Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Cell Host Microbe ; 32(9): 1519-1535.e7, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39106870

ABSTRACT

Identification of potential bacterial players in colorectal tumorigenesis has been a focus of intense research. Herein, we find that Clostridium symbiosum (C. symbiosum) is selectively enriched in tumor tissues of patients with colorectal cancer (CRC) and associated with higher colorectal adenoma recurrence after endoscopic polypectomy. The tumorigenic effect of C. symbiosum is observed in multiple murine models. Single-cell transcriptome profiling along with functional assays demonstrates that C. symbiosum promotes the proliferation of colonic stem cells and enhances cancer stemness. Mechanistically, C. symbiosum intensifies cellular cholesterol synthesis by producing branched-chain amino acids (BCAAs), which sequentially activates Sonic hedgehog signaling. Low dietary BCAA intake or blockade of cholesterol synthesis by statins could partially abrogate the C. symbiosum-induced cell proliferation in vivo and in vitro. Collectively, we reveal C. symbiosum as a bacterial driver of colorectal tumorigenesis, thus identifying a potential target in CRC prediction, prevention, and treatment.


Subject(s)
Amino Acids, Branched-Chain , Carcinogenesis , Cell Proliferation , Cholesterol , Colorectal Neoplasms , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Cholesterol/metabolism , Animals , Humans , Mice , Amino Acids, Branched-Chain/metabolism , Clostridium/metabolism , Clostridium/genetics , Signal Transduction , Hedgehog Proteins/metabolism , Cell Line, Tumor , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Male , Female
2.
Nat Microbiol ; 9(9): 2292-2307, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39169124

ABSTRACT

Fusobacterium nucleatum can bind to host cells and potentiate intestinal tumorigenesis. Here we used a genome-wide screen to identify an adhesin, RadD, which facilitates the attachment of F. nucleatum to colorectal cancer (CRC) cells in vitro. RadD directly binds to CD147, a receptor overexpressed on CRC cell surfaces, which initiated a PI3K-AKT-NF-κB-MMP9 cascade, subsequently enhancing tumorigenesis in mice. Clinical specimen analysis showed that elevated radD gene levels in CRC tissues correlated positively with activated oncogenic signalling and poor patient outcomes. Finally, blockade of the interaction between RadD and CD147 in mice effectively impaired F. nucleatum attachment and attenuated F. nucleatum-induced oncogenic response. Together, our study provides insights into an oncogenic mechanism driven by F. nucleatum RadD and suggests that the RadD-CD147 interaction could be a potential therapeutic target for CRC.


Subject(s)
Adhesins, Bacterial , Bacterial Adhesion , Basigin , Carcinogenesis , Colorectal Neoplasms , Fusobacterium nucleatum , Fusobacterium nucleatum/pathogenicity , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/physiology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Animals , Humans , Mice , Basigin/metabolism , Basigin/genetics , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Fusobacterium Infections/microbiology , Fusobacterium Infections/complications , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Signal Transduction , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Female
3.
Gut ; 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025494
4.
Gut Microbes ; 16(1): 2333790, 2024.
Article in English | MEDLINE | ID: mdl-38533566

ABSTRACT

Chemotherapy resistance is one of the main reasons for the poor prognosis of colorectal cancer (CRC). Moreover, dysbiosis of gut bacteria was found to be a specific environmental risk factor. In this study, enrichment of F. nucleatum was elucidated to be significantly associated with CRC recurrence after chemotherapy. Functional experiments showed that F. nucleatum could inhibit pyroptosis induced by chemotherapy drugs, thereby inducing chemoresistance. Furthermore, mechanistic investigation demonstrated that F. nucleatum could regulate the Hippo pathway and promote the expression of BCL2, thereby inhibiting the Caspase-3/GSDME pyroptosis-related pathway induced by chemotherapy drugs and mediating CRC cell chemoresistance. Taken together, these results validated the significant roles of F. nucleatum in CRC chemoresistance, which provided an innovative theoretical basis for the clinical diagnosis and therapy of CRC.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Fusobacterium nucleatum/physiology , Colorectal Neoplasms/microbiology , Hippo Signaling Pathway , Drug Resistance, Neoplasm , Pyroptosis , Neoplasm Recurrence, Local
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(2): 166941, 2024 02.
Article in English | MEDLINE | ID: mdl-37926361

ABSTRACT

OBJECTIVE: Branched-chain amino acid (BCAA) metabolism is involved in the development of colorectal cancer (CRC); however, the underlying mechanism remains unclear. Therefore, this study investigates the role of BCAA metabolism in CRC progression. METHODS: Dietary BCAA was administered to both azoxymethane-induced and azoxymethane/dextran sodium sulfate-induced CRC mouse models. The expression of genes related to BCAA metabolism was determined using RNA sequencing. Adjacent tissue samples, obtained from 58 patients with CRC, were subjected to quantitative real-time PCR and immunohistochemical analysis. Moreover, the suppressive role of branched-chain aminotransferase 2 (BCAT2) in cell proliferation, apoptosis, and xenograft mouse models was investigated. Alterations in BCAAs and activation of downstream pathways were also assessed using metabolic analysis and western blotting. RESULTS: High levels of dietary BCAA intake promoted CRC tumorigenesis in chemical-induced CRC and xenograft mouse models. Both the mRNA and protein levels of BCAT2 were decreased in tumor tissues of patients with CRC compared to those in normal tissues. Proliferation assays and xenograft models confirmed the suppressive role of BCAT2 in CRC progression. Furthermore, the accumulation of BCAAs caused by BCAT2 deficiency facilitated the chronic activation of mTORC1, thereby mediating the oncogenic effect of BCAAs. CONCLUSION: BCAT2 deficiency promotes CRC progression through inhibition of BCAAs metabolism and chronic activation of mTORC1.


Subject(s)
Colorectal Neoplasms , Pregnancy Proteins , Humans , Mice , Animals , Amino Acids, Branched-Chain/metabolism , RNA, Messenger , Mechanistic Target of Rapamycin Complex 1 , Azoxymethane , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Transaminases/genetics , Transaminases/metabolism , Pregnancy Proteins/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism
6.
Cell Host Microbe ; 31(5): 781-797.e9, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37130518

ABSTRACT

Immune checkpoint blockade therapy with anti-PD-1 monoclonal antibody (mAb) is a treatment for colorectal cancer (CRC). However, some patients remain unresponsive to PD-1 blockade. The gut microbiota has been linked to immunotherapy resistance through unclear mechanisms. We found that patients with metastatic CRC who fail to respond to immunotherapy had a greater abundance of Fusobacterium nucleatum and increased succinic acid. Fecal microbiota transfer from responders with low F. nucleatum, but not F. nucleatum-high non-responders, conferred sensitivity to anti-PD-1 mAb in mice. Mechanistically, F. nucleatum-derived succinic acid suppressed the cGAS-interferon-ß pathway, consequently dampening the antitumor response by limiting CD8+ T cell trafficking to the tumor microenvironment (TME) in vivo. Treatment with the antibiotic metronidazole reduced intestinal F. nucleatum abundance, thereby decreasing serum succinic acid levels and resensitizing tumors to immunotherapy in vivo. These findings indicate that F. nucleatum and succinic acid induce tumor resistance to immunotherapy, offering insights into microbiota-metabolite-immune crosstalk in CRC.


Subject(s)
Colorectal Neoplasms , Fusobacterium Infections , Animals , Mice , Fusobacterium nucleatum , Colorectal Neoplasms/drug therapy , Succinic Acid , Fusobacterium Infections/microbiology , Immunotherapy , Tumor Microenvironment
7.
Cell Metab ; 35(4): 651-666.e7, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36963394

ABSTRACT

Maladaptation of host-microbiota metabolic interplay plays a critical role in colorectal cancer initiation. Here, through a combination of single-cell transcriptomics, microbiome profiling, metabonomics, and clinical analysis on colorectal adenoma and carcinoma tissues, we demonstrate that host's urea cycle metabolism is significantly activated during colorectal tumorigenesis, accompanied by the absence of beneficial bacteria with ureolytic capacity, such as Bifidobacterium, and the overabundance of pathogenic bacteria lacking ureolytic function. Urea could enter into macrophages, inhibit the binding efficiency of p-STAT1 to SAT1 promotor region, and further skew macrophages toward a pro-tumoral phenotype characterized by the accumulation of polyamines. Treating a murine model using urea cycle inhibitors or Bifidobacterium-based supplements could mitigate urea-mediated tumorigenesis. Collectively, this study highlights the utility of urea cycle inhibitors or therapeutically manipulating microbial composition using probiotics to prevent colorectal cancer.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Microbiota , Mice , Animals , Gastrointestinal Microbiome/physiology , Carcinogenesis , Colorectal Neoplasms/pathology , Cell Transformation, Neoplastic
8.
EMBO Rep ; 24(4): e56325, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36794620

ABSTRACT

The frequency of p53 mutations in colorectal cancer (CRC) is approximately 40-50%. A variety of therapies are being developed to target tumors expressing mutant p53. However, potential therapeutic targets for CRC expressing wild-type p53 are rare. In this study, we show that METTL14 is transcriptionally activated by wild-type p53 and suppresses tumor growth only in p53-wild-type (p53-WT) CRC cells. METTL14 deletion promotes both AOM/DSS and AOM-induced CRC growth in mouse models with the intestinal epithelial cell-specific knockout of METTL14. Additionally, METTL14 restrains aerobic glycolysis in p53-WT CRC, by repressing SLC2A3 and PGAM1 expression via selectively promoting m6 A-YTHDF2-dependent pri-miR-6769b/pri-miR-499a processing. Biosynthetic mature miR-6769b-3p and miR-499a-3p decrease SLC2A3 and PGAM1 levels, respectively, and suppress malignant phenotypes. Clinically, METTL14 only acts as a beneficial prognosis factor for the overall survival of p53-WT CRC patients. These results uncover a new mechanism for METTL14 inactivation in tumors and, most importantly, reveal that the activation of METTL14 is a critical mechanism for p53-dependent cancer growth inhibition, which could be targeted for therapy in p53-WT CRC.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Animals , Mice , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , MicroRNAs/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Front Immunol ; 13: 1018334, 2022.
Article in English | MEDLINE | ID: mdl-36341436

ABSTRACT

Amino acid metabolism is closely related to the occurrence and development of colon adenocarcinoma (COAD). Studies on the relationship between COAD and the expression of amino acid metabolism are still rare. Based on in silico analysis, we used 358 amino acid metabolism-related genes (AAMRGs) to determine the amino acid metabolism characteristics and then classified COAD into two distinct subtypes, namely AA1 and AA2. Then we analyzed the clinical characteristics, somatic mutation landscape, transcriptome profile, metabolism signatures, immune infiltration, and therapy sensitivity of these two subtypes. The AA1 subtype had inferior overall survival and was characterized by lower amino acid metabolic activity, higher tumor mutation burden, and higher immune cell infiltration, while AA2 displayed higher metabolic activity and relatively better survival. Furthermore, the AA1 subtype was likely to benefit from irinotecan in chemotherapy and immune checkpoint blockade therapy including programmed cell death protein-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) immune checkpoint inhibitor but was resistant to targeted therapy cetuximab. The AA2 subtype showed higher sensitivity to 5-fluorouracil and oxaliplatin. To provide perspectives on cell-specific metabolism for further investigation, we explored metabolic activity in different cell types including lymphocytes, mast cells, myeloid cells stromal cells, and epithelial cells via colorectal cancer single-cell data. Additionally, to assist in clinical decision-making and prognosis prediction, a 60-AAMRG-based classifier was generated and validated in an independent cohort.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Humans , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Prognosis , Amino Acids
10.
Nat Commun ; 12(1): 5405, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34518538

ABSTRACT

Tumor cells evade T cell-mediated immunosurveillance via the interaction between programmed death-1 (PD-1) ligand 1 (PD-L1) on tumor cells and PD-1 on T cells. Strategies disrupting PD-1/PD-L1 have shown clinical benefits in various cancers. However, the limited response rate prompts us to investigate the molecular regulation of PD-L1. Here, we identify trafficking protein particle complex subunit 4 (TRAPPC4), a major player in vesicular trafficking, as a crucial PD-L1 regulator. TRAPPC4 interacts with PD-L1 in recycling endosomes, acting as a scaffold between PD-L1 and RAB11, and promoting RAB11-mediated recycling of PD-L1, thus replenishing its distribution on the tumor cell surface. TRAPPC4 depletion leads to a significant reduction of PD-L1 expression in vivo and in vitro. This reduction in PD-L1 facilitates T cell-mediated cytotoxicity. Overexpression of Trappc4 sensitizes tumor cells to checkpoint therapy in murine tumor models, suggesting TRAPPC4 as a therapeutic target to enhance anti-tumor immunity.


Subject(s)
B7-H1 Antigen/immunology , Colorectal Neoplasms/immunology , Gene Expression Regulation, Neoplastic/immunology , Nerve Tissue Proteins/immunology , Vesicular Transport Proteins/immunology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Endosomes/immunology , Endosomes/metabolism , HCT116 Cells , Humans , Intracellular Space/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Protein Transport , RNA Interference , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
11.
Gut ; 70(11): 2123-2137, 2021 11.
Article in English | MEDLINE | ID: mdl-33318144

ABSTRACT

OBJECTIVE: Microbiota disorder promotes chronic inflammation and carcinogenesis. High glycolysis is associated with poor prognosis in patients with colorectal cancer (CRC). However, the potential correlation between the gut microbiota and glucose metabolism is unknown in CRC. DESIGN: 18F-FDG (18F-fluorodeoxyglucose) PET (positron emission tomography)/CT image scanning data and microbiota PCR analysis were performed to measure the correlation between metabolic alterations and microbiota disorder in 33 patients with CRC. Multiple colorectal cancer models, metabolic analysis and Seahorse assay were established to assess the role of long non-coding RNA (lncRNA) enolase1-intronic transcript 1 (ENO1-IT1) in Fusobacterium (F.) nucleatum-induced glucose metabolism and colorectal carcinogenesis. RNA immunoprecipitation and chromatin immunoprecipitation sequencing were conducted to identify potential targets of lncRNA ENO1-IT1. RESULTS: We have found F. nucleatum abundance correlated with high glucose metabolism in patients with CRC. Furthermore, F. nucleatum supported carcinogenesis via increasing CRC cell glucose metabolism. Mechanistically, F. nucleatum activated lncRNA ENO1-IT1 transcription via upregulating the binding efficiency of transcription factor SP1 to the promoter region of lncRNA ENO1-IT1. Elevated ENO1-IT behaved as a guider modular for KAT7 histone acetyltransferase, specifying the histone modification pattern on its target genes, including ENO1, and consequently altering CRC biological function. CONCLUSION: F. nucleatum and glucose metabolism are mechanistically, biologically and clinically connected to CRC. Targeting ENO1 pathway may be meaningful in treating patients with CRC with elevated F. nucleatum.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Fusobacterium Infections/genetics , Glycolysis/genetics , RNA, Long Noncoding/genetics , Animals , Biomarkers, Tumor , Colorectal Neoplasms/diagnostic imaging , DNA-Binding Proteins , Fluorodeoxyglucose F18/pharmacokinetics , Fusobacterium nucleatum , Gastrointestinal Microbiome , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases , Humans , Mice , Phosphopyruvate Hydratase , Positron Emission Tomography Computed Tomography , Prognosis , Radiopharmaceuticals/pharmacokinetics , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Proteins
12.
Front Psychol ; 8: 1814, 2017.
Article in English | MEDLINE | ID: mdl-29089909

ABSTRACT

Despite various lines of evidence implicating impaired decision-making ability in individuals with obsessive-compulsive disorder (OCD), neuropsychological investigation has generated inconsistent findings. Although the cortico-striato-thalamo-cortical (CSTC) circuitry has been suggested, the involvement of the cortex has not yet been fully demonstrated. Moreover, it is unknown whether surgical intervention on the CSTC circuitry results in a predicted improvement of decision-making ability of OCD. Here we present a study of decision making based on the Iowa Gambling Task (IGT) to investigate decision making in a large sample of individuals with treatment-resistant OCD with and without anterior capsulotomy (AC). Task performance was evaluated in healthy subjects, individuals with OCD that had not undergone surgery, and postsurgical OCD patients with AC. The latter group was further divided into a short-term postsurgical group and a long-term postsurgical group. We found that the OCD patients without surgery performed significantly worse than the healthy controls on the IGT. There were no significant differences in decision-making between the presurgical OCD patients and those at the short-term postsurgical follow-up. Decision-making ability of the long-term postsurgical OCD patients was improved to the level comparable to that of healthy controls. All clinical symptoms (OCD, depression, and anxiety) assessed by psychiatric rating scales were significantly alleviated post-surgically, but exhibited no correlation with their IGT task performance. Our findings provide strong evidence that OCD is linked to impairments in decision-making ability; that impaired CSTC circuitry function is directly involved in the manifestation of OCD; and that AC related improvements in cognitive functions are caused by long-term plasticity in the brain circuitry.

SELECTION OF CITATIONS
SEARCH DETAIL