Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34
Filtrer
1.
J Clin Invest ; 2024 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-39325547

RÉSUMÉ

RNA N6-methyladenosine (m6A) reader YTHDF1 is implicated in cancer etiology and progression. We discovered that radiotherapy (RT) increased YTHDF1 expression in dendritic cells (DCs) of PBMCs from cancer patients, but not in other immune cells tested. Elevated YTHDF1 expression of DCs was associated with poor outcomes in patients receiving RT. We found that loss of Ythdf1 in DCs enhanced the antitumor effects of ionizing radiation (IR) via increasing the cross-priming capacity of DCs across multiple murine cancer models. Mechanistically, IR upregulated YTHDF1 expression in DCs through STING-IFN-I signaling. YTHDF1 in turn triggered STING degradation by increasing lysosomal cathepsins, thereby reducing IFN-I production. We created a YTHDF1 deletion/inhibition prototype DC vaccine, significantly improving the therapeutic effect of RT and radio-immunotherapy in a murine melanoma model. Our findings reveal a new layer of regulation between YTHDF1/m6A and STING in response to IR, which opens new paths for the development of YTHDF1-targeting therapies.

2.
J Transl Med ; 22(1): 769, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143573

RÉSUMÉ

BACKGROUND: Although immune checkpoint inhibitors (ICIs) have revolutionized the landscape of cancer treatment, only a minority of colorectal cancer (CRC) patients respond to them. Enhancing tumor immunogenicity by increasing major histocompatibility complex I (MHC-I) surface expression is a promising strategy to boost the antitumor efficacy of ICIs. METHODS: Dual luciferase reporter assays were performed to find drug candidates that can increase MHC-I expression. The effect of nilotinib on MHC-I expression was verified by dual luciferase reporter assays, qRT-PCR, flow cytometry and western blotting. The biological functions of nilotinib were evaluated through a series of in vitro and in vivo experiments. Using RNA-seq analysis, immunofluorescence assays, western blotting, flow cytometry, rescue experiments and microarray chip assays, the underlying molecular mechanisms were investigated. RESULTS: Nilotinib induces MHC-I expression in CRC cells, enhances CD8+ T-cell cytotoxicity and subsequently enhances the antitumor effects of anti-PDL1 in both microsatellite instability and microsatellite stable models. Mechanistically, nilotinib promotes MHC-I mRNA expression via the cGAS-STING-NF-κB pathway and reduces MHC-I degradation by suppressing PCSK9 expression in CRC cells. PCSK9 may serve as a potential therapeutic target for CRC, with nilotinib potentially targeting PCSK9 to exert anti-CRC effects. CONCLUSION: This study reveals a previously unknown role of nilotinib in antitumor immunity by inducing MHC-I expression in CRC cells. Our findings suggest that combining nilotinib with anti-PDL1 therapy may be an effective strategy for the treatment of CRC.


Sujet(s)
Tumeurs colorectales , Pyrimidines , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/immunologie , Tumeurs colorectales/métabolisme , Humains , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Animaux , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antigène CD274/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/génétique , Souris , Instabilité des microsatellites/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques
3.
J Clin Invest ; 133(24)2023 12 15.
Article de Anglais | MEDLINE | ID: mdl-38099498

RÉSUMÉ

Activation of TGF-ß signaling serves as an extrinsic resistance mechanism that limits the potential for radiotherapy. Bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) antagonizes TGF-ß signaling and is implicated in cancer progression. However, the molecular mechanisms of BAMBI regulation in immune cells and its impact on antitumor immunity after radiation have not been established. Here, we show that ionizing radiation (IR) specifically reduces BAMBI expression in immunosuppressive myeloid-derived suppressor cells (MDSCs) in both murine models and humans. Mechanistically, YTH N6-methyladenosine RNA-binding protein F2 (YTHDF2) directly binds and degrades Bambi transcripts in an N6-methyladenosine-dependent (m6A-dependent) manner, and this relies on NF-κB signaling. BAMBI suppresses the tumor-infiltrating capacity and suppression function of MDSCs via inhibiting TGF-ß signaling. Adeno-associated viral delivery of Bambi (AAV-Bambi) to the tumor microenvironment boosts the antitumor effects of radiotherapy and radioimmunotherapy combinations. Intriguingly, combination of AAV-Bambi and IR not only improves local tumor control, but also suppresses distant metastasis, further supporting its clinical translation potential. Our findings uncover a surprising role of BAMBI in myeloid cells, unveiling a potential therapeutic strategy for overcoming extrinsic radioresistance.


Sujet(s)
Tumeurs , Facteur de croissance transformant bêta , Animaux , Humains , Souris , Protéines membranaires/métabolisme , Tumeurs/génétique , Tumeurs/radiothérapie , Protéines de liaison à l'ARN/génétique , Facteurs de transcription/métabolisme , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/métabolisme , Microenvironnement tumoral
4.
Biol Pharm Bull ; 46(11): 1569-1575, 2023.
Article de Anglais | MEDLINE | ID: mdl-37914359

RÉSUMÉ

Ovarian cancer (OC) is one of the most common and high mortality type of cancer among women worldwide. The majority of patients with OC respond to chemotherapy initially; however, most of them become resistant to chemotherapy and results in a high level of treatment failure in OC. Therefore, novel agents for the treatment of OC are urgently required. Benzimidazole anthelmintics might have the promising efficacy for cancer therapy as their selectively binding activity to ß-tubulin. Recent study has shown that one of the benzimidazole anthelmintics oxfendazole inhibited cell growth of non-small cell lung cancer cells, revealing its anti-cancer activity; however, the pharmacological action and detailed mechanism underlying the effects of oxfendazole on OC cells remain unclear. Therefore, the present study investigated the cytotoxic effects of oxfendazole on OC cells. Our results demonstrated that oxfendazole significantly decreased the viability of OC cells. Oxfendazole inhibited the proliferation, induced G2/M phase arrest and apoptotic cell death in A2780 cells. The c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK) pathway was activated and reactive oxygen species (ROS) generation was increased in OC cells treated with oxfendazole; oxfendazole-induced apoptosis was notably abrogated when co-treated with JNK inhibitor SP600125 and ROS scavenger N-acetyl-L-cysteine (NAC), indicating that JNK/MAPK pathway activation and ROS accumulation was associated with the oxfendazole-induced apoptosis of OC cells. Moreover, oxfendazole could also induce the proliferation inhibition and apoptosis of cisplatin resistant cells. Collectively, these results revealed that oxfendazole may serve as a potential therapeutic agent for the treatment of OC.


Sujet(s)
Anthelminthiques , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Tumeurs de l'ovaire , Humains , Femelle , JNK Mitogen-Activated Protein Kinases/métabolisme , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Tumeurs de l'ovaire/traitement médicamenteux , Apoptose , Benzimidazoles/pharmacologie , Système de signalisation des MAP kinases , Anthelminthiques/pharmacologie
5.
Br J Biomed Sci ; 80: 10794, 2023.
Article de Anglais | MEDLINE | ID: mdl-37025162

RÉSUMÉ

Purpose: Additional effective therapeutic strategies for Type 2 diabetes (T2D) patients are urgently needed. Gut microbiota plays an important role in T2D development and is a promising treatment strategy for T2D patients. Faecalibacterium prausnitzii (F. prausnitzii) is regarded as one of the most important bacterial indicators for a healthy gut, but the mechanisms of its anti-diabetic properties are still unclear. Methods and Results: The abundance of F. prausnitzii in feces of patients with T2D was detected by using qPCR. The effects of F. prausnitzii on glucose homeostasis, insulin resistance (IR), dyslipidemia, hepatic steatosis and inflammation were investigated in type 2 diabetic (T2D) db/db mice. We also investigated F. prausnitzii in people. Our results showed that the abundance of F. prausnitzii was significantly lower in T2D patients compared to healthy subjects. In T2D mice, we found that F. prausnitzii treatment significantly decreased fasting blood glucose and IR index, indicating improved glucose intolerance as well as IR. Furthermore, based on evaluation of lipid-regulating enzyme activities and proinflammatory cytokine levels, F. prausnitzii was not only able to improve inflammation in both adipose tissue and liver, but also ameliorate hepatic steatosis through inhibiting the activity of hepatic lipogenic enzymes. Conclusion: These results suggested that F. prausnitzii might serve as a therapeutic option for T2D by improved IR, lipid metabolism and inflammation.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Stéatose hépatique , Insulinorésistance , Troubles du métabolisme lipidique , Humains , Souris , Animaux , Faecalibacterium prausnitzii/métabolisme , Métabolisme lipidique , Inflammation
6.
iScience ; 26(3): 106140, 2023 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-36879799

RÉSUMÉ

Antibody-secreting B cells have long been considered the central element of gut homeostasis; however, tumor-associated B cells in human colorectal cancer (CRC) have not been well characterized. Here, we show that the clonotype, phenotype, and immunoglobulin subclasses of tumor-infiltrating B cells have changed compared to adjacent normal tissue B cells. Remarkably, the tumor-associated B cell immunoglobulin signature alteration can also be detected in the plasma of patients with CRC, suggesting that a distinct B cell response was also evoked in CRC. We compared the altered plasma immunoglobulin signature with the existing method of CRC diagnosis. Our diagnostic model exhibits improved sensitivity compared to the traditional biomarkers, CEA and CA19-9. These findings disclose the altered B cell immunoglobulin signature in human CRC and highlight the potential of using the plasma immunoglobulin signature as a non-invasive method for the assessment of CRC.

7.
Adv Sci (Weinh) ; 10(14): e2205620, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36922750

RÉSUMÉ

The nuclear N6 -methyladenosine (m6 A) reader YT521-B homology-domain-containing protein 1 (YTHDC1) is required to maintain embryonic stem cell identity. However, little is known about its biological functions in intestinal-resident macrophages and inflammatory bowel disease (IBD). Herein, it is demonstrated that macrophage-specific depletion or insufficiency of YTHDC1 accelerates IBD development in animal models. On the molecular basis, YTHDC1 reduction in IBD-derived macrophages is attributed to Zinc finger protein 36 (ZFP36)-induced mRNA degradation. Importantly, transcriptome profiling and mechanistic assays unveil that YTHDC1 in macrophages regulates Ras homolog family member H (RHOH) to suppress inflammatory responses and fine-tunes NME nucleoside diphosphate kinase 1 (NME1) to enhance the integrity of colonic epithelial barrier, respectively. Collectively, this study identifies YTHDC1 as an important factor for the resolution of inflammatory responses and restoration of colonic epithelial barrier in the setting of IBD.


Sujet(s)
Maladies inflammatoires intestinales , Animaux , Macrophages/métabolisme , Côlon
8.
J Cancer Res Clin Oncol ; 149(8): 5095-5108, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-36348020

RÉSUMÉ

PURPOSE: Methyltransferase-like 3 (METTL3), a key member of the m6A methyltransferase complex, is upregulated in multiple human malignancies and plays a role in regulating tumor migration. This study aimed to reveal the underlying mechanism by which METTL3 in regulates the metastasis of colorectal cancer (CRC). METHODS: We compared METTL3 expression levels in CRC tumor tissues and adjacent nontumor tissues by immunohistochemistry (IHC). The functional roles of METTL3 in CRC were assessed by real-time cell migration assays, wound-healing assays and Transwell assays. miRNA sequencing (miRNA-seq), RNA-binding protein immunoprecipitation (RIP) assays and N6-methyladenosine immunoprecipitation (MeRIP) assays were performed to confirm the molecular mechanism underlying the involvement of METTL3 in CRC cell metastasis. RESULTS: We found that METTL3 was overexpressed in CRC tissues. METTL3 knockdown significantly inhibited CRC cell migration and invasion, while METTL3 overexpression had the opposite effects. Furthermore, we demonstrated that METTL3 regulates miR-196b expression via an N6-methyladenosine (m6A)-pri-miR-196b-dependent mechanism and thereby promotes CRC metastasis. CONCLUSION: This study shows the important role of METTL3 in CRC metastasis and provides novel insight into m6A modification in CRC metastasis.


Sujet(s)
Tumeurs colorectales , microARN , Humains , microARN/génétique , Adénosine , Mouvement cellulaire/génétique , Methyltransferases/génétique , Tumeurs colorectales/génétique
9.
Biol Pharm Bull ; 45(9): 1238-1245, 2022.
Article de Anglais | MEDLINE | ID: mdl-36047191

RÉSUMÉ

Despite advances in colorectal cancer (CRC) treatment, most advanced CRC patients who experience disease progression after chemotherapy, targeted therapy, and immunotherapy face a situation in which there is no available medicine. Thus, new therapeutic drugs for CRC are urgently needed. Studies have shown that cholesteryl ester transfer protein (CETP) has a vital role in tumor development and is a possible target for CRC therapy. We found that Evacetrapib, a CETP inhibitor, suppressed CRC cell growth by inhibiting the Wnt/ß-catenin signaling pathway and activating the c-Jun NH2-terminal kinase (JNK) signaling pathway in CRC. Therefore, Evacetrapib displays an anti-cancer effect and is a possible option for treating CRC.


Sujet(s)
Tumeurs colorectales , Voie de signalisation Wnt , Benzodiazépines , Lignée cellulaire tumorale , Prolifération cellulaire , Tumeurs colorectales/anatomopathologie , Régulation de l'expression des gènes tumoraux , Humains , Système de signalisation des MAP kinases , bêta-Caténine/métabolisme
10.
Mol Biol Rep ; 49(6): 4281-4292, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35262820

RÉSUMÉ

BACKGROUND: The emergence of nonresponse or resistance to traditional chemotherapeutic agents is one of the main challenges of colorectal cancer (CRC) therapies. Thus, novel therapeutic drugs that can improve the clinical outcomes of CRC patients are urgently needed. The purpose of this study was to investigate the effects and mechanisms of pyrimethamine in CRC. METHODS AND RESULTS: In this study, we assessed the role of pyrimethamine on CRC cell growth by cell counting kit-8 and colony formation assays. Cell cycle distribution and cellular senescence were determined by flow cytometry and senescence-associated ß-galactosidase staining respectively. RNA-seq analysis and western blotting were used to investigate the potential pathways of pyrimethamine in CRC development. Moreover, animal experiments were performed to evaluate the effect of pyrimethamine in vivo. Our results demonstrated that pyrimethamine could inhibit cell growth by inducing S phase arrest followed by cellular senescence in CRC cells, and the p38MAPK-p53 axis was probably involved in that effect. In addition, pyrimethamine could also boost CD8+ T-cell mediated cytotoxicity and exert antitumor activity in vivo. CONCLUSION: These results indicated that pyrimethamine may be a promising candidate agent for CRC treatment.


Sujet(s)
Tumeurs colorectales , Pyriméthamine , Animaux , Apoptose , Lymphocytes T CD8+ , Cycle cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Vieillissement de la cellule , Tumeurs colorectales/métabolisme , Pyriméthamine/pharmacologie , Pyriméthamine/usage thérapeutique , Lymphocytes T/métabolisme
11.
Cancer Biol Ther ; 23(1): 319-327, 2022 12 31.
Article de Anglais | MEDLINE | ID: mdl-35332847

RÉSUMÉ

Approximately 40% of patients with diffuse large B-cell lymphoma (DLBCL) are refractory or relapse to standard chemotherapy, and most of them are activated B cell-like DLBCLs (ABC-DLBCL) and germinal center B cell-like DLBCLs (GCB-DLBCL). SNS-032, a novel and selective CDK7/9 inhibitor, that the first phase clinical trials approved by US FDA for cancer treatment have been completed. In this study, we investigated the anti-tumor effect of SNS-032 in ABC- and GCB-DLBCL subtypes. We report that SNS-032 induced growth inhibition and cell apoptosis in both DLBCL cells in vitro, and inhibited the growth of both DLBCL xenografts in nude mice. Mechanistically, SNS-032 inhibited RNA polymerase II, which led to transcriptional-dependent suppression of NF-κB signaling pathway and its downstream targets involved in cell survival; SNS-032 also downregulates BCL-2 and c-MYC in both mRNA and protein levels. Significantly, these findings provide pre-clinical evidence for application of targeting the CDK7/9 in DLBCL.


Sujet(s)
Lymphome B diffus à grandes cellules , Récidive tumorale locale , Animaux , Apoptose , Kinases cyclines-dépendantes , Humains , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/anatomopathologie , Souris , Souris nude , Oxazoles , Thiazoles
12.
Cancer Lett ; 526: 322-334, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-34767926

RÉSUMÉ

The relationship between microRNA (miRNA) and hosting long non-coding RNA (lncRNA) remains unclear. Here, the expression levels of microRNA-210 (miR-210) and hosting lncRNA MIR210HG are significantly increased and positively correlated in gastric cancer (GC). Gain- and loss-of-function studies demonstrate that miR-210 and MIR210HG synergistically promote the migration and invasion of GC cells in vitro. Furthermore, GC sublines simultaneously expressing miR-210 and MIR210HG display synergistic promotion of lung metastasis in vivo. Mechanistically, MIR210HG interacts with DExH-box helicase 9 (DHX9) to increase DHX9/c-Jun complex's occupancy on the promoter of matrix metallopeptidases (MMPs), and thus promotes migration and invasion of GC cells. Additionally, miR-210 directly suppresses the expression of dopamine receptor D5 (DRD5), serine/threonine kinase 24 (STK24) and MAX network transcriptional repressor (MNT), resulting in enhanced migration and invasion. Finally, MYC proto-oncogene (c-Myc) transactivates miR-210 and MIR210HG. Overexpression of miR-210 or/and MIR210HG can rescue the inhibitory effect on the migration and invasion by silencing c-Myc. Moreover, c-Myc inhibitor significantly decreases lung metastasis of GC in vivo. Collectively, our findings identify a novel mechanism, by which c-Myc-activated miR-210 and MIR210HG synergistically promote the metastasis of GC.


Sujet(s)
microARN/génétique , Protéines proto-oncogènes c-myc/génétique , ARN long non codant/génétique , Tumeurs de l'estomac/génétique , Animaux , Lignée cellulaire tumorale , Femelle , Gènes myc , Hétérogreffes , Humains , Introns , Souris , Souris de lignée NOD , Souris SCID , microARN/métabolisme , Métastase tumorale , Protéines proto-oncogènes c-myc/métabolisme , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie
13.
Front Pharmacol ; 12: 729367, 2021.
Article de Anglais | MEDLINE | ID: mdl-34776955

RÉSUMÉ

There is an urgent need for novel agents for colorectal cancer (CRC) due to the increasing number of cases and drug-resistance related to current treatments. In this study, we aim to uncover the potential of chaetocin, a natural product, as a chemotherapeutic for CRC treatment. We showed that, regardless of 5-FU-resistance, chaetocin induced proliferation inhibition by causing G2/M phase arrest and caspase-dependent apoptosis in CRC cells. Mechanically, our results indicated that chaetocin could induce reactive oxygen species (ROS) accumulation and activate c-Jun N-terminal kinase (JNK)/c-Jun pathway in CRC cells. This was confirmed by which the JNK inhibitor SP600125 partially rescued CRC cells from chaetocin induced apoptosis and the ROS scavenger N-acetyl-L-cysteine (NAC) reversed both the chaetocin induced apoptosis and the JNK/c-Jun pathway activation. Additionally, this study indicated that chaetocin could down-regulate the expression of CD47 at both mRNA and protein levels, and enhance macrophages phagocytosis of CRC cells. Chaetocin also inhibited tumor growth in CRC xenograft models. In all, our study reveals that chaetocin induces CRC cell apoptosis, irrelevant to 5-FU sensitivity, by causing ROS accumulation and activating JNK/c-Jun, and enhances macrophages phagocytosis, which suggests chaetocin as a candidate for CRC chemotherapy.

14.
Chem Res Toxicol ; 34(1): 47-53, 2021 01 18.
Article de Anglais | MEDLINE | ID: mdl-33397103

RÉSUMÉ

Particulate matter (PM2.5) exposure is reported to have deleterious effects on health. Maternal PM2.5 exposure has been confirmed to damage the growth of somatic cells and enhance the incidence of chronic respiratory diseases in children. Here we aim to investigate the impact of in utero PM2.5 exposure on early birth weight and postnatal lung development. Pregnant Sprague-Dawley rats were administered PM2.5 (0.1, 0.5, 2.5, or 7.5 mg/kg) intraperitoneally every 3 days until birth. Maternal and birth outcomes and somatic growth were monitored. Lungs were collected on PND1 (where PND = postnatal day) and PND28; the lung wet-to-dry weight ratio (W/D) was analyzed, and reactive oxygen species (ROS) levels were measured. Expression of Toll-like receptor 4 (TLR4) and NF-κB were evaluated by Western blotting and quantitative RT-PCR. There were no significant intergroup differences for maternal outcomes; however, offspring exposed in utero to 2.5 and 7.5 mg/kg PM2.5 were significantly smaller in litter weight than the controls. In utero exposure to 2.5 and 7.5 mg/kg PM2.5 led to lower body weight after birth and disrupted lung development during infancy. ROS levels were significantly increased in the 7.5 mg/kg PM2.5 group. PM2.5-treated rats showed upregulated pulmonary expression of TLR4 and NF-κB. Maternal PM2.5 exposure enhances the risk of low birth weight and affects lung alveolar development. The underlying molecular mechanisms may involve TLR4/NF-κB signaling.


Sujet(s)
Polluants atmosphériques/pharmacologie , Poumon/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/génétique , Récepteur de type Toll-4/génétique , Polluants atmosphériques/composition chimique , Animaux , Animaux nouveau-nés , Poids de naissance/effets des médicaments et des substances chimiques , Femelle , Injections péritoneales , Poumon/métabolisme , Mâle , Facteur de transcription NF-kappa B/métabolisme , Taille de particule , Grossesse , Rats , Rat Sprague-Dawley , Récepteur de type Toll-4/métabolisme
15.
Onco Targets Ther ; 13: 10323-10333, 2020.
Article de Anglais | MEDLINE | ID: mdl-33116612

RÉSUMÉ

BACKGROUND: Ovarian cancer (OC) is one of the most common causes of cancer-related death among women; accordingly, new biomarkers of OC are urgently needed. Potassium voltage-gated channel sub-family H member 3 (KCNH3) is a voltage-gated potassium channel member involved in cognitive function and diabetes. Here, we aimed to elucidate the role and potential molecular mechanisms of KCNH3 in OC. MATERIALS AND METHODS: KCNH3 expression levels in OC tissues were analyzed using TCGA data and confirmed by RT-qPCR and immunohistochemistry in OC tissues. The cell counting kit-8 was used to assess cell proliferation in OC cells in which KCNH3 was knocked-down with small interference RNA (siRNA). Wound-healing and transwell invasion assays were used to assess migratory and invasive abilities, respectively. Cell cycle distribution and apoptosis were determined using a flow cytometer. Gene set enrichment analysis and Western blot were used to investigate the potential pathways of KCNH3 in OC development. RESULTS: TCGA data and RT-qPCR results from patients with OC revealed high KCNH3 expression in OC tissues compared to normal ovarian tissues. Survival analysis in patients with OC suggested that high KCNH3 expression might be an independent predictor for poor overall survival and disease-free survival. In vitro studies showed that KCNH3 silencing in OC cells could inhibit cell proliferation and migration ability, and induce apoptosis and G2/M phase arrest. Furthermore, Western blot results showed that KCNH3 silencing might induce downregulation of RPA1 and RPA2 expression level in both SKOV3 and COC1 cells. CONCLUSION: KCNH3 plays an important role in cancer progression in patients with OC. Further investigation might reveal KCNH3 as a potential biomarker for prognosis or diagnosis in OC.

16.
Cell Death Dis ; 11(9): 788, 2020 09 23.
Article de Anglais | MEDLINE | ID: mdl-32968054

RÉSUMÉ

Circular RNAs (circRNAs) are a newly discovered type of biological molecule that belongs to the noncoding RNA family. Abundant evidence has shown that circRNAs are involved in the progression of various cancers. However, the particular functions of circRNAs in colorectal cancer (CRC) remain elusive. In this study, we investigated the differentially expressed circRNAs in three pairs of cancer tissue and adjacent normal tissue of CRC. We revealed that circGLIS2 expression was higher in CRC tissue and cell lines. Gain-and-loss function assays showed that circGLIS2 was involved in the regulation of cell migration. Moreover, overexpressing circGLIS2 in CRC cells activated the NF-κB pathway and induced pro-inflammatory chemokine production, which evoked tumor-associated inflammation through recruiting leukocytes. In turn, when the cancer cells were exposed to the supernatant of circGLIS2 overexpressed cancer cells, they were endowed with the ability of migration and chemokines production. Furthermore, the rescue assay confirmed that circGLIS2 activated NF-κB signaling and promoted cell migration by sponging miR-671. Overall, our study reveals that circGLIS2, acting as a potential oncogene, maintains the abnormal activation state of the NF-κB signaling pathway via the miR-671 sponge mechanism in CRC cells. This study provides a scientific basis for targeting circGLIS2 in colorectal cancer interventions.


Sujet(s)
Tumeurs du côlon/génétique , Tumeurs colorectales/génétique , Facteurs de transcription Krüppel-like/sang , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Tumeurs du côlon/sang , Tumeurs colorectales/sang , Évolution de la maladie , Régulation de l'expression des gènes tumoraux/génétique , Humains , Facteurs de transcription Krüppel-like/génétique , ARN circulaire
18.
Oncol Lett ; 20(1): 373-381, 2020 Jul.
Article de Anglais | MEDLINE | ID: mdl-32565963

RÉSUMÉ

Ovarian cancer (OC) is one of the most fatal gynecological malignancies in the world and confers a poor 5-year survival rate. The present study was designed to discover novel prognostic markers for patients with OC in order to estimate disease metastasis or recurrence. Based on the large cohorts of transcriptome data from multicenter sources, a comprehensive analysis was performed to explore potential prognostic markers. A total of 269 differentially expressed genes were identified, of which 32 were upregulated and 237 downregulated in OC tissues compared with the corresponding expression in normal tissues. Kaplan-Meier analysis, log-rank test and nomogram analysis were employed to demonstrate that low expression levels of claudin 10 (CLDN10) were associated with a less favorable disease prognosis. The most promising prognostic marker for OC was subsequently selected. Additionally, the prognostic nomogram was constructed in order to assess the 5-year survival rate using CLDN10 expression as a prognostic marker for OC. Furthermore, gene set enrichment analysis and analysis of the tumor-associated competing endogenous RNA network were performed to elucidate the potential biological processes associated with CLDN10 expression. The current results indicated that CLDN10 may influence OC progression via transforming growth factor-ß (TGF-ß)- or WNT/ß-catenin-induced epithelial-to-mesenchymal transition (EMT). The associations among CLDN10, microRNA-486-5p, TGF-ß, WNT/ß-catenin and EMT should be further investigated in future studies.

19.
Biochem Pharmacol ; 177: 114014, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32387457

RÉSUMÉ

Developing new drugs for killing colorectal cancer (CRC) cells is urgently needed. Here, we explored the antitumor effects of toosendanin (TSN) in CRC, as well as explored its antitumor mechanisms and direct targets. Cell proliferation and apoptosis were analyzed by CCK8, colony formation, real-time cell impedance and flow cytometry. The signaling pathway and Wnt activity were analyzed by Wnt luciferase activity assay, quantitative real-time PCR and western blot. The interaction between TSN and the κ-opioid receptor was analyzed by a molecular docking simulation. BALB/c nude mice were used to detect the effects of TSN on tumor growth in vivo. We found that TSN inhibited proliferation, induced G1 phase arrest and caused caspase-dependent apoptosis in both 5-FU-sensitive and 5-FU-resistant CRC cells. Moreover, TSN effectively inhibited CRC growth in vivo. In terms of the mechanism, TSN inhibited Wnt/ß-catenin signaling in CRC cells, and the molecular docking results showed that TSN could bind to κ-opioid receptors directly. Additionally, TSN-induced apoptosis and ß-catenin decline were both reversed by the selective κ-opioid receptor agonist U50,488H. Our data demonstrate that TSN-induced apoptosis in CRC cells is associated with the κ-opioid receptor/ß-catenin signaling axis, and TSN has promising potential as an antitumor agent for CRC treatment.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Tumeurs colorectales/traitement médicamenteux , Médicaments issus de plantes chinoises/pharmacologie , Récepteur kappa/métabolisme , bêta-Caténine/métabolisme , 2-(3,4-Dichlorophényl)-N-méthyl-N-((1S,2S)-2-(pyrrolidin-1-yl)cyclohexyl)acétamide/pharmacologie , Animaux , Antinéoplasiques d'origine végétale/pharmacologie , Apoptose/physiologie , Lignée cellulaire tumorale , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/composition chimique , Femelle , Fluorouracil/pharmacologie , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Humains , Souris de lignée BALB C , Simulation de docking moléculaire , Récepteur kappa/agonistes , Récepteur kappa/composition chimique , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , bêta-Caténine/génétique
20.
Eur J Pharmacol ; 876: 173064, 2020 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-32179085

RÉSUMÉ

The selective BCR-ABL tyrosine kinase inhibitor imatinib is one of the first-line therapies in the management of chronic myeloid leukaemia (CML). However, acquired resistance to this inhibitor, which is especially conferred by the T315I point mutation in BCR-ABL, impedes the efficacy of imatinib therapy. Therefore, the discovery and development of novel agents to overcome imatinib resistance is urgently needed. Pseudolaric acid B (PAB), a small molecule isolated from the traditional Chinese medicine Cortex pseudolaricis, has been reported to be a potential candidate for immune disorders and cancer treatment. However, its effects on CML and the involved molecular mechanism have not been reported. In the current study, by performing both in vitro and in vivo experiments in CML cells, we showed that PAB blocked the cell cycle at G2/M phase and subsequently activated the caspase pathway, cleaved the BCR-ABL protein and inhibited the BCR-ABL downstream pathways, ultimately leading to cell proliferation inhibition, cytotoxicity and apoptosis. These events were observed in both imatinib-sensitive and imatinib-insensitive CML cell lines. Moreover, PAB decreased the viability of primary blood mononuclear cells from CML patients and induced apoptosis in these cells. Our findings suggest that PAB could be used as a novel agent to sensitize imatinib-resistant CML.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Diterpènes/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Mésilate d'imatinib/pharmacologie , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Mitose/effets des médicaments et des substances chimiques , Animaux , Cycle cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises , Femelle , Protéines de fusion bcr-abl/génétique , Humains , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Souris nude , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE