Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 38
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 15(1): 6225, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39043671

RÉSUMÉ

The deep ocean, a vast thermal reservoir, absorbs excess heat under greenhouse warming, which ultimately regulates the Earth's surface climate. Even if CO2 emissions are successfully reduced, the stored heat will gradually be released, resulting in a particular pattern of ocean warming. Here, we show that deep ocean warming will lead to El Niño-like ocean warming and resultant increased precipitation in the tropical eastern Pacific with southward shift of the intertropical convergence zone. Consequently, the El Niño-Southern Oscillation shifts eastward, intensifying Eastern Pacific El Niño events. In particular, the deep ocean warming could increase convective extreme El Niño events by 40 to 80% relative to the current climate. Our findings suggest that anthropogenic greenhouse warming will have a prolonged impact on El Niño variability through delayed deep ocean warming, even if CO2 stabilization is achieved.

2.
Mol Pharm ; 21(7): 3330-3342, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38875185

RÉSUMÉ

The aberrant assembly of amyloid-ß (Aß) is implicated in Alzheimer's disease (AD). Recent clinical outcomes of Aß-targeted immunotherapy reinforce the notion that clearing Aß burden is a potential therapeutic approach for AD. Herein, to develop drug candidates for chemically driven clearance of Aß aggregates, we synthesized 51 novel polyfunctionalized furo[2,3-b:4,5-b']dipyridine-chalcone hybrid compounds. After conducting two types of cell-free anti-Aß functional assays, Aß aggregation prevention and Aß aggregate clearance, we selected YIAD-0336, (E)-8-((1H-pyrrol-2-yl)methylene)-10-(4-chlorophenyl)-2,4-dimethyl-7,8-dihydropyrido[3',2':4,5]furo[3,2-b]quinolin-9(6H)-one, for further in vivo investigations. As YIAD-0336 exhibited a low blood-brain barrier penetration profile, it was injected along with aggregated Aß directly into the intracerebroventricular region of ICR mice and ameliorated spatial memory in Y-maze tests. Next, YIAD-0336 was orally administered to 5XFAD transgenic mice with intravenous injections of mannitol, and YIAD-0336 significantly removed Aß plaques from the brains of 5XFAD mice. Collectively, YIAD-0336 dissociated toxic aggregates in the mouse brain and hence alleviated cognitive deterioration. Our findings indicate that chemically driven clearance of Aß aggregates is a promising therapeutic approach for AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Modèles animaux de maladie humaine , Souris transgéniques , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Souris , Peptides bêta-amyloïdes/métabolisme , Chalcone/composition chimique , Chalcone/pharmacologie , Chalcone/analogues et dérivés , Chalcones/composition chimique , Chalcones/pharmacologie , Chalcones/administration et posologie , Mâle , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Humains , Mémoire/effets des médicaments et des substances chimiques , Agrégats de protéines/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Pyridines/composition chimique , Pyridines/pharmacologie , Pyridines/administration et posologie
3.
Heliyon ; 10(6): e27148, 2024 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-38500982

RÉSUMÉ

Breast cancer (BC) remains a significant global health threat, with triple-negative breast cancer (TNBC) standing out as a particularly aggressive subtype lacking targeted therapies. Addressing this gap, we propose Quiescin Q6 sulfhydryl oxidase 2 (QSOX2) as a potential therapeutic target, a disulfide bond-forming enzyme implicated in cancer progression. Using publicly available datasets, we conducted a comprehensive analysis of QSOX2 expression in BC tumor and non-tumor tissues, assessing its specificity across different molecular subtypes. We further explored correlations between QSOX2 expression and patient outcomes, utilizing datasets like TCGA and METABRIC. In addition, we performed in vitro experiments to evaluate QSOX2 expression in BC cell lines and investigate the effects of QSOX2 knockdown on various TNBC cellular processes, including cell proliferation, apoptosis resistance, migration, and the epithelial-to-mesenchymal transition (EMT). Our results reveal significantly elevated QSOX2 expression in BC tumor tissues, particularly in TNBC, and establish an association between high QSOX2 expression and increased patient mortality, cancer progression, and recurrence across various BC subtypes. Notably, QSOX2 knockdown in TNBC cell lines reduces cell proliferation, enhances apoptosis, and suppresses migration, potentially mediated through its influence on the EMT process. Furthermore, we identify a significant link between QSOX2 and integrin ß1 (ITGB1), suggesting that QSOX2 enhances ITGB1 stability, subsequently exacerbating the malignancy of TNBC. In conclusion, elevated QSOX2 expression emerges as a key factor associated with adverse patient outcomes in BC, particularly in TNBC, contributing to disease progression through various mechanisms, including the modulation of ITGB1 stability. Our findings underscore the potential of targeting QSOX2 as a therapeutic strategy for improving patient prognoses not only in TNBC but also in other BC subtypes.

4.
Int J Biol Sci ; 20(3): 1045-1063, 2024.
Article de Anglais | MEDLINE | ID: mdl-38322121

RÉSUMÉ

Homeobox genes and their encoded DNA-binding homeoproteins are master regulators of development. Consequently, these homeotic elements may regulate key steps in cancer pathogenesis. Here, using a combination of in silico analyses of large-scale patient datasets, in vitro RNAi phenotyping, and in vivo validation studies, we investigated the role of HOXB2 in different molecular subtypes of human breast cancer (BC). The gene expression signatures of HOXB2 are different across distinct BC subtypes due to various genetic alterations, but HOXB2 was specifically downregulated in the aggressive triple-negative subtype (TNBC). We found that the reduced expression of HOXB2 was correlated with the metastatic abilities (epithelial-to-mesenchymal transition) of TNBC cells. Further, we revealed that HOXB2 restrained TNBC aggressiveness by ECM organization. HOXB2 bound to the promoter regions of MATN3 and ECM2 and regulated their transcription levels. Forced expression of HOXB2 effectively prevented TNBC progression and metastasis in a mouse xenograft model. Reduction of HOXB2 and the HOXB2/MATN3/ECM2 transcriptional axis correlated with poor survival in patients with various cancers. Further, we found the long non-coding RNA HOXB-AS1 in complex with SMYD3, a lysine methyltransferase, as an epigenetic switch controlling HOXB2 expression. Overall, our results indicate a tumor-suppressive role of HOXB2 by maintaining ECM organization and delineate potential clinical utility of HOXB2 as a marker for TNBC patients.


Sujet(s)
Protéines à homéodomaine , Facteurs de transcription , Tumeurs du sein triple-négatives , Animaux , Humains , Souris , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire/génétique , Transformation cellulaire néoplasique/génétique , Transition épithélio-mésenchymateuse/génétique , Matrice extracellulaire/métabolisme , Régulation de l'expression des gènes tumoraux , Gènes homéotiques , Histone-lysine N-methyltransferase/génétique , Protéines à homéodomaine/génétique , Facteurs de transcription/génétique , Tumeurs du sein triple-négatives/métabolisme
5.
Mol Med ; 29(1): 114, 2023 09 18.
Article de Anglais | MEDLINE | ID: mdl-37718409

RÉSUMÉ

BACKGROUND: Sexual dimorphism in placental physiology affects the functionality of placental adaptation during adverse pregnancy. Defects of placental function compromise fetal programming, affecting the offspring's adult life. However, studies focusing on the relationship between sex-specific placental adaptation and consequent fetal maldevelopment under sub-optimal uterus milieu are still elusive. METHODS: Here, we investigated the effects of maternal lipopolysaccharide (LPS) exposure between placental sex. Pregnant ICR mice received intraperitoneal injection of phosphate-buffered saline or 100, 200, and 400 µg/kg LPS on the gestational day (GD) 15.5. To determine whether prenatal maternal LPS exposure resulted in complicated pregnancy outcomes, survival rate of embryos was calculated and the growth of embryos and placentas was examined. To elucidate global transcriptomic changes occurring in the placenta, total RNA-sequencing (RNA-seq) was performed in female and male placentas. RESULTS: LPS administration induced placental inflammation in both sexes at GD 17.5. Prenatal infection resulted in growth retardation in both sexes of embryos, and especially more prevalently in male. Impaired placental development was observed in a sex-specific manner. LPS 400 µg/kg reduced the percentage area of the labyrinth in females and junctional zone in males, respectively. RNA-sequencing revealed widespread sexually dimorphic transcriptional changes in placenta. In particular, representative changes were involved in biological processes such as trophoblast differentiation, nutrient/ion transporter, pregnancy, and immune system. CONCLUSIONS: Our results present the sexually dimorphic responses of placental physiology in intrauterine growth restriction model and provide tentative relationship further to be elucidated between sex-biased placental functional change and long-term effects on the offspring's later life.


Sujet(s)
Retard de croissance intra-utérin , Lipopolysaccharides , Femelle , Mâle , Grossesse , Souris , Animaux , Humains , Souris de lignée ICR , Retard de croissance intra-utérin/induit chimiquement , Placenta , ARN
6.
Sci Adv ; 9(25): eadh2412, 2023 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-37343086

RÉSUMÉ

Convective extreme El Niño (CEE) events, characterized by strong convective events in the eastern Pacific, are known to have a direct link to anomalous climate conditions worldwide, and it has been reported that CEE will occur more frequently under greenhouse warming. Here, using a set of CO2 ramp-up and ramp-down ensemble experiments, we show that frequency and maximum intensity of CEE events increase further in the ramp-down period from the ramp-up period. These changes in CEE are associated with the southward shift of the intertropical convergence zone and intensified nonlinear rainfall response to sea surface temperature change in the ramp-down period. The increasing frequency of CEE has substantial impacts on regional abnormal events and contributed considerably to regional mean climate changes to the CO2 forcings.

7.
Exp Mol Med ; 55(4): 725-734, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-37009802

RÉSUMÉ

Comparative oncology is a field of study that has been recently adopted for studying cancer and developing cancer therapies. Companion animals such as dogs can be used to evaluate novel biomarkers or anticancer targets before clinical translation. Thus, the value of canine models is increasing, and numerous studies have been conducted to analyze similarities and differences between many types of spontaneously occurring cancers in canines and humans. A growing number of canine cancer models as well as research-grade reagents for these models are becoming available, leading to substantial growth in comparative oncology research spanning from basic science to clinical trials. In this review, we summarize comparative oncology studies that have been conducted on the molecular landscape of various canine cancers and highlight the importance of the integration of comparative biology into cancer research.


Sujet(s)
Tumeurs , Animaux de compagnie , Humains , Animaux , Chiens , Modèles animaux de maladie humaine , Tumeurs/génétique , Tumeurs/thérapie
8.
Nat Immunol ; 24(1): 148-161, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36577929

RÉSUMÉ

Regulatory T (Treg) cells have an immunosuppressive function and highly express the immune checkpoint receptor PD-1 in the tumor microenvironment; however, the function of PD-1 in tumor-infiltrating (TI) Treg cells remains controversial. Here, we showed that conditional deletion of PD-1 in Treg cells delayed tumor progression. In Pdcd1fl/flFoxp3eGFP-Cre-ERT2(+/-) mice, in which both PD-1-expressing and PD-1-deficient Treg cells coexisted in the same tissue environment, conditional deletion of PD-1 in Treg cells resulted in impairment of the proliferative and suppressive capacity of TI Treg cells. PD-1 antibody therapy reduced the TI Treg cell numbers, but did not directly restore the cytokine production of TI CD8+ T cells in TC-1 lung cancer. Single-cell analysis indicated that PD-1 signaling promoted lipid metabolism, proliferation and suppressive pathways in TI Treg cells. These results suggest that PD-1 ablation or inhibition can enhance antitumor immunity by weakening Treg cell lineage stability and metabolic fitness in the tumor microenvironment.


Sujet(s)
Tumeurs , Lymphocytes T régulateurs , Animaux , Souris , Lymphocytes T CD8+ , Expression des gènes , Lymphocytes TIL , Tumeurs/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Microenvironnement tumoral
9.
Cell J ; 24(1): 51-54, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-35182065

RÉSUMÉ

General control non-derepressible 5 (Gcn5) is a member of histone acetyltransferase (HAT) that plays key roles during embryogenesis as well as in the development of various human cancers. Gcn5, an epigenetic regulator of Hoxc11, has been reported to be negatively regulated by Akt1 in the mouse embryonic fibroblasts (MEFs). However, the exact mechanism by which Akt1 regulates Gcn5 is not well understood. Using protein stability chase assay, we observed that Gcn5 is negatively regulated by Akt1 at the post-translational level in MEFs. The stability of Gcn5 protein is determined by the competitive binding with the protein partner that interacts with Gcn5. The interaction of Gcn5 and Cul4a-Ddb1 complex predominates and promotes ubiquitination of Gcn5 in the wild-type MEFs. On the other hand, in the Akt1-null MEFs, the interaction of Gcn5 and And-1 inhibits binding of Gcn5 and Cul4a-Dbd1 E3 ubiquitin ligase complex, thereby increasing the stability of the Gcn5 protein. Taken together, our study indicates that Akt1 negatively controls Gcn5 via the proteasomal degradation pathway, suggesting a potential mechanism that regulates the expression of Hox genes.

10.
J Immunol ; 207(7): 1703-1709, 2021 10 01.
Article de Anglais | MEDLINE | ID: mdl-34544812

RÉSUMÉ

Advances in data collection (high-throughput shotgun metagenomics, transcriptomics, and metabolomics) and analysis (bioinformatics and multiomics) led to the realization that all mammals are metaorganisms, shaped not only by their own genome but also by the genomes of the microbes that colonize them. To date, most studies have focused on the bacterial microbiome, whereas curated databases for viruses, fungi, and protozoa are still evolving. Studies on the interdependency of microbial kingdoms and their combined effects on host physiology are just starting. Although it is clear that past and present exposure to commensals and pathogens profoundly affect human physiology, such exposure is lacking in standard preclinical models such as laboratory mice. Laboratory mouse colonies are repeatedly rederived in germ-free status and subjected to restrictive, pathogen-free housing conditions. This review summarizes efforts to bring the wild microbiome into the laboratory setting to improve preclinical models and their translational research value.


Sujet(s)
Animaux de laboratoire/physiologie , Animaux sauvages/physiologie , Infections/immunologie , Animaux , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Axénie , Interactions hôte-microbes , Humains , Métabolomique , Métagénomique , Souris
11.
Nat Metab ; 3(8): 1042-1057, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34417593

RÉSUMÉ

Obesity and its consequences are among the greatest challenges in healthcare. The gut microbiome is recognized as a key factor in the pathogenesis of obesity. Using a mouse model, we show here that a wild-derived microbiome protects against excessive weight gain, severe fatty liver disease and metabolic syndrome during a 10-week course of high-fat diet. This phenotype is transferable only during the first weeks of life. In adult mice, neither transfer nor severe disturbance of the wild-type microbiome modifies the metabolic response to a high-fat diet. The protective phenotype is associated with increased secretion of metabolic hormones and increased energy expenditure through activation of brown adipose tissue. Thus, we identify a microbiome that protects against weight gain and its negative consequences through metabolic programming in early life. Translation of these results to humans may identify early-life therapeutics that protect against obesity.


Sujet(s)
Régime alimentaire , Résistance à la maladie , Prédisposition aux maladies , Exposition environnementale , Interactions hôte-microbes , Microbiote , Obésité/étiologie , Aliment pour animaux , Animaux , Régime alimentaire/effets indésirables , Alimentation riche en graisse , Modèles animaux de maladie humaine , Métabolisme énergétique , Microbiome gastro-intestinal , Souris , Facteurs temps , Prise de poids
12.
Anticancer Res ; 41(7): 3409-3417, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34230136

RÉSUMÉ

BACKGROUND/AIM: ER-positive breast cancer patients commonly undergo endocrine therapy with drugs such as tamoxifen. Despite tamoxifen being a highly effective drug, long-term treatment results in resistance in one-third of the patients. Although many explanations for the development of tamoxifen resistance have been put forward, a clearly defined underlying mechanism is still lacking. MATERIALS AND METHODS: The expression level of HOXB5 was evaluated between MCF7 breast cancer cells and tamoxifen-resistant MCF7 (TAMR) cells by RT-PCR. Then, the effect of HOXB5 on invasion and migration abilities as well as on cancer stemness were investigated through 3D culture and spheroid formation assay. RESULTS: In this study, we provide evidence that HOXB5 is up-regulated in TAMR cells. EGFR is concurrently overexpressed, and the EGFR signaling cascade is activated, resulting in migratory and invasive phenotypes in TAMR cells compared to MCF7 cells. However, HOXB5 knockdown in TAMR cells resulted in the de-activation of the EGFR signaling pathway, less aggressive phenotypes and restoration of sensitivity to tamoxifen treatment. More interestingly, TAMR cells expressed higher levels of stem cell markers, and as a result, their enhanced stemness allowed for a better formation of spheroids than MCF7 cells. When HOXB5 was overexpressed in MCF7 cells, they were able to form a larger number of spheroids as in TAMR cells. CONCLUSION: HOXB5 is one of the key factors involved in tumor aggression and progression in tamoxifen-resistant breast cancer cells.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Mouvement cellulaire/génétique , Résistance aux médicaments antinéoplasiques/génétique , Protéines à homéodomaine/génétique , Marqueurs biologiques tumoraux/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Évolution de la maladie , Récepteurs ErbB/génétique , Femelle , Régulation de l'expression des gènes tumoraux/génétique , Humains , Cellules MCF-7 , Transduction du signal/génétique , Cellules souches/anatomopathologie , Tamoxifène/pharmacologie
13.
Cells ; 10(7)2021 06 29.
Article de Anglais | MEDLINE | ID: mdl-34209871

RÉSUMÉ

Endocrine therapy is used to treat estrogen receptor (ER)-positive breast cancer. Tamoxifen is effective against this cancer subtype. Nonetheless, approximately 30% of patients treated with tamoxifen acquire resistance, resulting in therapeutic challenges. NR4A1 plays key roles in processes associated with carcinogenesis, apoptosis, DNA repair, proliferation, and inflammation. However, the role of NR4A1 in tamoxifen-resistant ER-positive breast cancer has not yet been elucidated. Here, we propose that NR4A1 is a promising target to overcome tamoxifen resistance. NR4A1 gene expression was downregulated in tamoxifen-resistant MCF7 (TamR) cells compared to that in MCF7 cells. Kaplan-Meier plots were used to identify high NR4A1 expression correlated with increased survival rates in patients with ER-positive breast cancer following tamoxifen treatment. Gain and loss of function experiments showed that NR4A1 restores sensitivity to tamoxifen by regulating cell proliferation, migration, invasion, and apoptosis. NR4A1 localized to the cytoplasm enhanced the expression of apoptotic factors. In silico and in vitro analyses revealed that NR4A1 enhanced responsiveness to tamoxifen by suppressing ERK signaling in ER-positive breast cancer, suggesting that the NR4A1/ERK signaling axis modulates tamoxifen resistance. These results indicate that NR4A1 could be a potential therapeutic target to overcome tamoxifen resistance in ER-positive breast cancer.


Sujet(s)
Tumeurs du sein/métabolisme , Résistance aux médicaments antinéoplasiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Récepteurs des oestrogènes/métabolisme , Tamoxifène/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Tumeurs du sein/traitement médicamenteux , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Invasion tumorale , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , ARN messager/génétique , ARN messager/métabolisme , Tamoxifène/usage thérapeutique
14.
J Cancer ; 12(15): 4626-4637, 2021.
Article de Anglais | MEDLINE | ID: mdl-34149926

RÉSUMÉ

Tamoxifen is a commonly used drug to treat estrogen receptor-positive patients with breast cancer. Despite the outstanding efficacy of tamoxifen, approximately one-third of patients develop resistance toward it, thereby presenting a therapeutic challenge. HOX genes may be involved in the acquisition of tamoxifen resistance. In this study, we identified HOXA5, a member of the HOX gene family, as a marker of tamoxifen resistance. Using ChIP assay, we found that HOXA5 expression was significantly overexpressed in tamoxifen-resistant MCF7 (TAMR) breast cancer cells because of reduced H3K27me3 binding. HOXA5 upregulation resulted in activation of the PI3K/AKT signaling cascade, which in turn, led to p53 and p21 reduction, ultimately making the TAMR cells less apoptotic. Furthermore, elevated HOXA5 expression resulted in breast cancer cells acquiring more mesenchymal-like and stem cell traits associated with aggressive breast cancer phenotypes. In conclusion, our results delineate a mechanism by which HOXA5 promotes tumorigenesis, cancer progression, and tamoxifen resistance in breast cancer cells.

16.
J Nat Prod ; 84(2): 195-203, 2021 02 26.
Article de Anglais | MEDLINE | ID: mdl-33534559

RÉSUMÉ

Separating the immunosuppressive activity of FK506 (1) from its neurotrophic activity is required to develop FK506 analogues as drugs for the treatment of neuronal diseases. Two new FK506 analogues, 9-deoxo-36,37-dihydro-prolylFK506 (2) and 9-deoxo-31-O-demethyl-36,37-dihydro-prolylFK506 (3) containing a proline moiety instead of the pipecolate ring at C-1 and modifications at the C-9/C-31 and C-36-C-37 positions, respectively, were biosynthesized, and their biological activities were evaluated. The proline substitution in 9-deoxo-36,37-dihydroFK506 and 9-deoxo-31-O-demethyl-36,37-dihydroFK506 reduced immunosuppressive activity by more than 120-fold, as previously observed. Compared with FK506 (1), 2 and 3 exhibited ∼1.2 × 105- and 2.2 × 105-fold reductions in immunosuppressive activity, respectively, whereas they retained almost identical neurite outgrowth activity. Furthermore, these compounds significantly increased the strength of synaptic transmission, confirming that replacement of the pipecolate ring with a proline is critical to reduce the strong immunosuppressive activity of FK506 (1) while enhancing its neurotrophic activity.


Sujet(s)
Excroissance neuronale/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Tacrolimus/analogues et dérivés , Animaux , Cellules cultivées , Fermentation , Hippocampe/cytologie , Immunosuppresseurs , Souris de lignée ICR , Structure moléculaire , Acides pipécoliques , Streptomyces/métabolisme
17.
Cell ; 184(3): 615-627.e17, 2021 02 04.
Article de Anglais | MEDLINE | ID: mdl-33453153

RÉSUMÉ

The microbiota shields the host against infections in a process known as colonization resistance. How infections themselves shape this fundamental process remains largely unknown. Here, we show that gut microbiota from previously infected hosts display enhanced resistance to infection. This long-term functional remodeling is associated with altered bile acid metabolism leading to the expansion of taxa that utilize the sulfonic acid taurine. Notably, supplying exogenous taurine alone is sufficient to induce this alteration in microbiota function and enhance resistance. Mechanistically, taurine potentiates the microbiota's production of sulfide, an inhibitor of cellular respiration, which is key to host invasion by numerous pathogens. As such, pharmaceutical sequestration of sulfide perturbs the microbiota's composition and promotes pathogen invasion. Together, this work reveals a process by which the host, triggered by infection, can deploy taurine as a nutrient to nourish and train the microbiota, promoting its resistance to subsequent infection.


Sujet(s)
Microbiome gastro-intestinal , Interactions hôte-pathogène , Animaux , Infections bactériennes/immunologie , Infections bactériennes/microbiologie , Numération de colonies microbiennes , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Interactions hôte-pathogène/effets des médicaments et des substances chimiques , Immunité , Souris de lignée C57BL , Sulfures/métabolisme , Taurine/pharmacologie
18.
Cancer Lett ; 495: 145-155, 2020 12 28.
Article de Anglais | MEDLINE | ID: mdl-32987137

RÉSUMÉ

Approximately 70% of breast cancers are estrogen receptor (ER)-positive and treated with endocrine therapy. A commonly used treatment agent, tamoxifen, shows high efficacy for improving prognosis. However, approximately one-third of patients treated with tamoxifen develop resistance to this drug. Here, we investigated the function of general control non-derepressible 5 (GCN5) and its downstream effectors in tamoxifen-resistant (TamR) breast cancer. TamR-MCF7 breast cancer cells maintained high GCN5 levels due to its attenuated proteasomal degradation. GCN5 overexpression upregulated amplified in breast cancer 1 (AIB1) expression, resulting in decreased p53 stability and tamoxifen resistance. Conversely, the sensitivity of GCN5-AIB1-overexpressing MCF7 cells to tamoxifen was restored by forced p53 expression. An in vivo study demonstrated a positive correlation between GCN5 and AIB1 and their contribution to tamoxifen resistance. We concluded that GCN5 promotes AIB1 expression and tamoxifen resistance in breast cancer by reducing p53 levels, suggesting the utility of GCN5 and its downstream effectors as therapeutic targets to either prevent or overcome tamoxifen resistance in breast cancer.


Sujet(s)
Tumeurs du sein/métabolisme , Protéines de liaison à l'ADN/métabolisme , Résistance aux médicaments antinéoplasiques , Tamoxifène/administration et posologie , Régulation positive , Facteurs de transcription CBP-p300/métabolisme , Animaux , Tumeurs du sein/traitement médicamenteux , Lignée cellulaire tumorale , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Cellules MCF-7 , Souris , Transplantation tumorale , Pronostic , Récepteurs des oestrogènes/métabolisme , Tamoxifène/pharmacologie , Protéine p53 suppresseur de tumeur/métabolisme
19.
Proc Natl Acad Sci U S A ; 117(33): 19982-19993, 2020 08 18.
Article de Anglais | MEDLINE | ID: mdl-32753382

RÉSUMÉ

The underlying mechanism of necroptosis in relation to cancer is still unclear. Here, MYC, a potent oncogene, is an antinecroptotic factor that directly suppresses the formation of the RIPK1-RIPK3 complex. Gene set enrichment analyses reveal that the MYC pathway is the most prominently down-regulated signaling pathway during necroptosis. Depletion or deletion of MYC promotes the RIPK1-RIPK3 interaction, thereby stabilizing the RIPK1 and RIPK3 proteins and facilitating necroptosis. Interestingly, MYC binds to RIPK3 in the cytoplasm and inhibits the interaction between RIPK1 and RIPK3 in vitro. Furthermore, MYC-nick, a truncated form that is mainly localized in the cytoplasm, prevented TNF-induced necroptosis. Finally, down-regulation of MYC enhances necroptosis in leukemia cells and suppresses tumor growth in a xenograft model upon treatment with birinapant and emricasan. MYC-mediated suppression of necroptosis is a mechanism of necroptosis resistance in cancer, and approaches targeting MYC to induce necroptosis represent an attractive therapeutic strategy for cancer.


Sujet(s)
Leucémies/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Femelle , Humains , Leucémies/génétique , Leucémies/physiopathologie , Souris , Souris de lignée BALB C , Nécroptose , Liaison aux protéines , Transport des protéines , Protéines proto-oncogènes c-myc/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Transduction du signal
20.
J Cancer ; 11(12): 3416-3423, 2020.
Article de Anglais | MEDLINE | ID: mdl-32284737

RÉSUMÉ

Breast cancer is one of the most commonly diagnosed cancers in women worldwide. Approximately 40% of patients with breast cancer acquire endocrine resistance following therapy with tamoxifen. Many explanations for the development of endocrine resistance have been put forward, one of them being the dysregulation of long non-coding RNAs (lncRNAs). The lncRNA HOTAIRM1, known to be involved in myelopoiesis as well as transcriptional regulation of the HOXA genes in embryonic stem cells, is also expressed in breast cancer cells. This study explored the molecular mechanisms of HOTAIRM1 involved in acquired tamoxifen resistance. We showed that HOTAIRM1 and HOXA1 are concurrently up-regulated in tamoxifen-resistant MCF7 (TAMR) cells. Knockdown of HOTAIRM1 down-regulated HOXA1 expression and restored sensitivity to tamoxifen. In addition, the knockdown of HOXA1 showed similar effects, suggesting that the HOTAIRM1/HOXA1 axis regulates tamoxifen resistance. Furthermore, we showed that HOTAIRM1 directly interacts with EZH2 and prevents the PRC2 complex from binding and depositing H3K27me3 on the putative promoter of HOXA1. Together, our findings suggest that HOXA1 and its neighboring lncRNA, HOTAIRM1, might serve as potential therapeutic targets for ER+ breast cancer patients who have acquired tamoxifen resistance.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE