vivo. Analysis of intestinal tissues also demonstrated that PFOS exposure leads to upregulation of proteins implicated in colorectal carcinogenesis, including ß-catenin, c-MYC, mTOR and FASN. Consistent with the in vivo results, PFOS exposure leads to downregulation of HMGCS2 in mouse and human normal intestinal organoids in vitro. Furthermore, we show that shRNA-mediated knockdown of HMGCS2 in a human normal intestinal cell line resulted in increased cell proliferation and upregulation of key proliferation-associated proteins such as cyclin D, survivin, ERK1/2 and AKT, along with an increase in lipid accumulation. In summary, our results suggest that PFOS exposure may contribute to pathological changes in normal intestinal cells via downregulation of HMGCS2 expression and upregulation of pro-carcinogenic signaling pathways that may increase the risk of colorectal cancer development.


Subject(s)
Alkanesulfonic Acids , Carcinogenesis , Down-Regulation , Fluorocarbons , Hydroxymethylglutaryl-CoA Synthase , Mice, Inbred C57BL , Animals , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Hydroxymethylglutaryl-CoA Synthase/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Mice , Down-Regulation/drug effects , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Up-Regulation/drug effects , Environmental Pollutants/toxicity , Intestines/drug effects , Humans , Intestinal Mucosa/metabolism
12.
Cell Rep ; 43(4): 113975, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38507411

ABSTRACT

The intestine is a highly metabolic tissue, but the metabolic programs that influence intestinal crypt proliferation, differentiation, and regeneration are still emerging. Here, we investigate how mitochondrial sirtuin 4 (SIRT4) affects intestinal homeostasis. Intestinal SIRT4 loss promotes cell proliferation in the intestine following ionizing radiation (IR). SIRT4 functions as a tumor suppressor in a mouse model of intestinal cancer, and SIRT4 loss drives dysregulated glutamine and nucleotide metabolism in intestinal adenomas. Intestinal organoids lacking SIRT4 display increased proliferation after IR stress, along with increased glutamine uptake and a shift toward de novo nucleotide biosynthesis over salvage pathways. Inhibition of de novo nucleotide biosynthesis diminishes the growth advantage of SIRT4-deficient organoids after IR stress. This work establishes SIRT4 as a modulator of intestinal metabolism and homeostasis in the setting of DNA-damaging stress.


Subject(s)
Cell Proliferation , Intestinal Neoplasms , Intestines , Sirtuins , Animals , Humans , Mice , Glutamine/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intestinal Neoplasms/genetics , Intestines/metabolism , Intestines/pathology , Mice, Inbred C57BL , Mitochondrial Proteins , Nucleotides/metabolism , Organoids/metabolism , Sirtuins/metabolism
13.
Cell Oncol (Dordr) ; 47(4): 1183-1199, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38315283

ABSTRACT

PURPOSE: Microbial dysbiosis is considered as a hallmark of colorectal cancer (CRC). Trimethylamine-N-oxide (TMAO) as a gut microbiota-dependent metabolite has recently been implicated in CRC development. Nevertheless, evidence relating TMAO to intestinal carcinogenesis remains largely unexplored. Herein, we aimed to examine the crucial role of TMAO in CRC progression. METHODS: Apcmin/+ mice were treated with TMAO or sterile PBS for 14 weeks. Intestinal tissues were isolated to evaluate the effects of TMAO on the malignant transformation of intestinal adenoma. The gut microbiota of mouse feces was detected by 16S rRNA sequencing analysis. HCT-116 cells were used to provide further evidence of TMAO on the progression of CRC. RESULTS: TMAO administration increased tumor cell and stem cell proliferation, and decreased apoptosis, accompanied by DNA damage and gut barrier impairment. Gut microbiota analysis revealed that TMAO induced changes in the intestinal microbial community structure, manifested as reduced beneficial bacteria. Mechanistically, TMAO bound to farnesoid X receptor (FXR), thereby inhibiting the FXR-fibroblast growth factor 15 (FGF15) axis and activating the Wnt/ß-catenin signaling pathway, whereas the FXR agonist GW4064 could blunt TMAO-induced Wnt/ß-catenin pathway activation. CONCLUSION: The microbial metabolite TMAO can enhance intestinal carcinogenesis by inhibiting the FXR-FGF15 pathway.


Subject(s)
Carcinogenesis , Gastrointestinal Microbiome , Methylamines , Receptors, Cytoplasmic and Nuclear , Signal Transduction , Wnt Signaling Pathway , Methylamines/metabolism , Methylamines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans , Gastrointestinal Microbiome/drug effects , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Mice , Signal Transduction/drug effects , Wnt Signaling Pathway/drug effects , Mice, Inbred C57BL , HCT116 Cells , Cell Proliferation/drug effects , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/microbiology , Male , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/chemically induced , Apoptosis/drug effects , Fibroblast Growth Factors/metabolism
14.
Endocr Rev ; 45(3): 343-350, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38123518

ABSTRACT

Midgut neuroendocrine neoplasms (NENs) are one of the most common subtypes of NEN, and their incidence is rising globally. Despite being the most frequently diagnosed malignancy of the small intestine, little is known about their underlying molecular biology. Their unusually low mutational burden compared to other solid tumors and the unexplained occurrence of multifocal tumors makes the molecular biology of midgut NENs a particularly fascinating field of research. This review provides an overview of recent advances in the understanding of the interplay of the genetic, epigenetic, and transcriptomic landscape in the development of midgut NENs, a topic that is critical to understanding their biology and improving treatment options and outcomes for patients.


Subject(s)
Intestinal Neoplasms , Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Animals , Epigenesis, Genetic
15.
Am J Physiol Gastrointest Liver Physiol ; 325(6): G508-G517, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37788331

ABSTRACT

High-fat (HF) diets (HFDs) and inflammation are risk factors for colon cancer; however, the underlying mechanisms remain to be fully elucidated. The transcriptional corepressor HDAC3 has recently emerged as a key regulator of intestinal epithelial responses to diet and inflammation with intestinal-specific Hdac3 deletion (Hdac3IKO) in mice increasing fatty acid oxidation genes and the rate of fatty acid oxidation in enterocytes. Hdac3IKO mice are also predisposed to experimentally induced colitis; however, whether this is driven by the intestinal metabolic reprogramming and whether this predisposes these mice to intestinal tumorigenesis is unknown. Herein, we examined the effects of intestinal-specific Hdac3 deletion on colitis-associated intestinal tumorigenesis in mice fed a standard (STD) or HFD. Hdac3IKO mice were highly prone to experimentally induced colitis, which was further enhanced by an HFD. Hdac3 deletion also accelerated intestinal tumor development, specifically when fed an HFD and most notably in the small intestine where lipid absorption is maximal. Expression of proteins involved in fatty acid metabolism and oxidation (SCD1, EHHADH) were elevated in the small intestine of Hdac3IKO mice fed an HFD, and these mice displayed increased levels of lipid peroxidation, DNA damage, and apoptosis in their villi, as well as extensive expansion of the stem cell and progenitor cell compartment. These findings reveal a novel role for Hdac3 in suppressing colitis and intestinal tumorigenesis, particularly in the context of consumption of an HFD, and reveal a potential mechanism by which HFDs may increase intestinal tumorigenesis by increasing fatty acid oxidation, DNA damage, and intestinal epithelial cell turnover.NEW & NOTEWORTHY We reveal a novel role for the transcriptional corepressor Hdac3 in suppressing colitis and intestinal tumorigenesis, particularly in the context of consumption of an HFD, and reveal a potential mechanism by which HFDs may increase intestinal tumorigenesis by increasing fatty acid oxidation, DNA damage, and intestinal epithelial cell turnover. We also identify a unique mouse model for investigating the complex interplay between diet, metabolic reprogramming, and tumor predisposition in the intestinal epithelium.


Subject(s)
Colitis , Intestinal Neoplasms , Animals , Mice , Carcinogenesis/metabolism , Co-Repressor Proteins/metabolism , Colitis/metabolism , Diet, High-Fat , Fatty Acids/metabolism , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Mice, Inbred C57BL
16.
Am J Physiol Gastrointest Liver Physiol ; 325(1): G80-G91, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37158470

ABSTRACT

Regulation of small intestinal epithelial growth by endogenous and environmental factors is critical for intestinal homeostasis and recovery from insults. Depletion of the intestinal microbiome increases epithelial proliferation in small intestinal crypts, similar to the effects observed in animal models of serotonin potentiation. Based on prior evidence that the microbiome modulates serotonin activity, we hypothesized that microbial depletion-induced epithelial proliferation is dependent on host serotonin activity. A mouse model of antibiotic-induced microbial depletion (AIMD) was employed. Serotonin potentiation was achieved through either genetic knockout of the serotonin transporter (SERT) or pharmacological SERT inhibition, and inhibition of serotonin synthesis was achieved with para-chlorophenylalanine. AIMD and serotonin potentiation increased intestinal villus height and crypt proliferation in an additive manner, but the epithelial proliferation observed after AIMD was blocked in the absence of endogenous serotonin. Using Lgr5-EGFP-reporter mice, we evaluated intestinal stem cell (ISC) quantity and proliferation. AIMD increased the number of ISCs per crypt and ISC proliferation compared with controls, and changes in ISC number and proliferation were dependent on the presence of host serotonin. Furthermore, Western blotting demonstrated that AIMD reduced epithelial SERT protein expression compared with controls. In conclusion, host serotonin activity is necessary for microbial depletion-associated changes in villus height and ISC proliferation in crypts, and microbial depletion produces a functional serotonin-potentiated state through reduced SERT protein expression. These findings provide an understanding of how changes to the microbiome contribute to intestinal pathology and can be applied therapeutically.NEW & NOTEWORTHY Antibiotic-induced microbial depletion of the murine small intestine results in a state of potentiated serotonin activity through reduced epithelial expression of the serotonin transporter. Specifically, serotonin-dependent mechanisms lead to increased intestinal surface area and intestinal stem cell proliferation. Furthermore, the absence of endogenous serotonin leads to blunting of small intestinal villi, suggesting that serotonin signaling is required for epithelial homeostasis.


Subject(s)
Intestinal Neoplasms , Serotonin , Mice , Animals , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Intestines , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Intestinal Neoplasms/metabolism , Cell Proliferation
17.
J Gastroenterol ; 58(5): 444-457, 2023 05.
Article in English | MEDLINE | ID: mdl-36739585

ABSTRACT

BACKGROUND: Amino acid transporters play an important role in supplying nutrition to cells and are associated with cell proliferation. L-type amino acid transporter 1 (LAT1) is highly expressed in many types of cancers and promotes tumor growth; however, how LAT1 affects tumor development is not fully understood. METHODS: To investigate the role of LAT1 in intestinal tumorigenesis, mice carrying LAT1 floxed alleles that also expressed Cre recombinase from the promoter of gene encoding Villin were crossed to an ApcMin/+ background (LAT1fl/fl; vil-cre; ApcMin/+), which were subject to analysis; organoids derived from those mice were also analyzed. RESULTS: This study showed that LAT1 was constitutively expressed in normal crypt base cells, and its conditional deletion in the intestinal epithelium resulted in fewer Paneth cells. LAT1 deletion reduced tumor size and number in the small intestine of ApcMin/+ mice. Organoids derived from LAT1-deleted ApcMin/+ intestinal crypts displayed fewer spherical organoids with reduced Wnt/ß-catenin target gene expression, suggesting a low tumor-initiation capacity. Wnt3 expression was decreased in the absence of LAT1 in the intestinal epithelium, suggesting that loss of Paneth cells due to LAT1 deficiency reduced the risk of tumor initiation by decreasing Wnt3 production. CONCLUSIONS: LAT1 affects intestinal tumor development in a cell-extrinsic manner through reduced Wnt3 expression in Paneth cells. Our findings may partly explain how nutrient availability can affect the risk of tumor development in the intestines.


Subject(s)
Adenomatous Polyposis Coli Protein , Amino Acid Transport System y+L , Intestinal Neoplasms , Paneth Cells , Animals , Mice , Cell Transformation, Neoplastic/genetics , Intestinal Mucosa/pathology , Intestinal Neoplasms/metabolism , Intestine, Small/pathology , Intestines , Paneth Cells/metabolism , Paneth Cells/pathology , Adenomatous Polyposis Coli Protein/metabolism , Amino Acid Transport System y+L/metabolism
18.
Histopathology ; 82(4): 541-554, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36507623

ABSTRACT

AIMS: Criteria for the interpretation of digestive system neuroendocrine neoplasms (NENs) continue to evolve. Although there are some literature recommendations regarding workup and diagnosis of these lesions, different practice patterns exist among pathologists when signing out these specimens. The aim of this study was to assess practice trends among pathologists worldwide when reporting these neoplasms. METHODS AND RESULTS: We created an online survey with multiple questions pertaining to digestive NENs. The results were analysed based on type of practice setting, years of sign-out experience, and practice location. Respondents included 384 practicing pathologists: 70% academic, 30% private practice; 63% gastrointestinal (GI) pathology-subspecialised, 37% not; 39% North American, 42% European, 19% others; 45% with ≤10 years in practice; 55% with >10 years. Some question responses were chosen by the majority (e.g. 85% use both mitotic count and Ki67 index for grading NENs, 82% complete a synoptic, and Ki67 stain even for small incidental appendiceal neuroendocrine tumours [NETs], and 96% utilize the diagnosis of grade 3 NET). However, some questions showed varying responses, including counting mitotic figures, Ki67 stain interpretation, and pancreatic grade 3 NEN workup. Pathologists also had some variability in interpreting regional metastatic foci of small bowel NETs and in choosing blocks for Ki67 staining in multifocal lesions. CONCLUSION: There existed scenarios wherein practice patterns varied despite recommendations in the literature, and there were also scenarios lacking clear guidelines wherein pathologists used varying judgement. This survey highlights current key grey areas in digestive system NEN evaluation, leading to variation in practice patterns.


Subject(s)
Digestive System Neoplasms , Ki-67 Antigen , Neuroendocrine Tumors , Humans , Appendiceal Neoplasms/genetics , Appendiceal Neoplasms/metabolism , Appendiceal Neoplasms/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Ki-67 Antigen/analysis , Ki-67 Antigen/genetics , Neoplasm Grading , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Digestive System Neoplasms/genetics , Digestive System Neoplasms/metabolism , Digestive System Neoplasms/pathology
19.
Am J Physiol Gastrointest Liver Physiol ; 324(3): G177-G189, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36537709

ABSTRACT

Small intestinal neuroendocrine tumors (SI-NETs) are serotonin-secreting well-differentiated neuroendocrine tumors of putative enterochromaffin (EC) cell origin. However, EC cell-derived tumorigenesis remains poorly understood. Here, we examined whether the gain of Myc and the loss of RB1 and Trp53 function in EC cells result in SI-NET using tryptophan hydroxylase 1 (TPH1) Cre-ERT2-driven RB1fl Trp53fl MycLSL (RPM) mice. TPH1-Cre-induced gain of Myc and loss of RB1 and Trp53 function resulted in endocrine or neuronal tumors in pancreas, lung, enteric neurons, and brain. Lineage tracing indicated that the cellular origin for these tumors was TPH1-expressing neuroendocrine, neuronal, or their precursor cells in these organs. However, despite that TPH1 is most highly expressed in EC cells of the small intestine, we observed no incidence of EC cell tumors. Instead, the tumor of epithelial cell origin in the intestine was exclusively nonendocrine adenocarcinoma, suggesting dedifferentiation of EC cells into intestinal stem cells (ISCs) as a cellular mechanism. Furthermore, ex vivo organoid studies indicated that loss of functions of Rb1 and Trp53 accelerated dedifferentiation of EC cells that were susceptible to apoptosis with expression of activated MycT58A, suggesting that the rare dedifferentiating cells escaping cell death went on to develop adenocarcinomas. Lineage tracing demonstrated that EC cells in the small intestine were short-lived compared with neuroendocrine or neuronal cells in other organs. In contrast, EC cell-derived ISCs were long-lasting and actively cycling and thus susceptible to transformation. These results suggest that tissue- and cell-specific properties of EC cells such as rapid cell turnover and homeostatic dedifferentiation, affect the fate and rate of tumorigenesis induced by genetic alterations and provide important insights into EC cell-derived tumorigenesis.NEW & NOTEWORTHY Small intestinal neuroendocrine tumors are of putative enterochromaffin (EC) cell origin and are the most common malignancy in the small intestine, followed by adenocarcinoma. However, the tumorigenesis of these tumor types remains poorly understood. The present lineage tracing studies showed that tissue- and cell-specific properties of EC cells such as rapid cell turnover and homeostatic dedifferentiation affect the fate and rate of tumorigenesis induced by genetic alterations toward a rare occurrence of adenocarcinoma.


Subject(s)
Adenocarcinoma , Intestinal Neoplasms , Neuroendocrine Tumors , Mice , Animals , Enterochromaffin Cells/metabolism , Intestine, Small/pathology , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Intestinal Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Adenocarcinoma/genetics
20.
Cell Mol Biol (Noisy-le-grand) ; 69(14): 101-108, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38279469

ABSTRACT

The occurrence and progression of intestinal cancer are complex, multifactorial processes in which tumor stem cells are believed to play a crucial role. An in-depth understanding of their molecular mechanisms holds imperative clinical significance for improving intestinal cancer treatment. HT-29 and HCT116intestinal cancer cell lines were utilized as research models. The experimental group (group E) and control group (group C) were established by transfecting the hnRNPA1 subtype and empty vector, respectively. The expression (EP) levels of hnRNPA1 and changes in Wnt/ß-catenin signaling-related markers were evaluated using techniques such as RNA extraction, reverse transcription reactions, real-time quantitative PCR (RT qPCR), and protein extraction. The EP of tumor stem cell markers CD133 and CD44 was assessed using immunohistochemistry. Additionally, cell invasion assays, scratch assays, and cell counting kit-8 (CCK-8) proliferation assays were conducted. Furthermore, a mouse tumor model was established to observe the growth of tumors in both groups. overexpression (OP) of hnRNPA1 in group E greatly increased the protein EP levels of ß-catenin, Axin2, and Cyclin D1 and exhibited a higher positivity rate for CD133 and CD44. The invasive, migratory, and proliferative abilities of cells in group E were notably enhanced, and tumor growth was observably faster versus group C. OP of hnRNPA1 was closely associated with the activation of Wnt/ß-catenin signaling and promoted the proliferation and invasive capacity of tumor cells. hnRNPA1 played an imperative role in intestinal cancer stem cells.


Subject(s)
Intestinal Neoplasms , beta Catenin , Animals , Mice , beta Catenin/genetics , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Neoplastic Stem Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL