Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 35
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cancer Cell ; 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39303724

RÉSUMÉ

Microsatellite stable (MSS) colorectal cancers (CRCs) are often resistant to anti-programmed death-1 (PD-1) therapy. Here, we show that a CRC pathogen, Fusobacterium nucleatum (Fn), paradoxically sensitizes MSS CRC to anti-PD-1. Fecal microbiota transplantation (FMT) from patients with Fn-high MSS CRC to germ-free mice bearing MSS CRC confers sensitivity to anti-PD-1 compared to FMT from Fn-low counterparts. Single Fn administration also potentiates anti-PD-1 efficacy in murine allografts and CD34+-humanized mice bearing MSS CRC. Mechanistically, we demonstrate that intratumoral Fn generates abundant butyric acid, which inhibits histone deacetylase (HDAC) 3/8 in CD8+ T cells, inducing Tbx21 promoter H3K27 acetylation and expression. TBX21 transcriptionally represses PD-1, alleviating CD8+ T cell exhaustion and promoting effector function. Supporting this notion, knockout of a butyric acid-producing gene in Fn abolishes its anti-PD-1 boosting effect. In patients with MSS CRC, high intratumoral Fn predicts favorable response to anti-PD-1 therapy, indicating Fn as a potential biomarker of immunotherapy response in MSS CRC.

2.
Cancer Cell ; 42(8): 1386-1400.e8, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39137727

RÉSUMÉ

Changes in plasma and fecal metabolomes in colorectal cancer (CRC) progression (normal-adenoma-CRC) remain unclear. Here, plasma and fecal samples were collected from four independent cohorts of 1,251 individuals (422 CRC, 399 colorectal adenoma [CRA], and 430 normal controls [NC]). By metabolomic profiling, signature plasma and fecal metabolites with consistent shift across NC, CRA, and CRC are identified, including CRC-enriched oleic acid and CRC-depleted allocholic acid. Oleic acid exhibits pro-tumorigenic effects in CRC cells, patient-derived organoids, and two murine CRC models, whereas allocholic acid has opposing effects. By integrative analysis, we found that oleic acid or allocholic acid directly binds to α-enolase or farnesoid X receptor-1 in CRC cells, respectively, to modulate cancer-associated pathways. Clinically, we establish a panel of 17 plasma metabolites that accurately diagnoses CRC in a discovery and three validation cohorts (AUC = 0.848-0.987). Overall, we characterize metabolite signatures, mechanistic significance, and diagnostic potential of plasma and fecal metabolomes in CRC.


Sujet(s)
Adénomes , Marqueurs biologiques tumoraux , Tumeurs colorectales , Évolution de la maladie , Fèces , Métabolomique , Humains , Tumeurs colorectales/diagnostic , Tumeurs colorectales/métabolisme , Tumeurs colorectales/sang , Tumeurs colorectales/anatomopathologie , Fèces/composition chimique , Adénomes/métabolisme , Adénomes/diagnostic , Adénomes/anatomopathologie , Adénomes/sang , Métabolomique/méthodes , Animaux , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/sang , Souris , Mâle , Femelle , Dépistage précoce du cancer/méthodes , Métabolome , Adulte d'âge moyen , Acide oléique/métabolisme , Acide oléique/sang , Sujet âgé
3.
Pharmacol Res ; 208: 107373, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39197712

RÉSUMÉ

Gastrointestinal cancer is a worldwide health challenge due to its dramatically increasing prevalence and as a leading cause of cancer-related mortality. Increasing evidence has illustrated the vital role of gut microbes-derived metabolites in gastrointestinal cancer progression and treatment. Microbial metabolites are produced by the gut microbiota that utilizes both extrinsic dietary components and intrinsic host-generated compounds. Meanwhile, certain categories of metabolites such as short-chain fatty acids, bile acids, tryptophan, and indole derivatives, are linked to gastrointestinal malignancy. In this review, the major classes of microbial metabolites and their impacts on various gastrointestinal cancers including colorectal cancer, gastric cancer, and hepatocellular carcinoma, have been introduced. The application of microbial metabolites as predictive biomarkers for early diagnosis and prognosis of gastrointestinal cancer has also been explored. In addition, therapeutic potential of strategies that target microbial metabolites against gastrointestinal cancer is further evaluated.


Sujet(s)
Microbiome gastro-intestinal , Tumeurs gastro-intestinales , Humains , Tumeurs gastro-intestinales/diagnostic , Tumeurs gastro-intestinales/métabolisme , Tumeurs gastro-intestinales/thérapie , Tumeurs gastro-intestinales/microbiologie , Animaux , Acides gras volatils/métabolisme , Acides et sels biliaires/métabolisme
4.
Cell Rep Med ; 5(7): 101627, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38964315

RÉSUMÉ

The efficacy of chemotherapy varies significantly among patients with gastric cancer (GC), and there is currently no effective strategy to predict chemotherapeutic outcomes. In this study, we successfully establish 57 GC patient-derived organoids (PDOs) from 73 patients with GC (78%). These organoids retain histological characteristics of their corresponding primary GC tissues. GC PDOs show varied responses to different chemotherapeutics. Through RNA sequencing, the upregulation of tumor suppression genes/pathways is identified in 5-fluorouracil (FU)- or oxaliplatin-sensitive organoids, whereas genes/pathways associated with proliferation and invasion are enriched in chemotherapy-resistant organoids. Gene expression biomarker panels, which could distinguish sensitive and resistant patients to 5-FU and oxaliplatin (area under the dose-response curve [AUC] >0.8), are identified. Moreover, the drug-response results in PDOs are validated in patient-derived organoids-based xenograft (PDOX) mice and are consistent with the actual clinical response in 91.7% (11/12) of patients with GC. Assessing chemosensitivity in PDOs can be utilized as a valuable tool for screening chemotherapeutic drugs in patients with GC.


Sujet(s)
Fluorouracil , Organoïdes , Médecine de précision , Tumeurs de l'estomac , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Humains , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/anatomopathologie , Organoïdes/métabolisme , Animaux , Médecine de précision/méthodes , Fluorouracil/pharmacologie , Fluorouracil/usage thérapeutique , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Mâle , Femelle , Tests d'activité antitumorale sur modèle de xénogreffe , Tests de criblage d'agents antitumoraux/méthodes , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Oxaliplatine/pharmacologie , Oxaliplatine/usage thérapeutique , Adulte d'âge moyen , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Sujet âgé , Pertinence clinique
5.
Cancer Lett ; 598: 217087, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-38964732

RÉSUMÉ

Human appendix is critical for the maintenance of intestinal homeostasis. Appendicectomy has been the optimal treatment of acute appendicitis, yet the cancer incidence after appendix removal remains unclear. In this territory-wide retrospective cohort study, adult participants who underwent appendicectomy from 2000 to 2018 were retrieved from a population database (n = 43,983), while matched reference participants were retrieved as controls (n = 85,853). After appendicectomy, the overall cancer risk was significantly increased (subdistribution hazard ratio (SHR) = 1.124) compared to the non-appendicectomy group. Appendicectomy-treated males had higher cancer risk than males without appendicectomy (SHR = 1.197), while such difference was not observed in female participants. Significant increase in cancer risk was also observed in elder participants (age >60) with appendicectomy (SHR = 1.390). Appendicectomy was positively correlated with the risk of digestive tract and respiratory cancers including colon (SHR = 1.440), pancreas (SHR = 1.930), and trachea, bronchus, and lung (SHR = 1.394). In contrast, the risk of liver cancer was markedly decreased after appendicectomy (SHR = 0.713). In conclusion, we reported the association of appendicectomy with subsequent cancer incidence. These findings highlight the potential complication after appendix removal and the necessity of post-operative management to monitor and prevent long-term adverse events.


Sujet(s)
Appendicectomie , Appendicite , Humains , Appendicectomie/effets indésirables , Mâle , Femelle , Adulte d'âge moyen , Adulte , Études rétrospectives , Incidence , Appendicite/chirurgie , Appendicite/épidémiologie , Facteurs de risque , Sujet âgé , Tumeurs/épidémiologie , Asiatiques , Appendice vermiforme/chirurgie , Appendice vermiforme/anatomopathologie , Jeune adulte
6.
Gut ; 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886045

RÉSUMÉ

Gastric cancer (GC) is one of the most common malignancies and a prominent cause of cancer mortality worldwide. A distinctive characteristic of GC is its intimate association with commensal microbial community. Although Helicobacter pylori is widely recognised as an inciting factor of the onset of gastric carcinogenesis, increasing evidence has indicated the substantial involvement of microbes that reside in the gastric mucosa during disease progression. In particular, dysregulation in gastric microbiota could play pivotal roles throughout the whole carcinogenic processes, from the development of precancerous lesions to gastric malignancy. Here, current understanding of the gastric microbiota in GC development is summarised. Potential translational and clinical implications of using gastric microbes for GC diagnosis, prognosis and therapeutics are also evaluated, with further discussion on conceptual haziness and limitations at present. Finally, we highlight that modulating microbes is a novel and promising frontier for the prevention and management of GC, which necessitates future in-depth investigations.

7.
Nat Microbiol ; 9(6): 1467-1482, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38750176

RÉSUMÉ

Bacteria such as the oral microbiome member Peptostreptococcus anaerobius can exacerbate colorectal cancer (CRC) development. Little is known regarding whether these immunomodulatory bacteria also affect antitumour immune checkpoint blockade therapy. Here we show that administration of P. anaerobius abolished the efficacy of anti-PD1 therapy in mouse models of CRC. P. anaerobius both induced intratumoral myeloid-derived suppressor cells (MDSCs) and stimulated their immunosuppressive activities to impair effective T cell responses. Mechanistically, P. anaerobius administration activated integrin α2ß1-NF-κB signalling in CRC cells to induce secretion of CXCL1 and recruit CXCR2+ MDSCs into tumours. The bacterium also directly activated immunosuppressive activity of intratumoral MDSCs by secreting lytC_22, a protein that bound to the Slamf4 receptor on MDSCs and promoted ARG1 and iNOS expression. Finally, therapeutic targeting of either integrin α2ß1 or the Slamf4 receptor were revealed as promising strategies to overcome P. anaerobius-mediated resistance to anti-PD1 therapy in CRC.


Sujet(s)
Tumeurs colorectales , Cellules myéloïdes suppressives , Récepteur-1 de mort cellulaire programmée , Animaux , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme , Souris , Tumeurs colorectales/immunologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/microbiologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Humains , Lignée cellulaire tumorale , Intégrine alpha2bêta1/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Famille des molécules de signalisation de l'activation des lymphocytes/métabolisme , Souris de lignée C57BL , Transduction du signal , Résistance aux médicaments antinéoplasiques , Modèles animaux de maladie humaine , Femelle , Facteur de transcription NF-kappa B/métabolisme
8.
Gut ; 73(9): 1478-1488, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-38599786

RÉSUMÉ

OBJECTIVE: Probiotic Lactococcus lactis is known to confer health benefits to humans. Here, we aimed to investigate the role of L. lactis in colorectal cancer (CRC). DESIGN: L. lactis abundance was evaluated in patients with CRC (n=489) and healthy individuals (n=536). L. lactis was isolated from healthy human stools with verification by whole genome sequencing. The effect of L. lactis on CRC tumourigenesis was assessed in transgenic Apc Min/+ mice and carcinogen-induced CRC mice. Faecal microbiota was profiled by metagenomic sequencing. Candidate proteins were characterised by nano liquid chromatography-mass spectrometry. Biological function of L. lactis conditioned medium (HkyuLL 10-CM) and functional protein was studied in human CRC cells, patient-derived organoids and xenograft mice. RESULTS: Faecal L. lactis was depleted in patients with CRC. A new L. lactis strain was isolated from human stools and nomenclated as HkyuLL 10. HkyuLL 10 supplementation suppressed CRC tumourigenesis in Apc Min/+ mice, and this tumour-suppressing effect was confirmed in mice with carcinogen-induced CRC. Microbiota profiling revealed probiotic enrichment including Lactobacillus johnsonii in HkyuLL 10-treated mice. HkyuLL 10-CM significantly abrogated the growth of human CRC cells and patient-derived organoids. Such protective effect was attributed to HkyuLL 10-secreted proteins, and we identified that α-mannosidase was the functional protein. The antitumourigenic effect of α-mannosidase was demonstrated in human CRC cells and organoids, and its supplementation significantly reduced tumour growth in xenograft mice. CONCLUSION: HkyuLL 10 suppresses CRC tumourigenesis in mice through restoring gut microbiota and secreting functional protein α-mannosidase. HkyuLL 10 administration may serve as a prophylactic measure against CRC.


Sujet(s)
Carcinogenèse , Tumeurs colorectales , Fèces , Microbiome gastro-intestinal , Lactococcus lactis , Probiotiques , alpha-Mannosidase , Animaux , Tumeurs colorectales/microbiologie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/prévention et contrôle , Microbiome gastro-intestinal/physiologie , Humains , Souris , Probiotiques/usage thérapeutique , Fèces/microbiologie , alpha-Mannosidase/métabolisme , Souris transgéniques , Femelle , Mâle
9.
Cell Rep Med ; 5(4): 101478, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38631285

RÉSUMÉ

Immunotherapy has emerged as a robust approach against cancer, yet its efficacy has varied among individuals, accompanied by the occurrence of immune-related adverse events. As a result, the efficacy of immunotherapy is far from satisfactory, and enormous efforts have been invested to develop strategies to improve patient outcomes. The gut microbiome is now well acknowledged for its critical role in immunotherapy, with better understanding on host-microbes interaction in the context of cancer treatment. Also, an increasing number of trials have been conducted to evaluate the potential and feasibility of microbiome-targeting approaches to enhance efficacy of cancer treatment in patients. Here, the role of the gut microbiome and metabolites (e.g., short-chain fatty acids, tryptophan metabolites) in immunotherapy and the underlying mechanisms are explored. The application of microbiome-targeting approaches that aim to improve immunotherapy efficacy (e.g., fecal microbiota transplantation, probiotics, dietary intervention) is also elaborated, with further discussion on current challenges and suggestions for future research.


Sujet(s)
Microbiome gastro-intestinal , Microbiote , Tumeurs , Humains , Immunothérapie , Résultat thérapeutique , Transplantation de microbiote fécal
10.
Nat Commun ; 15(1): 427, 2024 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-38199995

RÉSUMÉ

The microbiome in a specific human organ has been well-studied, but few reports have investigated the multi-organ microbiome as a whole. Here, we aim to analyse the intra-individual inter-organ and intra-organ microbiome in deceased humans. We collected 1608 samples from 53 sites of 7 surface organs (oral cavity, esophagus, stomach, small intestine, appendix, large intestine and skin; n = 33 subjects) and performed microbiome profiling, including 16S full-length sequencing. Microbial diversity varied dramatically among organs, and core microbial species co-existed in different intra-individual organs. We deciphered microbial changes across distinct intra-organ sites, and identified signature microbes, their functional traits, and interactions specific to each site. We revealed significant microbial heterogeneity between paired mucosa-lumen samples of stomach, small intestine, and large intestine. Finally, we established the landscape of inter-organ relationships of microbes along the digestive tract. Therefore, we generate a catalogue of bacterial composition, diversity, interaction, functional traits, and bacterial translocation in human at inter-organ and intra-organ levels.


Sujet(s)
Appendice vermiforme , Microbiote , Humains , Translocation bactérienne , Estomac , Microbiote/génétique , Bouche
11.
EBioMedicine ; 100: 104952, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38176203

RÉSUMÉ

BACKGROUND: Gut probiotic depletion is associated with non-alcoholic fatty liver disease-associated hepatocellular carcinoma (NAFLD-HCC). Here, we investigated the prophylactic potential of Lactobacillus acidophilus against NAFLD-HCC. METHODS: NAFLD-HCC conventional and germ-free mice were established by diethylnitrosamine (DEN) injection with feeding of high-fat high-cholesterol (HFHC) or choline-deficient high-fat (CDHF) diet. Orthotopic NAFLD-HCC allografts were established by intrahepatic injection of murine HCC cells with HFHC feeding. Metabolomic profiling was performed using liquid chromatography-mass spectrometry. Biological functions of L. acidophilus conditional medium (L.a CM) and metabolites were determined in NAFLD-HCC human cells and mouse organoids. FINDINGS: L. acidophilus supplementation suppressed NAFLD-HCC formation in HFHC-fed DEN-treated mice. This was confirmed in orthotopic allografts and germ-free tumourigenesis mice. L.a CM inhibited the growth of NAFLD-HCC human cells and mouse organoids. The protective function of L. acidophilus was attributed to its non-protein small molecules. By metabolomic profiling, valeric acid was the top enriched metabolite in L.a CM and its upregulation was verified in liver and portal vein of L. acidophilus-treated mice. The protective function of valeric acid was demonstrated in NAFLD-HCC human cells and mouse organoids. Valeric acid significantly suppressed NAFLD-HCC formation in HFHC-fed DEN-treated mice, accompanied by improved intestinal barrier integrity. This was confirmed in another NAFLD-HCC mouse model induced by CDHF diet and DEN. Mechanistically, valeric acid bound to hepatocytic surface receptor GPR41/43 to inhibit Rho-GTPase pathway, thereby ablating NAFLD-HCC. INTERPRETATION: L. acidophilus exhibits anti-tumourigenic effect in mice by secreting valeric acid. Probiotic supplementation is a potential prophylactic of NAFLD-HCC. FUNDING: Shown in Acknowledgments.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Stéatose hépatique non alcoolique , Acides pentanoïques , Probiotiques , Humains , Animaux , Souris , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/étiologie , Stéatose hépatique non alcoolique/étiologie , Stéatose hépatique non alcoolique/complications , Lactobacillus acidophilus , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/étiologie , Foie/métabolisme , Transformation cellulaire néoplasique/métabolisme , Carcinogenèse/anatomopathologie , Alimentation riche en graisse , Choline/métabolisme , Probiotiques/pharmacologie , Probiotiques/usage thérapeutique , Souris de lignée C57BL
12.
Gut ; 72(12): 2272-2285, 2023 Nov 24.
Article de Anglais | MEDLINE | ID: mdl-37770127

RÉSUMÉ

OBJECTIVE: Gut microbiota is a key player in dictating immunotherapy response. We aimed to explore the immunomodulatory effect of probiotic Lactobacillus gallinarum and its role in improving anti-programmed cell death protein 1 (PD1) efficacy against colorectal cancer (CRC). DESIGN: The effects of L. gallinarum in anti-PD1 response were assessed in syngeneic mouse models and azoxymethane/dextran sulfate sodium-induced CRC model. The change of immune landscape was identified by multicolour flow cytometry and validated by immunohistochemistry staining and in vitro functional assays. Liquid chromatography-mass spectrometry was performed to identify the functional metabolites. RESULTS: L. gallinarum significantly improved anti-PD1 efficacy in two syngeneic mouse models with different microsatellite instability (MSI) statuses (MSI-high for MC38, MSI-low for CT26). Such effect was confirmed in CRC tumourigenesis model. L. gallinarum synergised with anti-PD1 therapy by reducing Foxp3+ CD25+ regulatory T cell (Treg) intratumoural infiltration, and enhancing effector function of CD8+ T cells. L. gallinarum-derived indole-3-carboxylic acid (ICA) was identified as the functional metabolite. Mechanistically, ICA inhibited indoleamine 2,3-dioxygenase (IDO1) expression, therefore suppressing kynurenine (Kyn) production in tumours. ICA also competed with Kyn for binding site on aryl hydrocarbon receptor (AHR) and antagonised Kyn binding on CD4+ T cells, thereby inhibiting Treg differentiation in vitro. ICA phenocopied L. gallinarum effect and significantly improved anti-PD1 efficacy in vivo, which could be reversed by Kyn supplementation. CONCLUSION: L. gallinarum-derived ICA improved anti-PD1 efficacy in CRC through suppressing CD4+Treg differentiation and enhancing CD8+T cell function by modulating the IDO1/Kyn/AHR axis. L. gallinarum is a potential adjuvant to augment anti-PD1 efficacy against CRC.


Sujet(s)
Tumeurs colorectales , Inhibiteurs de points de contrôle immunitaires , Cynurénine , Lactobacillus , Animaux , Souris , Lymphocytes T CD8+ , Tumeurs colorectales/traitement médicamenteux , Cynurénine/métabolisme , Récepteurs à hydrocarbure aromatique/effets des médicaments et des substances chimiques , Récepteurs à hydrocarbure aromatique/métabolisme , Lymphocytes T régulateurs , Lactobacillus/composition chimique , Récepteur-1 de mort cellulaire programmée/effets des médicaments et des substances chimiques , Récepteur-1 de mort cellulaire programmée/immunologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Bacterial Lysates/pharmacologie , Bacterial Lysates/usage thérapeutique
13.
Gut ; 72(11): 2112-2122, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37491158

RÉSUMÉ

OBJECTIVE: Roseburia intestinalis is a probiotic species that can suppress intestinal inflammation by producing metabolites. We aimed to study the role of R. intestinalis in colorectal tumourigenesis and immunotherapy. DESIGN: R. intestinalis abundance was evaluated in stools of patients with colorectal cancer (CRC) (n=444) and healthy controls (n=575). The effects of R. intestinalis were studied in ApcMin/+ or azoxymethane (AOM)-induced CRC mouse models, and in syngeneic mouse xenograft models of CT26 (microsatellite instability (MSI)-low) or MC38 (MSI-high). The change of immune landscape was evaluated by multicolour flow cytometry and immunohistochemistry staining. Metabolites were profiled by metabolomic profiling. RESULTS: R. intestinalis was significantly depleted in stools of patients with CRC compared with healthy controls. R. intestinalis administration significantly inhibited tumour formation in ApcMin/+ mice, which was confirmed in mice with AOM-induced CRC. R. intestinalis restored gut barrier function as indicated by improved intestinal permeability and enhanced expression of tight junction proteins. Butyrate was identified as the functional metabolite generated by R. intestinalis. R. intestinalis or butyrate suppressed tumour growth by inducing cytotoxic granzyme B+, interferon (IFN)-γ+ and tumour necrosis factor (TNF)-α+ CD8+ T cells in orthotopic mouse models of MC38 or CT26. R. intestinalis or butyrate also significantly improved antiprogrammed cell death protein 1 (anti-PD-1) efficacy in mice bearing MSI-low CT26 tumours. Mechanistically, butyrate directly bound to toll-like receptor 5 (TLR5) receptor on CD8+ T cells to induce its activity through activating nuclear factor kappa B (NF-κB) signalling. CONCLUSION: R. intestinalis protects against colorectal tumourigenesis by producing butyrate, which could also improve anti-PD-1 efficacy by inducing functional CD8+ T cells. R. intestinalis is a potential adjuvant to augment anti-PD-1 efficacy against CRC.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs colorectales , Humains , Souris , Animaux , Butyrates/pharmacologie , Carcinogenèse , Transformation cellulaire néoplasique , Tumeurs colorectales/métabolisme
14.
FEMS Microbiol Rev ; 47(4)2023 07 05.
Article de Anglais | MEDLINE | ID: mdl-37407433

RÉSUMÉ

The gut microbiota plays a crucial role in regulating various host metabolic, immune, and neuroendocrine functions, and has a significant impact on human health. Several lines of evidence suggest that gut dysbiosis is associated with a variety of diseases, including cancer. The gut microbiota can impact the development and progression of cancer through a range of mechanisms, such as regulating cell proliferation and death, modulating the host immune response, and altering the host metabolic state. Gene regulatory programs are considered critical mediators between the gut microbiota and host phenotype, of which RNA N6-methyladenosine (m6A) modifications have attracted much attention recently. Aberrant m6A modifications have been shown to play a crucial role in cancer development. This review aims to provide an overview of the diverse roles of gut microbiota and RNA m6A modifications in cancer and highlight their potential interactions in cancer development.


Sujet(s)
Microbiome gastro-intestinal , Tumeurs , Humains , Microbiome gastro-intestinal/génétique , Prolifération cellulaire , Tumeurs/génétique , ARN
15.
Gut Microbes ; 15(1): 2203968, 2023.
Article de Anglais | MEDLINE | ID: mdl-37095682

RÉSUMÉ

Trillions of microbes are indigenous to the human gastrointestinal tract, together forming an ecological community known as the gut microbiota. The gut microbiota is involved in dietary digestion to produce various metabolites. In healthy condition, microbial metabolites have unneglectable roles in regulating host physiology and intestinal homeostasis. However, increasing studies have reported the correlation between metabolites and the development of colorectal cancer (CRC), with the identification of oncometabolites. Meanwhile, metabolites can also influence the efficacy of cancer treatments. In this review, metabolites derived from microbes-mediated metabolism of dietary carbohydrates, proteins, and cholesterol, are introduced. The roles of pro-tumorigenic (secondary bile acids and polyamines) and anti-tumorigenic (short-chain fatty acids and indole derivatives) metabolites in CRC development are then discussed. The impacts of metabolites on chemotherapy and immunotherapy are further elucidated. Collectively, given the importance of microbial metabolites in CRC, therapeutic approaches that target metabolites may be promising to improve patient outcome.


Sujet(s)
Tumeurs colorectales , Microbiome gastro-intestinal , Humains , Microbiome gastro-intestinal/physiologie , Carcinogenèse , Intestins
16.
Semin Cancer Biol ; 92: 16-27, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-36965839

RÉSUMÉ

Excess body weight is a global health problem due to sedentary lifestyle and unhealthy diet, affecting 2 billion population worldwide. Obesity is a major risk factor for metabolic diseases. Notably, the metabolic risk of obesity largely depends on body weight distribution, of which visceral adipose tissues but not subcutaneous fats are closely associated with obesity comorbidities, including type 2 diabetes, non-alcoholic fatty liver disease, cardiovascular disease and certain types of cancer. Latest multi-omics and mechanistical studies reported the crucial involvement of genetic and epigenetic alterations, adipokines dysregulation, immunity changes, imbalance of white and brown adipose tissues, and gut microbial dysbiosis in mediating the pathogenic association between visceral adipose tissues and comorbidities. In this review, we explore the epidemiology of excess body weight and the up-to-date mechanism of how excess body weight and obesity lead to chronic complications. We also examine the utilization of visceral fat measurement as an accurate clinical parameter for risk assessment in healthy individuals and clinical outcome prediction in obese subjects. In addition, current approaches for the prevention and treatment of excess body weight and its related metabolic comorbidities are further discussed.


Sujet(s)
Diabète de type 2 , Humains , Diabète de type 2/complications , Obésité/complications , Obésité/épidémiologie , Obésité/métabolisme , Comorbidité , Facteurs de risque , Régime alimentaire
17.
Genomics Proteomics Bioinformatics ; 21(1): 84-96, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-35914737

RÉSUMÉ

Colorectal cancer (CRC) is one of the most frequently diagnosed cancers and the leading cause of cancer-associated deaths. Epidemiological studies have shown that both genetic and environmental risk factors contribute to the development of CRC. Several metagenomic studies of CRC have identified gut dysbiosis as a fundamental risk factor in the evolution of colorectal malignancy. Although enormous efforts and substantial progresses have been made in understanding the relationship between human gut microbiome and CRC, the precise mechanisms involved remain elusive. Recent data have shown a direct causative role of the gut microbiome in DNA damage, inflammation, and drug resistance in CRC, suggesting that modulation of gut microbiome could act as a powerful tool in CRC prevention and therapy. Here, we provide an overview of the relationship between gut microbiome and CRC, and explore relevant mechanisms of colorectal tumorigenesis. We next highlight the potential of bacterial species as clinical biomarkers, as well as their roles in therapeutic response. Factors limiting the clinical translation of gut microbiome and strategies for resolving current challenges are further discussed.


Sujet(s)
Tumeurs colorectales , Microbiome gastro-intestinal , Microbiote , Humains , Microbiome gastro-intestinal/génétique , Tumeurs colorectales/diagnostic , Tumeurs colorectales/étiologie , Tumeurs colorectales/thérapie , Marqueurs biologiques , Carcinogenèse
19.
Cancers (Basel) ; 14(14)2022 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-35884552

RÉSUMÉ

N6-methyladenosine (m6A) is the most abundant internal modification in eukaryotic cell mRNA, and this modification plays a key role in regulating mRNA translation, splicing, and stability. Emerging evidence implicates aberrant m6A as a crucial player in the occurrence and development of diseases, especially GI cancers. Among m6A regulators, YTHDF1 is the most abundant m6A reader that functionally connects m6A-modified mRNA to its eventual fate, mostly notably protein translation. Here, we summarized the function, molecular mechanisms, and clinical implications of YTHDF1 in GI cancers. YTHDF1 is largely upregulated in multiple GI cancer and its high expression predicts poor patient survival. In vitro and in vivo experimental evidence largely supports the role of YTDHF1 in promoting cancer initiation, progression, and metastasis, which suggests the oncogenic function of YTHDF1 in GI cancers. Besides, YTHDF1 overexpression is associated with changes in the tumor microenvironment that are favorable to tumorigenesis. Mechanistically, YTHDF1 regulates the expression of target genes by promoting translation, thereby participating in cancer-related signaling pathways. Targeting YTHDF1 holds therapeutic potential, as the overexpression of YTHDF1 is associated with tumor resistance to chemotherapy and immunotherapy. In summary, YTHDF1-mediated regulation of m6A modified mRNA is an actionable target and a prognostic factor for GI cancers.

20.
Biomedicines ; 10(7)2022 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-35885015

RÉSUMÉ

Colorectal cancer (CRC) is a malignant disease that is the second most common cancer worldwide. CRC arises from the complex interactions among a variety of genetic and environmental factors. To understand the mechanism of colon tumorigenesis, preclinical studies have developed various mouse models including carcinogen-induced and transgenic mice to recapitulate CRC in humans. Using these mouse models, scientific breakthroughs have been made on the understanding of the pathogenesis of this complex disease. Moreover, the availability of transgenic knock-in or knock-out mice further increases the potential of CRC mouse models. In this review, the overall features of carcinogen-induced (focusing on azoxymethane and azoxymethane/dextran sulfate sodium) and transgenic (focusing on ApcMin/+) mouse models, as well as their mechanisms to induce colon tumorigenesis, are explored. We also discuss limitations of these mouse models and their applications in the evaluation and study of drugs and treatment regimens against CRC. Through these mouse models, a better understanding of colon tumorigenesis can be achieved, thereby facilitating the discovery of novel therapeutic strategies against CRC.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE